Open Access

Metformin prevents the onset and progression of intervertebral disc degeneration: New insights and potential mechanisms (Review)

  • Authors:
    • Wenzhi Yang
    • Yipin Yang
    • Yong Wang
    • Zongshi Gao
    • Jingtang Zhang
    • Weimin Gao
    • Yanjun Chen
    • You Lu
    • Haoyu Wang
    • Lingyan Zhou
    • Yifan Wang
    • Jie Li
    • Hui Tao
  • View Affiliations

  • Published online on: July 2, 2024     https://doi.org/10.3892/ijmm.2024.5395
  • Article Number: 71
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metformin has been the go‑to medical treatment for addressing type 2 diabetes mellitus (T2DM) as a frontline oral antidiabetic. Obesity, cancer and bone deterioration are linked to T2DM, which is considered a metabolic illness. Numerous diseases associated with T2DM, such as tumours, cardiovascular disease and bone deterioration, may be treated with metformin. Intervertebral disc degeneration (IVDD) is distinguished by degeneration of the spinal disc, accompanied by the gradual depletion of proteoglycans and water in the nucleus pulposus (NP) of the IVD, resulting in lower back pain. The therapeutic effect of metformin on IVDD has also attracted much attention. By stimulating AMP‑activated kinase, metformin could enhance autophagy and suppress cell senescence, apoptosis and inflammation, thus effectively delaying IVDD. The present review aimed to systematically explain the development of IVDD and mechanism of metformin in the treatment and prevention of IVDD to provide a reference for the clinical application of metformin as adjuvant therapy in the treatment of IVDD.

1. Introduction

Intervertebral disc degeneration (IVDD) is a disease characterized by degeneration of the spinal disc, accompanied by the gradual loss of proteoglycans and water in the nucleus pulposus (NP) of the IVD (1). IVDD can lead to changes in the structure of the spine, which can adversely affect the normal physiological function of the spine (2). In addition, IVDD is the prerequisite and pathological basis for degenerative disease of the lumbar spine, including lumbar disc herniation and stenosis (3). Epidemiological studies have shown that IVDD is the primary factor of lower back pain (LBP), which is a common clinical symptom (4). In 2019, the global adult incidence of LBP was 7.34% (5,6). As the leading factor in the global disability burden, LBP-related disability cases increased by 47% from 1990 to 2019 (5). The lifetime instances of LBP in Americans amounts to ~80% (7). A notable burden is imposed on society when people struggle with recurrent LBP that causes harm to their physical and mental health (8,9). However, the exact pathogenesis of IVDD is not clear and there is a lack of effective treatment options for IVDD and secondary LBP.

Oral metformin is a prescription medication most frequently used for treating diabetes, including type 2 diabetes mellitus (T2DM). Patients with T2DM who use metformin have excellent safety profiles, along with satisfactory blood glucose control. This drug rarely causes adverse reactions such as hypoglycaemia (10-12). The primary cause of fasting hyperglycaemia in patients with T2DM is increased liver glucose production. Various studies have noted that metformin controls hyperglycaemia by inhibiting gluconeogenesis in the liver (13-16). Metformin is efficient at alleviating a wide range of conditions, such as obesity (17), eating disorders (18), tumours (19), cardiovascular disease (20,21), polycystic ovary syndrome (22), kidney disease (23), cell senescence (24), osteoporosis (OP) (25) and osteoarthritis (OA) (26). DM is significantly associated with IVDD (27). DM can promote IVDD by abnormal blood glucose regulation and damaging IVD metabolism, which makes IVDD a complication of diabetes (28). Due to the association between IVDD and DM, metformin has been demonstrated to alleviate IVDD (29-34). Recent research has revealed that metformin may boost its curative effect on IVDD by encouraging the release of mesenchymal stem cell (MSC)-derived extracellular nanovesicles (30).

The hypoglycaemic effect of metformin is primarily mediated by inhibiting the mitochondrial respiratory chain in the liver and decreasing the production of ATP, which leads to the activation of AMP-activated kinase (AMPK) and increases the ratio of AMP/ATP, thereby inhibiting liver glycogenolysis and glucose production (35,36). Improvement of IVDD is associated with the AMPK signalling pathway (29,37). Autophagy is an internal recycling mechanism that has been preserved throughout evolution to degrade damaged or old cytoplasm in order to preserve cells in a state of equilibrium (38). Impaired autophagy in IVD cells is a primary contributor to IVDD. Appropriate induction of autophagy can slow progression of IVDD, but excessive autophagy can lead to IVD cell death (3,39). Hypoxia can decrease excessive autophagy by limiting reactive oxygen species (ROS) production and inactivating the AMPK/mTOR signalling pathway, thereby contributing to the survival of IVD cells (37). Metformin induces autophagy in NP cells of IVD in an AMPK-dependent manner, which suggests the importance of the AMPK pathway for the treatment of IVDD with metformin (29). The present review summarizes research advances on metformin and IVDD and potential mechanisms of metformin in controlling the onset and progression of IVDD. These results may provide fresh perspectives on how metformin is clinically used to prevent and treat IVDD and facilitate further research into its mechanism of action.

2. IVDD

Composition of IVD

The IVD is a key structure in the human spinal joint, connecting the upper and lower cones to perform movements and carry the load of the spine. The IVD is made up of three components: NP, anulus fibrosus (AF) and the cartilage endplate (CEP) and cone endplate on the upper and lower surfaces (40) (Fig. 1). The NP is composed of chondrocyte-like cells and has a large number of randomly distributed collagen fibres and radially arranged elastic fibers, the most important component of which is aggregated proteoglycan (aggrecan) (41,42), which provides the osmotic properties needed to resist compression and maintain the height of the IVD (43). The AF is composed primarily of water, collagen, aggregated and non-aggregated proteoglycan and non-collagenous protein (44). The AF is structurally composed of 15-25 concentric rings or lamellae composed of type I collagen fibres that are reversed in their alignment between consecutive cones (45). The AF is used to accommodate the NP and maintain NP pressurization under compressive loading (43,46). The CEP mostly is composed of type II collagen, glycosaminoglycan (GAG), and water (47) in similar proportions to those found in articular cartilage (48). The CEP exists in a highly hydrated proteoglycan matrix reinforced by collagen fibres, which fibers may contribute to the connection between the CEP and AF (49), while elastic fibers are key for the connection between the NP and adjacent CEP (50). CEP can separate the IVD from the adjacent cone while accommodating the NP (51). CEP can act as a semipermeable membrane that facilitates material exchange between cone layers (52), which provides nutrients to the cone and eliminates metabolites. CEP can act as a cushion and evenly distribute the load (53).

Mechanisms of IVDD

Aberrant cell-mediated response to increasing collapse of structure is a prevalent route of IVDD (Fig. 1). Degenerative IVD is characterized by structural failure accompanied by accelerated cellular senescence (45). Although mechanical overload is hypothesized to be the direct cause of IVDD, it is only an aggravating factor. The premature weakening of IVD is one of the key factors leading to disc-related issues. This degeneration can be caused by various factors, including genetic predisposition, age, insufficient transport of metabolites, and loading history (45).

A key stage in the development of the IVDD is the degradation of the extracellular matrix (ECM) by NP and AF cells. In NP cells, the matrix exhibits a decrease in GAG, aggrecan and elastin, an alteration in collagen and an increase in fragmented aggrecan, fibronectin and advanced glycosylation end products (AGES) (54). The decrease in type II collagen expression in NP cells is accompanied by an increase in type I and III collagen, ensuring that the absolute amount of collagen remains constant. However, there is an association between changes in type X collagen and calcification of the CEP. High expression of type X collagen may stimulate preosteoblasts to undergo osteogenesis in chondrocytes, leading to calcification of CEP (55,56). In addition, fragments of aggrecan are filtered out of the IVD (57), and the decrease in aggrecan is hypothesized to be closely related to a decrease in the hydration capacity and ability to withstand load of NP cells (58). Additionally, the formation of AGES can increase stiffness and fragility of collagen fibres (54). This may lead to a decrease in the elasticity of the IVD, making it more susceptible to injury and degeneration, which in turn affects the stability and function of the spine (54). In addition, syndecan-4, a transmembrane acetyl heparan sulfate proteoglycan, may mediate degradation of ECM and aggregated proteins via thrombospondin motif 5 and MMPs (59,60). The small leucine-rich proteoglycans are also impaired by degradation (61). This may lead to a loss of elasticity and stability of the ECM, affecting cell-matrix interactions and, consequently, the physiological function and structural stability of the tissue (62). Decrease expression of islet amyloid polypeptide, calcitonin receptor and receptor activity-modifying proteins in AF cells disrupts ECM homeostasis, thereby promoting IVDD (61). Furthermore, the reduction of type I and type II collagen and lectins in AF cells leads to a transition of their load-bearing form to an inner annular elastic cushioning. Prolonged transition may result in compressed pain and AF damage (51). Alterations in the activity of certain enzymes, such as histoproteinases, matrix proteases and polymerase, may indirectly trigger degeneration of the disc (55). For example, MMPs may cause apoptosis in NP cells and calcification of the CEP (63,64).

Numerous signalling pathways are thought to be closely associated with IVDD, such as the NF-κB and MAPK pathways, which advance the progression of IVDD by regulating proinflammatory mediators (65) such as TNF-α, IL-1β and IL-6. In addition, initiation and development of disc disease may be dependent on the Wnt and the Notch pathways. Activation of the Wnt pathway accelerates NP cell senescence (66), while the hypoxia-activated Notch pathway can influence the course of IVDD via proinflammatory factors (66). DNA damage also plays a role in IVDD, leading to the loss of proteoglycans, decreased disc height and increased cellular senescence (67,68). DNA damage and telomere shortening induce replicative senescence in IVD cells primarily through activation of the p53/p21-retinoblastoma (Rb)/p16-Rb signalling pathway (69,70). In addition, oxidative stress serves an important role in IVDD and ROS damage the structure and function of IVD by damaging lipids, DNA and protein (71). Hydrogen peroxide promotes senescence by activating the senescence pathway, which stops NP cells at the G0/G1 phase of the cell cycle (72). The effects of oxidative stress also indicate activation of the p53/p21-Rb/p16-Rb signalling pathway (73). Inflammation promotes senescence in IVD cells and the inflammatory factors TNF-α and IL-1α, -1β, -6 and -17 enhance catabolism of the ECM (73).

Sirtuin (SIRT)1 can protect against IVD by inhibiting the p53-dependent and p16-dependent cellular senescence pathways and it can activate the Akt signalling pathway to protect NP cells from apoptosis (73,74). Moderate autophagy can mitigate apoptosis and delay IVDD in response to oxidative stress (75). Most autophagy is activated via AMPK/mTOR-dependent pathways (76). Overactivated autophagy may adversely affect IVD and hypoxic conditions can inactivate the AMPK/mTOR signalling pathway by limiting ROS production and decreasing activation of autophagy in NP cells through a pathway involving hypoxia-inducible factor 1α (75). In addition, autophagy safeguards CEP cells against calcification brought on by the intermittent, cyclic mechanical strain (77).

Apoptosis is associated with IVDD in NP cells, which are associated with activation of caspase-3 activity, oxidative stress and SIRT1 (37). Extrinsically, TNF-α regulates Fas/Fas ligand levels and thus activates caspase-3/9 activity (66), and may cause a significant increase in a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) family of disintegrins and ADAMTS-7 in degenerating NP cells (78). In addition, in degenerating IVD, microRNAs (miRs) are differentially expressed, including downregulation of miR-155, production of miR-10b and miR-27a, and elevation of miR-21, and downregulation of miR-155 also activates caspase-3 activity to promote apoptosis (37). Additionally, p38 MAPK, JNK1/2 and ERK1/2 in the MAPK family may damage the IVD (78). It has been shown that the MAPK family exerts proapoptotic effects on senescence via direct or indirect activation of the p53/p21-Rb/p16-Rb pathway (73). In addition, apoptosis in AF and CEP cells is associated with the presence of the MAPK family and caspase-3, -8 and -9 (37).

3. Metformin

Physical and chemical properties

The structural formula of metformin (N, N-dimethylbiguanide) is C4H11N5 (molecular weight, 129.16 g/mol); it is composed of a single protonated planar molecule in a non-polar methyl group and two imino groups. Metformin hydrochloride (molecular weight, 165.6 g/mol), which is widely used in the clinic, is constructed of dimethylamine hydrochloride and dicyandiamide (79). It is a white crystalline powder at room temperature. The density is 1.28 g/cm3, the melting point is 223-226°C and the boiling point is 224.1°C (79). This substance is almost insoluble in chloroform and ether. It can be dissolved in water at a ratio of 1:2, and in ethanol at a ratio of 1:100 (80,81). Metformin has low lipophilicity, so it cannot quickly and passively diffuse through the cell membrane (24,82,83). As a hydrophilic base, metformin has an acid-base constant of 2.8 and 11.5. It has numerous tautomeric structures, is monoprotonated at neutral pH (84) and exists as a cation at physiological pH. The concentration of metformin in physiological fluids is represented by the free base (molecular weight, 129.16 g/mol) (83) (Fig. 2).

Pharmacokinetics

Following oral administration, metformin is dispersed in the gastrointestinal tract, liver and kidney (85). The mean oral bioavailability of metformin is 55±16%. Absorption is mainly limited to the proximal small intestine, and it accumulates in the liver after absorption (83). The peak plasma concentration is reached ~3 h after oral administration, and the peak in the liver appears within 1 h of administration (83,86). The plasma concentrations of metformin are 1-4 mg/l in patients who received 1-2 g/day metformin oral treatment (12). At 4 h after drug intake, the liver absorbs 80% of metformin and the body eliminates 98% within 24 h (87). Metformin is primarily excreted through the kidneys. Elimination half-life and apparent total clearance in individuals with normal renal function are 5 h and 1,140±330 ml/min, respectively (83). The absorption, liver utilization and renal excretion of metformin are primarily mediated by organic cation transporters (OCTs). Studies have shown that proton inhibitors decrease metformin transport by inhibiting OCTs (24,83).

To lessen gastrointestinal adverse reactions such as diarrhoea, malignant vomiting and so on, metformin should be taken with meals. Eating can affect drug absorption, but the decreased rate of absorption is not clinically significant for most patients (24,83,88). The faecal recovery of metformin is 20-30% of the oral dose, and metformin is not readily detectable in faeces after intravenous administration, suggesting that faecal metformin is not absorbed or utilized (83).

General mechanisms of metformin in degenerative bone disease

Degenerative skeletal diseases include OP, OA and IVDD, as well as diseases such as ankylosing spondylitis (AS) and periodontitis (24) (Fig. 2). The use of metformin in the treatment of degenerative bone diseases has been verified to be beneficial (24,89). The incidence of OP is lower among patients who have diabetes with carcinoma in situ who are treated with metformin than those not receiving metformin (25). Metformin improves the long-term outcome for people who are significantly overweight and have knee OA (90). To the best of our knowledge however, there have been no clinical studies showing the effectiveness of metformin in the treatment of IVDD. Since the NP is similar to articular cartilage, which consists of water, collagen and proteoglycans, (91-93) and NP and bone tissue have common components and origins (94,95), the mechanisms by which metformin affects them are similar.

Many studies have shown that activation of AMPK has a notable effect on musculoskeletal disorder (96-98). AMPK activity is significantly downregulated in human and mouse chondrocytes with OA and significantly elevated following metformin administration (82). Metformin can lead to osteoblast differentiation in vitro by activating AMPK, promoting bone matrix synthesis and osteoblast proliferation (99). Metformin also stimulates the regenerative process in bone lesions in diabetic and non-diabetic rats (99). Metformin serves an effective role in inhibiting ossification and inflammation in AS fibroblasts by activating the AMPK pathway (100). The aforementioned studies suggest an important role of the AMPK signalling pathway in treatment of degenerative skeletal diseases with metformin. However, the direct targets of metformin that act via the AMPK pathway in these diseases remain unclear. Recent studies have shown that metformin binds presenilin Enhancer 2 (PEN2) and initiates a signalling pathway that intersects with the AMPK/activated lysosomal glucose sensing pathway via ATP6AP1 (101-103). Whether metformin acts through this signalling pathway in skeletal degenerative disease remains unclear.

Phosphoinositide 3-kinase (PI3K)/Akt is one of the key pathways that regulates cell behaviour (including apoptosis, proliferation and differentiation); previous studies have shown that this pathway is a key regulator of osteoblasts and is associated with fracture healing (104-107). Metformin ability to regulate the PI3K/Akt pathway could contribute to its efficacy in the treatment of AS (100). Metformin reduces insulin and insulin-like growth factor-1 (IGF-1) levels, thereby inhibiting phosphorylation of insulin receptor substrate 1/2 (IRS-1/2) and PI3K/Akt/mTOR signalling (24), which is key for the control of skeletal degenerative lesions.

SIRT family is a group of antiaging genes (108) that are closely associated with the aetiology of degenerative bone disease (109). Studies have demonstrated that metformin inhibits IL-1β-induced oxidative and osteoarthritic inflammatory changes by enhancing the SIRT3/PINK1/Parkin signalling pathway. In addition, metformin attenuates IL-1β-induced expression of catabolic genes, such as MMP3 and 13 and enhances the anabolic factor collagen II, which is beneficial for treatment of OA (110-113). In addition, metformin-induced activation of SIRT1 decreases inflammatory mediators and matrix degradation substances, inhibits accumulation of receptor for AGE (RAGE) and protects articular cartilage, thus preventing the development of OA (114). Another study confirmed that metformin inhibits lipopolysaccharide (LPS)-mediated enhancement of IL-6, IL-1β and TNF-α production in human gingival fibroblasts (HGFs) by increasing activating transcription factor-3 (ATF3) expression. Decreases in TNF-α and IL-1β protein expression suggest that metformin mitigates LPS-induced inflammatory responses via the AMPK/NF-κB pathway (115). However, whether this mechanism of metformin is involved in IVDD requires further experimental verification.

Side effects

Metformin is an efficient and typically well-tolerated drug with notable hypoglycaemic effects but there are some adverse events (Fig. 2); the most prevalent negative reactions include gastrointestinal discomfort, including abdominal distension and pain, nausea and vomiting. These symptoms are typically mild, but medication may need to be suspended and the dose adjusted (24,88,116,117). In rare cases, metformin is related to lactic acidosis. Studies have shown that cardiovascular disease and liver and kidney damage following treatment are likely to be the main causes of lactic acidosis and there is no causal relationship with metformin (24,118,119). It has been reported that 1.4% of patients treated with metformin have insomnia, which may be associated with changes in blood glucose levels (120). DM with depression often suffer from insomnia (121,122). In addition, long-term metformin treatment may lead to vitamin B12 deficiency and require regular monitoring to decrease the incidence of side effects (123-125). Researchers determined the milk-to-plasma ratio of metformin in lactating patients and showed that metformin was safe during lactation and did not cause side effects (10,24). However, maternal and infant risks and individual differences should be considered and metformin is not recommended for pregnant patients (24).

4. Mechanism of metformin in IVDD

Metformin induces autophagy through the AMPK pathway and inhibits senescence and apoptosis in myeloid cells

As a highly evolutionarily conserved serine/threonine protein kinase, AMPK is commonly expressed in eukaryotic cells (126). The role of AMPK in the development of IVDD is becoming more obvious (Table I). Activation of the AMPK signalling pathway has been observed in human IVDD (127). AMPK phosphorylation is increased in rat IVDD. IVDD is a complex process involving both apoptosis and senescence (29). In degenerating rat NP cells, AMPK activation further dephosphorylates mTOR and inhibition of AMPK-mediated mTOR facilitates apoptosis and senescence while suppressing cell proliferation and cell cycle progression (34,128). Meanwhile, human AF cell apoptosis is influenced by AMPK (129).

Table I

Preclinical study of metformin in the treatment of IVDD.

Table I

Preclinical study of metformin in the treatment of IVDD.

Treatment groupsExperimental modelResultsConclusion(Refs.)
100 and 200 μM metforminTBHP-induced senescence in NP cells/puncture-induced rat IVDD modelAutophagy, as well as its upstream regulator AMPK, is activated by metformin in NP cells in a dose- and time-dependent manner. Metformin promotes expression of anabolic genes such as COL2A1 and Acan expression while inhibiting expression of catabolic genes such as MMP3 and ADAMTS5 in nucleus pulposus cells. In vivo, metformin ameliorates IVDDMetformin activates autophagy via the AMPK pathway to protect NP cells from apoptosis and senescence and improve IVDD in vivo(29)
EVs, EVs + metformin (1 mM, 24 h)Puncture-induced rat IVDD modelMetformin increases EV markers CD63 and Alix proteins, autophagy markers and the EVs transport-related proteins total and phosphorylated SNAP29, increases expression of ITIH4 in EVs and ameliorates senescence of NP cellsMetformin enhances release of EVs from MSCs via autophagy-associated pathways and ameliorates IVD cell senescence both in vitro and in vivo(30)
10, 50 and 100 μM metforminTBHP-induced senescence in NP cells/puncture-induced rat IVDD modelMetformin inhibits TBHP-induced expression of cell senescence markers P16, P21. With increasing concentrations of metformin, ratio of autophagy marker LC3-II/LC3-I increases, level of p62 decreases and autophagic vesicles in cytoplasmincrease. Metformin activates autophagy and decreases level of DNA damage marker γ-H2AXMetformin ameliorates IVDD by inactivating the cGAS/STING pathway via autophagy(31)
Metformin (100 mM Metformin for 4 hMechanical stretch and IL-1β-induced inflammation in rat AF cells in vitroMetformin increases COL1, COX-2 and MMP13 gene expression, decreases PGE2 production and NF-κB nuclear translocation following 4 h IL-1β treatmentAnti-inflammatory effect of metformin in IVD cells may be mediated by the NF-κB pathway; effect of metformin is enhanced by mechanical stretch(33)
Metformin (0-10 mM for 7 days)LPS-induced inflammation in rabbit AFSCs in vitroMetformin inhibits LPS-induced release of HMGB1 from AFSC nuclei, upregulation of certain inflammation-associated genes (IL-1β and -6, COX-2 and TNF-α) and proteins (IL-1β, COX-2 and TNF-α) as well as catabolic genes (MMP3 and 13) and decrease in anabolic gene (collagen I and II) expressionMetformin exerts an anti-inflammatory effect by blocking HMGB1 translocation, catabolic production and senescence in AFSCs(32)
20 μg/ml metformin for 24-72 hIn vitro culture of IVD cells from patients undergoing lumbar microdiscectomy and sequestrectomyExtracellular matrix structure is disrupted by metformin treatment, with decreased reproductive capacity at 72 h, and inhibited the expression of CHAD and COMP and decreased the expression levels of IL-1β, MMP7 and MMP19.Metformin adversely affects IVD tissue cell culture, including inhibition of cell. proliferation and alteration of gene expression associated with extracellular matrix synthesis and degradation(34)

[i] IVDD, intervertebral disc degeneration; TBHP, Tert-butyl hydroperoxide; NP, Nucleus pulposus; COL2A1, Collagen type II alpha 1 chain; ADAMTS, A disintegrin and metalloproteinase with thrombospondin motifs; EV, Extracellular vesicle; MSC, Mesenchymal stem cell; SNAP, Synaptosome associated protein; ITIH, Inter-alpha-trypsin inhibitor heavy chain; cGAS, cyclic GMP-AMP synthase; STING, Stimulator of interferon genes; AFSC, Anulus fibrosus stem cells; PGE, Prostaglandin E; LPS, Lipopolysaccharide; HMGB, High mobility group box; CHAD, Chondroadherin; COMP, Cartilage oligomeric matrix protein.

Several in vivo studies have demonstrated a significant increase in the concentration of oxidative products in aging and degenerating IVD (130-132). Oxidative stress has been shown to induce apoptosis via the mitochondrial pathway and to induce premature cellular senescence (72,133). Chen et al (29) modelled apoptosis and senescence in medullary cells by inducing oxidative stress using tert-butyl hydroperoxide (TBHP) (29). The aforementioned study investigated the effects of metformin on apoptosis and senescence of myeloid cells under oxidative stress and examined expression of anabolic and catabolic genes in myeloid cells following metformin treatment; metformin had a significant protective effect against TBHP-induced apoptosis. Metformin pretreatment inhibits the TBHP-induced increase in cleaved-caspase3 and Cyclin-dependent kinase inhibitor 2A (p16INK4a) protein content in a dose-dependent manner. In addition, TBHP treatment decreases mRNA levels of the key ECM synthesizing gene collagen type II alpha 1 chain (Col2a1), whereas metformin significantly inhibits this downregulation. The effects of metformin are realized through the activation of autophagy by the AMPK pathway. Metformin significantly decreases Bax and caspase expression and increases BCL-2 under oxidative stress conditions via the mitochondrial pathway. Metformin also significantly decreases the senescence marker senescence-associated β-galactosidase (SA-β-gal) activity and expression of the senescence mediators p16INK4a, p21WAF1 and phosphorylated-p53, which results in the inhibition of oxidative stress-induced senescence in NP cells. Following metformin treatment, expression of catabolic enzymes MMP3 and ADAMTS-5 is inhibited and synthesis of type II collagen and aggrecan is increased, maintaining NP cell homeostasis from an ECM perspective (29). However, more investigation is necessary to show how metformin triggers autophagy via the AMPK pathway. Metformin binds to PEN2 and starts a signalling cascade that interacts with the AMPK-activated lysosomal glucose sensing pathway via ATP6AP1 (101). Low doses of metformin activate the PEN2/ATP6AP1/AMPK pathway, while high concentrations of metformin directly block mitochondrial respiratory chain complex I, thereby lowering ATP and increasing AMP to enable AMPK activation (102). To the best of our knowledge, however, there have been no studies that directly show that metformin improves IVDD by activating AMPK by binding to PEN2 (Fig. 3).

Metformin promotes extracellular vesicle (EV) production and inactivates the cGAS/STING signalling pathway via autophagy, thereby improving myeloid cell senescence

EVs are vesicular secretions produced by MSCs that mediate the biological effects of MSCs (134-136). Compared with MSC-based cell treatment, EV therapy provides a number of potential benefits, including decreased manufacturing costs, greater stability and ease of sterilization, storage and infusion therapy (134). EV therapy has been considered a promising alternative strategy for treatment with MSCs. Autophagic activity is associated with EV biogenesis and secretion and the regulation of autophagy may influence EV production (137,138). Metformin, which is an AMPK activator, is associated with autophagy activation (139). In a recent study, Liao et al (30) showed that metformin treatment effectively promotes the secretion of EVs in vitro and that this process is associated with autophagy activation (30). Metformin activates autophagy in MSCs primarily via AMPK signalling and promotes formation of multivesicular bodies (MVBs) or amphipathic bodies, which is followed by membrane fusion and EV release. In addition, metformin treatment induces phosphorylation of synaptosome associated protein (SNAP29), a key SNARE protein that mediates fusion of MVBs with the plasma membrane, thereby facilitating the release of EVs (30). Liao et al (30) also hypothesized that metformin facilitates release of EVs by altering the expression of inter-α-trypsin inhibitor heavy chain 4, which serves a key role in the regulation of cell proliferation and inflammatory responses. The secretome of MSCs (including EVs) exerts antioxidant and anti-inflammatory effects to rescue resident senescent cells in the IVD; EV treatment reduces the rate of senescent NP cells and delays the progression of IVDD (140).

One of the key indicators of the innate immune response is cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) signalling and its primary function is to recognize and respond to cytoplasmic DNA. After cGAS recognizes and binds DNA in the cytoplasm, STING initiates downstream responses (31). Senescent cells secrete a variety of cytokines called the senescence-associated secretory phenotype (SASP). SASP factors affect the intracellular environment and induce local or systemic dysfunction. However, it has been demonstrated that cGAS/STING signalling can stimulate production of inflammatory factors that lead to SASP factor secretion by senescent cells and accelerate ageing (141). Ren et al (31) demonstrated that metformin inhibits the activation of the cGAS/STING pathway by activating autophagy, thereby inhibiting the inflammatory response and NP cell ageing and ultimately delaying IVDD (31). During IVDD, release of inflammatory factors is the primary cause of excessive senescence in myeloid cells (31). Several studies have proven that oxidative stress is present in degenerative discs and ROS production disrupts intracellular homeostasis and leads to inflammation and accelerated senescence in NP cells (142-144). The cGAS/STING signalling axis consists of the second messenger loop cGAS of the interferon gene and circulating GMP-AMP receptor-stimulating factor. The inflammatory response and cellular senescence both rely on this pathway, which is part of the innate immune system. cGAS sequesters damaged DNA fragments in the cytoplasm in senescent myeloid cells, which then recruits downstream STING to activate the NF-κB signalling pathway, promoting the release of inflammatory cytokines (145). In addition, the cGAS/STING signalling pathway is a pathway that is responsive to cytoplasmic DNA. Under normal conditions, intracellular DNA is stable and present in the nucleus and mitochondria (146-148). When DNA is damaged, histone γ-H2AX (DNA damage marker) is phosphorylated and damaged DNA fragments enter the cytoplasm and are recognized by cGAS, thus activating STING and its downstream molecules (149). NF-κB has been identified as a downstream molecule of cGAS-STING. Damaged DNA fragments induce cGAS/STING activation, which phosphorylates P65 and promotes its translocation to the nucleus (150,151). This promotes the production of inflammatory cytokines, which then induce oxidative stress, leading to degradation and destruction of ECM and accelerated cellular senescence (152). The study has also shown that silencing STING gene inhibits senescence and that overexpression of STING promotes senescence in myeloid cells (152), which suggests that cGAS/STING signalling may serve as a novel therapeutic target to inhibit senescence and mitigate the progression of IVDD (Fig. 3).

Metformin inhibits myeloid cell senescence and apoptosis by suppressing the inflammatory response

NF-κB is a key mediator of cellular responses to inflammation; the downstream factor prostaglandin E2 (PGE2) disrupts IVD cell matrix homeostasis, thereby destroying the disc (153,154). There is substantial evidence that inhibiting NF-κB inhibits the inflammatory response and the production of catabolic factors in IVD cells (153,155), thus providing a potential strategy for inhibiting disc degeneration. Unlike conventional non-steroidal anti-inflammatory drugs, metformin does not inhibit COX-2 (PGE2 synthase) through an enzymatic response but rather regulates COX-2 expression via NF-κB (33). Metformin results in a large decrease in nuclear ectopic NF-κB and a significant decrease in PGE2 levels following dual stimulation of mechanical and inflammatory stress (33). The effect of metformin on proinflammatory gene expression is likely due to blockade of NF-κB translocation to the nucleus via the SIRT1/FOXO/Peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1) pathway induced by activated AMPK (33,156).

High mobility group box 1 (HMGB1) is a nuclear protein that binds to DNA and is a cofactor for gene transcription (157). Under normal conditions, HMGB1 is in the nucleus and serves a role in regulating DNA stability and transcription, but it is released into ECM in response to certain stimulatory conditions (such as LPS) (158), which promotes expression of PGE2 and some inflammatory factors (TNF-α, IL-1β and IL-6) (159,160). Metformin exerts antiaging and anti-inflammatory effects by inhibiting HMGB1 translocation and catabolic products of AFSCs. HMGB1 also significantly decreases the levels of SA-β-gal activity, inflammatory factors (TNF-, IL-1 and IL-6), and certain catabolic enzymes (MMP3 and 13) in AFSCs (32) (Fig. 4).

IL-1, which is an inflammatory factor, is key to the development of IVDD (161). AGEs amplify the inflammatory response via the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome/IL-1β inflammatory response axis, which can lead to IVDD (162,163). In addition, IL-1β leads to a significant increase in ADAMTS and MMP expression, which promotes ECM degradation and leads to IVDD (164,165). Additionally, IL-1β can induce senescence and apoptosis in myeloid cells and inhibit their proliferation (161). It has also been shown that IL-1β increases intracellular ROS levels in murine vertebral MSCs, thereby exacerbating oxidative stress in IVD cells (166). Metformin inhibits IL-1β production by the SIRT3/PINK1/Parkin and the AMPK/SIRT1 pathway (110,114) and inhibits LPS-mediated enhancement of IL-1β in HGFs by increasing ATF3 expression (115). In addition to decreasing IL-1β production, metformin also reduces the effects of existing IL-1β by inhibiting activation of the NLRP3 inflammasome (167). Therefore, metformin may inhibit the proinflammatory effect of IL-1β via several pathways, thus exerting a protective effect against IVDD. In addition, resveratrol inhibits IL-1β-mediated apoptosis in the NP by regulating the PI3K/Akt pathway (168); metformin regulates the PI3K/Akt pathway (169), suggesting that metformin may also inhibit IL-1β-mediated apoptosis in myeloid cells by modulating the PI3K/Akt pathway; nonetheless, additional experiments are required to confirm this hypothesis (Fig. 4).

Metformin can act on IVDD by activating the SIRT signalling pathway

SIRT family is composed of NAD+-dependent histone deacetylases, which participate in biological processes, such as cell survival, senescence, proliferation, apoptosis, DNA repair, metabolism and caloric restriction (170). SIRT1-7 are considered attractive therapeutic targets for ageing-associated diseases such as T2DM and inflammatory and neurodegenerative disease (171). SIRT1 inhibits senescence and apoptosis in myeloid cells via autophagy. SIRT1 blocks the expression of MMP1 and 3 at the mRNA and protein levels in human dermal fibroblasts (172) and enhances the expression of cartilage-specific ECM genes, such as type II collagen and aggregated proteoglycan (173). SIRT1 inhibits matrix metalloproteinase (MMP) expression and activity, and SIRT1 also promotes the synthesis of matrix components, delaying IVDD through these pathways (174,175). SIRT1 acts on the disc via the PI3K/Akt pathway (74,176). There has been much evidence that metformin has a strong activating effect on SIRT1 (177,178). To the best of our knowledge, however, few studies have shown that metformin affects IVDD through SIRT1 signalling and further investigations still need to be conducted (179,180).

SIRT3 is a key factor for IVDD. A key mechanism of AGE-induced oxidative stress and secondary human NP apoptosis is impairment of SIRT3 activity and the mitochondrial antioxidant network (181). Nicotinamide mononucleotide (NMN) activates SIRT3 via the AMPK/PGC-1α pathways (181,182). By preventing compression-induced mitochondrial division, SIRT3 expression improves mitochondrial function, decreases the formation of ROS (183) and activates the SIRT3/FOXO3/SOD2 signalling pathway; SIRT3 activates transcription factor FOXO3a, which in turn promotes superoxide dismutase 2 (SOD2) synthesis and ROS catabolism (184), which reduces NP cell stress and the degradation of ECM. The activating effect of metformin on SIRT3 is evident, and metformin significantly increases the expression of Histone H3 Lysine 79 trimethylation (H3K79me3) in the SIRT3 promoter region by stimulating AMPK production, thereby increasing levels of SIRT3 (185). In addition, metformin inhibits the inflammatory response in myeloid cells via the SIRT3/PINK1/Parkin signalling pathway (110). When NAD is used to inhibit the effect of SIRT3, a significant decrease in the effect of metformin also occurs (186). This finding suggests that metformin may act via SIRT3 to improve IVDD.

In senescent NP cells, expression of SIRT6 is decreased, and SIRT6 decreases levels of the replicative senescence marker SA-β-gal, which inhibits IL-1β-induced apoptosis and activates autophagy in NP cells by inhibiting phosphorylation of mTOR; activation of autophagy inhibits stress-induced apoptosis in NP cells and regulates the expression and activity of certain mechanistic catabolic enzymes (187). For example, MMP-3 (Matrix Metalloproteinases) and MMP-13 are enzymes that are frequently regulated during autophagy, and they play important roles in extracellular matrix degradation and interstitial cell remodelling (188). ADAMTS-4 (A Disintegrin and Metalloproteinase with Thrombospondin Motifs) and ADAMTS-5 are typical examples of autophagy-regulated enzymes, and their activity and expression change significantly during articular cartilage and intervertebral disc degeneration (189). Autophagy may affect cell metabolism and survival by regulating the number and function of lysosomes to influence the activity of endogenous mechanolytic enzymes (190). Metformin augments the expression of SIRT6 in vitro (191). It has also been shown that SIRT2 can decrease the expression of P21 and P53 and thus inhibit ageing and oxidative stress in NP cells (192), and there is a relationship between metformin and SIRT2 (193). However, whether metformin ameliorates IVDD directly by acting on SIRT-related signalling pathways requires further study.

5. Clinical studies

To the best of our knowledge, the use of metformin in the treatment of IVDD has not been validated by clinical data. However, in clinically relevant conditions, such as obesity, aging and diabetes, that may be a direct or indirect inducer of IVDD, metformin has been used in clinical treatment or trials (24,194,195). Patients experiencing insulin sensitivity and resistance who are overweight exhibit increased weight loss with metformin (196). To the best of our knowledge, few clinical trials directly examine the effects of metformin on human aging (197,198). A randomized placebo-controlled clinical trial of metformin is underway to prevent frailty in elderly patients with poor glucose tolerance and examine the effects of metformin on several markers of aging (199). However, multiple preclinical experiments have established that metformin can be beneficial in treating IVDD (29-34). By promoting autophagy or controlling release of EVs from bone marrow MSCs, metformin prevents apoptosis and senescence in NP cells (29,30). In addition, a randomized, double-blind, placebo-controlled, crossover trial showed that metformin delayed senescence in the elderly population via metabolic and non-metabolic effects (200), suggesting that metformin plays a role in ageing-related diseases. The Targeting Aging with Metformin (TAME) trial was approved by the Food and Drug Administration (FDA, America) in 2015 (199,200). The TAME trial may make metformin the first approved anti-aging drug; as it is not testing metformin against a single disease, but rather a collection of age-related diseases, it establishes aging as a medical condition that can be intervened in or treated, rather than an irreversible process outside of human control. This shift in the concept of aging may make it easier to conduct future anti-aging clinical trials and the increase in the number of clinical trials associated with cellular senescence as a major contributor to IVDD may facilitate the clinical scope of investigating metformin for IVDD in future (201,202). Metformin is the first-line drug for T2DM, especially for obesity-associated and gestational diabetes (203). Metformin positively regulates obesity, aging and diabetes through AMPK pathway signaling, PI3K/AKT/mTOR pathway phosphorylation, oxidative stress, autophagy and other pathways to alleviate and improve disease progression (194,204). Ramanathan et al (33) confirmed that the inflammatory response in IVD treated with metformin is significantly inhibited. In addition, studies have shown that metformin promotes angiogenesis in the injured spinal cord by activating enzyme pathways, thereby improving neurological function after spinal cord injury in aged mice and promoting recovery of motor function following injury (205,206). Although metformin has not yet been used for clinical trials of IVDD, the aforementioned preclinical and clinical trials of diseases closely related to IVDD suggest potential mechanisms.

In clinical practice, metformin is often used as an adjunctive therapeutic agent. For example, metformin has anti-inflammatory properties (165). When combined with drugs that also have certain anti-inflammatory effects, metformin produces significant synergistic anti-inflammatory effects. Metformin and pioglitazone have different mechanisms of action on oxidative stress; combination of these two drugs is more effective in improving oxidative stress (161). Sildenafil + metformin + leucine and metformin + atorvastatin combination showed higher efficacy in lowering oxidative stress and decreasing inflammatory markers compared with each drug alone (162,163). IVDD often occurs in conjunction with DM, aging and inflammation (207). Especially in elderly patients with DM, chronic inflammation and IVDD, metformin can be used to treat DM as well as to slow down the further progression of IVDD. There have been numerous clinical studies of metformin in the treatment of OA (ICTRP, https://trialsearch.who.int/) (Table II), and there are many similarities between IVDD and OA, both in terms of the mechanism of occurrence and at the preventive and therapeutic levels (7,208-210). Specifically, for example, both IVDD and OA involve chronic inflammation and apoptotic processes, and elevated inflammatory markers such as IL-1β, IL-6, and TNF-α, to name a few, are present in patients with both IVDD and OA (211,212). Metformin may have potential for the treatment of IVDD. In the future, whether metformin can be used independently to delay IVDD or as an adjunctive drug to delay IVDD while assisting in the treatment of other diseases, and in what ways its benefits can be increased, need to be corroborated by basic research studies and clinical trials.

Table II

Clinical trials using metformin.

Table II

Clinical trials using metformin.

Trial ID no.TitleRecruitment statusPhasePrimary sponsorConditionPrimary outcome
NCT06328426Vitamin D With omega-3 or metformin in osteoarthritisNot recruitingN/AAssiut University (Assiut, Egypt)OsteoarthritisEffusion-synovitis volume on musculoskeletal ultrasound
IRCT20200522047538N2Evaluation of the effectiveness of metformin in WOMAC index of overweight knee osteoarthritis patientsRecruiting3Mashhad University of Medical Sciences (Mashhad, Iran)Knee. osteoarthritisWOMAC index of osteoarthritis before the study and 4 months later
NCT06231758Metformin safety and efficacy in osteoarthritisRecruiting3PhD Mostafa Bahaa (Damietta, Egypt)Knee osteoarthritisChange of VAS pain score from baseline to post-treatment
NCT06126029Effect of metformin as add-on therapy to ibuprofen on disease activity in knee osteoarthritis patientsRecruiting2Bangabandhu Sheikh Mujib Medical University (Dhaka, Bangladesh)Knee osteoarthritisKOOS
PACTR202311507335269Effects of metformin phonophoresis and exercise therapy on knee pain, range of motion, and function in knee osteoarthritisNot recruiting4Cairo University (Cairo, Egypt)Musculoskeletal diseaseKnee pain and function
NCT06096259Preventing injured knees from osteoarthritis: Severity outcomesNot recruiting2Brigham and Women's Hospital (Boston, United States)Knee osteoarthritis, post-traumatic osteoarthritis, ACL tearKOOS; modified MOAKS cartilage score
NCT05638893Metformin as adjuvant therapy in obese knee osteoarthritis patientsNot recruiting2/3Tanta University (Tanta, Egypt)Knee osteoarthritisWOMAC, weight
NCT05034029Effect of metformin on patients with osteoarthritisNot recruitingN/AGuangzhou First People's Hospital (Guangzhou, China)OsteoarthritisChange of tibiofemoral cartilage volume and WOMAC from baseline to 24-month follow up
ACTRN12621000710820Effectiveness of metformin for knee osteoarthritis with obesity-a randomised, double-blind, placebo-controlled trial of a potential disease modifying therapyNot recruiting3Prof Flavia Cicuttini (Melbourne, Australia)Knee osteoarthritis, obesityChange in VAS knee pain from baseline to 6-month follow-up
NCT04767841The AMPK modulator metformin as a novel adjunct to conventional therapy in patients with knee osteoarthritisRecruiting1/2Sadat City University (Sadat, Egypt)Knee osteoarthritisVAS assessment of pain
RCT20150202020908N2Evaluation of the molecular effects of metformin and improvement of symptoms in osteoarthritis patientsNot recruiting2/3Shiraz University of Medical Sciences (Shiraz, Iran)Knee osteoarthritisWOMAC questionnaire score at the beginning of the study and 16 weeks after starting metformin treatment
ChiCTR2000037081Clinical study of autologous fat glue alone or combined with metformin in the treatment of knee osteoarthritisRecruiting0Xinhua Hospital Affiliated to Medical College of Shanghai Jiaotong University (Shanghai, China)OsteoarthritisWeight, MRI, DNA methylation, knee function score, telomerase
ChiCTR2000036958Clinical study of autologous adipose derived mesenchymal stem cells alone or in combination with small molecule drugs in the treatment of osteoarthritisRecruiting0Xinhua Hospital Affiliated to Medical School of Shanghai Jiaotong University (Shanghai, China)OsteoarthritisMRI, DNA methylation, knee function score, telomerase

[i] WOMAC, Western Ontario and McMaster Universities osteoarthritis index; VAS, visual analog scale; KOOS, knee injury and osteoarthritis outcome score; MOAKS, Magnetic resonance imaging osteoarthritis knee score; ACL, Anterior cruciate ligament.

6. Perspectives

Although metformin affects IVDD in numerous molecular processes, there are still notable difficulties in its therapeutic use. The first is that the mechanism by which metformin acts on IVDD has not been fully elucidated and confirmed. Metformin has been shown to induce autophagy via AMPK, control MSC EV release and activate the cGAS/STING pathway to prevent apoptosis and senescence in NP cells (29-31). More research is required to understand additional distinct anti-inflammatory and autophagy-activating mechanisms of metformin in IVD cells. In addition, existing studies are cell culture and preclinical studies; to the best of our knowledge, there is a lack large-scale, randomized, double-blind, placebo-controlled clinical trials (29-33). Designing controls is challenging due to the number of potential confounding variables, including sample size, age, presence of other common conditions (particularly diabetes), adherence to treatment and follow-up accuracy. The drug concentration of oral metformin in the target area, feasibility of the local route of administration, the half-life of the drug and effect of the combined drug on efficacy are also factors that need to be explored in clinical experiments.

There are many studies showing that metformin does not lead to lower blood glucose when treating other conditions in non-diabetic patients because metformin does not stimulate insulin secretion (213-215). In recent years, metformin has been studied for the treatment of other clinical conditions, including cancer, heart disease and neurodegenerative disease such as IVDD (24). However, when metformin is used in non-diabetic patients, doctors may closely monitor their blood glucose levels to ensure they are in the normal range as other medications or health conditions may interact with metformin to affect blood glucose (216). Therefore, structure-activity relationship (SAR) or quantitative SAR studies could be considered to enhance the effect of metformin by modifying its structure to some extent and ensuring that it does not lower blood glucose levels.

Following oral administration, bioavailability of metformin is relatively low as it is primarily absorbed from the upper part of the small intestine (217). Incomplete absorption may be ameliorated by use of convenient drug delivery systems, such as bioadhesion and gastric retention drug delivery systems (218). In addition, due to the short biological half-life of metformin, repeated high doses of metformin are required, which usually results in decreased patient compliance and an increased incidence of side effects, such as diarrhoea, nausea and weight loss. Metformin also causes lactic acidosis, which is sometimes fatal (217). It is also necessary to develop new formulation strategies for metformin to improve its bioavailability, reduce the frequency of administration and minimize gastrointestinal side effects and toxicity. Drug delivery systems (such as particles, nanoparticles, liposomes and lipoid) are useful systems to overcome difficulties associated with conventional dosage forms. These systems offer advantages over conventional dosage forms, such as effective protection of the drug from enzymatic degradation, decreased drug side effects, improvement of patient comfort and compliance, control or site of drug release and improvement of relative bioavailability of the drug (219). In particulate dosage form, nanoparticle drug delivery systems are unique and may enhance targeted delivery of metformin to specific sites of action, which has been used as a promising approach for the treatment of cancer (220). However, whether metformin in the form of particles and nanoparticles as carriers is potentially important for its goal of delaying IVDD needs to be further investigated. Additionally, adverse medication reactions, particularly gastrointestinal symptoms, need to be detected and assessed further before metformin is used in the clinical treatment of IVDD. Besides, the combination of metformin with other drugs also needs to be considered in terms of drug interactions and adverse effects, as well as whether metformin can be used clinically for the treatment of IVDD. Specific experiments need to be designed to validate this on a case-by-case basis, as well as more basic research.

7. Conclusion

Globally, IVDD has notable health implications. For example, IVDD can lead to low back or neck pain (212), IVDD may lead to reduced disc height and spinal deformity, which can affect a patient's motor function (221), and IVDD may also lead to nerve compression, causing neurological symptoms such as sciatica and myelopathy (222). Metformin, as a first-line drug for the treatment of T2DM, may also contribute to the treatment of IVDD. The present review summarizes potential mechanisms of metformin for the treatment of IVDD. Metformin induces autophagy via the AMPK pathway and inhibits senescence and apoptosis of NP cells (29). Metformin promotes the production of EVs through autophagy and inactivates the cGAS/STING signalling pathway, improving the senescence of myeloid cells (30,31). Moreover, it has been shown that metformin inhibits senescence and apoptosis of medullary cells by suppressing inflammatory responses (32,33). Finally, metformin may act on IVDD by activating the SIRT signalling pathway, providing a theoretical basis for more basic and clinical studies of metformin in IVDD and associated disease. For elderly patients with both diabetes and IVDD-related diseases, metformin may serve as an adjunctive medication for the treatment of IVDD. The present review provides direction and for future preclinical and clinical studies of metformin in IVDD.

Availability of data and materials

Not applicable.

Authors' contributions

HT, JL and WY conceived the study. WY, YY, YW and ZG wrote and edited the manuscript. ZG and WY constructed figures and tables. JZ, WG and YC performed the literature review. YL, HW, LZ and YW revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 82072492) and the Excellent Youth Scientific Research Projects in Anhui Universities (grant no. 2022AH030117).

References

1 

Wang F, Cai F, Shi R, Wang XH and Wu XT: Aging and age related stresses: A senescence mechanism of intervertebral disc degeneration. Osteoarthritis Cartilage. 24:398–408. 2016. View Article : Google Scholar

2 

Cornaz F, Widmer J, Farshad-Amacker NA, Spirig JM, Snedeker JG and Farshad M: Biomechanical contributions of spinal structures with different degrees of disc degeneration. Spine (Phila Pa 1976). 46:E869–E877. 2021. View Article : Google Scholar

3 

Lan T, Shiyu H, Shen Z, Yan B and Chen J: New insights into the interplay between miRNAs and autophagy in the aging of intervertebral discs. Ageing Res Rev. 65:1012272021. View Article : Google Scholar

4 

Risbud MV and Shapiro IM: Role of cytokines in intervertebral disc degeneration: Pain and disc content. Nat Rev Rheumatol. 10:44–56. 2014. View Article : Google Scholar

5 

GBD 2019 Diseases and Injuries Collaborators: Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: A systematic analysis for the global burden of disease study 2019. Lancet. 396:1204–1222. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Yang Y, Lai X, Li C, Yang Y, Gu S, Hou W, Zhai L and Zhu Y: Focus on the impact of social factors and lifestyle on the disease burden of low back pain: Findings from the global burden of disease study 2019. BMC Musculoskelet Disord. 24:6792023. View Article : Google Scholar : PubMed/NCBI

7 

Goode AP, Carey TS and Jordan JM: Low back pain and lumbar spine osteoarthritis: How are they related? Curr Rheumatol Rep. 15:3052013. View Article : Google Scholar : PubMed/NCBI

8 

Maher C, Underwood M and Buchbinder R: Non-specific low back pain. Lancet. 389:736–747. 2017. View Article : Google Scholar

9 

Chou R: Low back pain. Ann Intern Med. 174:ITC113–ITC128. 2021. View Article : Google Scholar : PubMed/NCBI

10 

Apostolova N, Iannantuoni F, Gruevska A, Muntane J, Rocha M and Victor VM: Mechanisms of action of metformin in type 2 diabetes: Effects on mitochondria and leukocyte-endothelium interactions. Redox Biol. 34:1015172020. View Article : Google Scholar : PubMed/NCBI

11 

Sanchez-Rangel E and Inzucchi SE: Metformin: Clinical use in type 2 diabetes. Diabetologia. 60:1586–1593. 2017. View Article : Google Scholar : PubMed/NCBI

12 

LaMoia TE and Shulman GI: Cellular and molecular mechanisms of metformin action. Endocr Rev. 42:77–96. 2021. View Article : Google Scholar :

13 

An H and He L: Current understanding of metformin effect on the control of hyperglycemia in diabetes. J Endocrinol. 228:R97–R106. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Foretz M, Hébrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G, Sakamoto K, Andreelli F and Viollet B: Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 120:2355–2369. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Caton PW, Nayuni NK, Kieswich J, Khan NQ, Yaqoob MM and Corder R: Metformin suppresses hepatic gluconeogenesis through induction of SIRT1 and GCN5. J Endocrinol. 205:97–106. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Kim YD, Park KG, Lee YS, Park YY, Kim DK, Nedumaran B, Jang WG, Cho WJ, Ha J, Lee IK, et al: Metformin inhibits hepatic gluconeogenesis through AMP-activated protein kinase-dependent regulation of the orphan nuclear receptor SHP. Diabetes. 57:306–314. 2008. View Article : Google Scholar

17 

Masarwa R, Brunetti VC, Aloe S, Henderson M, Platt RW and Filion KB: Efficacy and safety of metformin for obesity: A systematic review. Pediatrics. 147:e202016102021. View Article : Google Scholar : PubMed/NCBI

18 

Day EA, Ford RJ, Smith BK, Mohammadi-Shemirani P, Morrow MR, Gutgesell RM, Lu R, Raphenya AR, Kabiri M, McArthur AG, et al: Metformin-induced increases in GDF15 are important for suppressing appetite and promoting weight loss. Nat Metab. 1:1202–1208. 2019. View Article : Google Scholar

19 

Kamarudin MNA, Sarker MMR, Zhou JR and Parhar I: Metformin in colorectal cancer: Molecular mechanism, preclinical and clinical aspects. J Exp Clin Cancer Res. 38:4912019. View Article : Google Scholar : PubMed/NCBI

20 

Luo F, Das A, Chen J, Wu P, Li X and Fang Z: Metformin in patients with and without diabetes: A paradigm shift in cardiovascular disease management. Cardiovasc Diabetol. 18:542019. View Article : Google Scholar : PubMed/NCBI

21 

Griffin SJ, Leaver JK and Irving GJ: Impact of metformin on cardiovascular disease: A meta-analysis of randomised trials among people with type 2 diabetes. Diabetologia. 60:1620–1629. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Diamanti-Kandarakis E, Economou F, Palimeri S and Christakou C: Metformin in polycystic ovary syndrome. Ann N Y Acad Sci. 1205:192–198. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Song A, Zhang C and Meng X: Mechanism and application of metformin in kidney diseases: An update. Biomed Pharmacother. 138:1114542021. View Article : Google Scholar : PubMed/NCBI

24 

Chen S, Gan D, Lin S, Zhong Y, Chen M, Zou X, Shao Z and Xiao G: Metformin in aging and aging-related diseases: Clinical applications and relevant mechanisms. Theranostics. 12:2722–2740. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Lu CH, Chung CH, Kuo FC, Chen KC, Chang CH, Kuo CC, Lee CH, Su SC, Liu JS, Lin FH, et al: Metformin attenuates osteoporosis in diabetic patients with carcinoma in situ: A nationwide, retrospective, matched-cohort study in Taiwan. J Clin Med. 9:28392020. View Article : Google Scholar : PubMed/NCBI

26 

Li J, Zhang B, Liu WX, Lu K, Pan H, Wang T, Oh CD, Yi D, Huang J, Zhao L, et al: Metformin limits osteoarthritis development and progression through activation of AMPK signalling. Ann Rheum Dis. 79:635–645. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Li X, Liu X, Wang Y, Cao F, Chen Z, Hu Z, Yu B, Feng H, Ba Z, Liu T, et al: Intervertebral disc degeneration in mice with type II diabetes induced by leptin receptor deficiency. BMC Musculoskelet Disord. 21:772020. View Article : Google Scholar : PubMed/NCBI

28 

Cannata F, Vadalà G, Ambrosio L, Fallucca S, Napoli N, Papalia R, Pozzilli P and Denaro V: Intervertebral disc degeneration: A focus on obesity and type 2 diabetes. Diabetes Metab Res Rev. 36:e32242020. View Article : Google Scholar

29 

Chen D, Xia D, Pan Z, Xu D, Zhou Y, Wu Y, Cai N, Tang Q, Wang C, Yan M, et al: Metformin protects against apoptosis and senescence in nucleus pulposus cells and ameliorates disc degeneration in vivo. Cell Death Dis. 7:e24412016. View Article : Google Scholar : PubMed/NCBI

30 

Liao Z, Li S, Lu S, Liu H, Li G, Ma L, Luo R, Ke W, Wang B, Xiang Q, et al: Metformin facilitates mesenchymal stem cell-derived extracellular nanovesicles release and optimizes therapeutic efficacy in intervertebral disc degeneration. Biomaterials. 274:1208502021. View Article : Google Scholar : PubMed/NCBI

31 

Ren C, Jin C, Li C, Xiang J, Wu Y, Zhou Y, Sun L, Zhang X and Tian N: Metformin inactivates the cGAS-STING pathway through autophagy and suppresses senescence in nucleus pulposus cells. J Cell Sci. 135:jcs2597382022. View Article : Google Scholar : PubMed/NCBI

32 

Han Y, Yuan F, Deng C, He F, Zhang Y, Shen H, Chen Z and Qian L: Metformin decreases LPS-induced inflammatory response in rabbit annulus fibrosus stem/progenitor cells by blocking HMGB1 release. Aging (Albany NY). 11:1252–10265. 2019.

33 

Ramanathan R, Firdous A, Dong Q, Wang D, Lee J, Vo N and Sowa G: Investigation into the anti-inflammatory properties of metformin in intervertebral disc cells. JOR Spine. 5:e11972022. View Article : Google Scholar : PubMed/NCBI

34 

Kaya YE, Karaarslan N, Yilmaz I, Sirin DY, Akalan H and Ozbek H: A study of the effects of metformin, a biguanide derivative, on annulus fibrosus and nucleus pulposus cells. Turk Neurosurg. 30:434–441. 2020.PubMed/NCBI

35 

Stephenne X, Foretz M, Taleux N, van der Zon GC, Sokal E, Hue L, Viollet B and Guigas B: Metformin activates AMP-activated protein kinase in primary human hepatocytes by decreasing cellular energy status. Diabetologia. 54:3101–3110. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Maruthur NM, Tseng E, Hutfless S, Wilson LM, Suarez-Cuervo C, Berger Z, Yue Chu Y, Iyoha E, Segal JB and Bolen S: Diabetes medications as monotherapy or metformin-based combination therapy for type 2 diabetes: A systematic review and meta-analysis. Ann Intern Med. 164:740–751. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Zhang F, Zhao X, Shen H and Zhang C: Molecular mechanisms of cell death in intervertebral disc degeneration (Review). Int J Mol Med. 37:1439–1448. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Gong CY and Zhang HH: Autophagy as a potential therapeutic target in intervertebral disc degeneration. Life Sci. 273:1192662021. View Article : Google Scholar : PubMed/NCBI

39 

Li J, Wang C, Xue L, Zhang F and Liu J: Melatonin suppresses apoptosis of nucleus pulposus cells through inhibiting autophagy via the PI3K/Akt pathway in a high-glucose culture. Biomed Res Int. 2021:46042582021.PubMed/NCBI

40 

Humzah MD and Soames RW: Human intervertebral disc: Structure and function. Anat Rec. 220:337–356. 1988. View Article : Google Scholar : PubMed/NCBI

41 

Bach FC, de Vries SAH, Krouwels A, Creemers LB, Ito K, Meij BP and Tryfonidou MA: The species-specific regenerative effects of notochordal cell-conditioned medium on chondrocyte-like cells derived from degenerated human intervertebral discs. Eur Cell Mater. 30:132–147. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Urban JPG and Roberts S: Degeneration of the intervertebral disc. Arthritis Res Ther. 5:120–130. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Dowdell J, Erwin M, Choma T, Vaccaro A, Iatridis J and Cho SK: Intervertebral disk degeneration and repair. Neurosurgery. 80(3S): S46–S54. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Oegema TR Jr: Biochemistry of the intervertebral disc. Clin Sports Med. 12:419–439. 1993. View Article : Google Scholar : PubMed/NCBI

45 

Adams MA and Roughley PJ: What is intervertebral disc degeneration, and what causes it? Spine (Phila Pa 1976). 31:2151–2161. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Bruehlmann SB, Rattner JB, Matyas JR and Duncan NA: Regional variations in the cellular matrix of the annulus fibrosus of the intervertebral disc. J Anat. 201:159–171. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Roberts S, Menage J, Duance V, Wotton S and Ayad S: 1991 Volvo Award in basic sciences. Collagen types around the cells of the intervertebral disc and cartilage end plate: An immunolocalization study. Spine (Phila Pa 1976). 16:1030–1038. 1991. View Article : Google Scholar : PubMed/NCBI

48 

Mwale F, Roughley P and Antoniou J: Distinction between the extracellular matrix of the nucleus pulposus and hyaline cartilage: A requisite for tissue engineering of intervertebral disc. Eur Cell Mater. 8:58–64. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Maroudas A, Stockwell RA, Nachemson A and Urban J: Factors involved in the nutrition of the human lumbar intervertebral disc: Cellularity and diffusion of glucose in vitro. J Anat. 120:113–130. 1975.PubMed/NCBI

50 

Yu J, Winlove PC, Roberts S and Urban JPG: Elastic fibre organization in the intervertebral discs of the bovine tail. J Anat. 201:465–475. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Pattappa G, Li Z, Peroglio M, Wismer N, Alini M and Grad S: Diversity of intervertebral disc cells: Phenotype and function. J Anat. 221:480–496. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Moon SM, Yoder JH, Wright AC, Smith LJ, Vresilovic EJ and Elliott DM: Evaluation of intervertebral disc cartilaginous endplate structure using magnetic resonance imaging. Eur Spine J. 22:1820–1828. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Broberg KB: On the mechanical behaviour of intervertebral discs. Spine (Phila Pa 1976). 8:151–165. 1983. View Article : Google Scholar : PubMed/NCBI

54 

Rider SM, Mizuno S and Kang JD: Molecular mechanisms of intervertebral disc degeneration. Spine Surg Relat Res. 3:1–11. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Raj PP: Intervertebral disc: Anatomy-physiology-pathophysiology-treatment. Pain Pract. 8:18–44. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Colombini A, Lombardi G, Corsi MM and Banfi G: Pathophysiology of the human intervertebral disc. Int J Biochem Cell Biol. 40:837–842. 2008. View Article : Google Scholar : PubMed/NCBI

57 

Lyons G, Eisenstein SM and Sweet MB: Biochemical changes in intervertebral disc degeneration. Biochim Biophys Acta. 673:443–453. 1981. View Article : Google Scholar : PubMed/NCBI

58 

Roughley PJ, Melching LI, Heathfield TF, Pearce RH and Mort JS: The structure and degradation of aggrecan in human intervertebral disc. Eur Spine J. 15(Suppl 3): S326–S332. 2006. View Article : Google Scholar : PubMed/NCBI

59 

Binch ALA, Shapiro IM and Risbud MV: Syndecan-4 in intervertebral disc and cartilage: Saint or synner? Matrix Biol. 52-54:355–362. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Xu K, Wang X, Zhang Q, Liang A, Zhu H, Huang D, Li C and Ye W: Sp1 downregulates proinflammatory cytokine-induced catabolic gene expression in nucleus pulposus cells. Mol Med Rep. 14:3961–3968. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Roughley PJ: Biology of intervertebral disc aging and degeneration: Involvement of the extracellular matrix. Spine (Phila Pa 1976). 29:2691–2699. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Sjöberg AP, Manderson GA, Mörgelin M, Day AJ, Heinegård D and Blom AM: Short leucine-rich glycoproteins of the extracellular matrix display diverse patterns of complement interaction and activation. Mol Immunol. 46:830–839. 2009. View Article : Google Scholar :

63 

Huang YC, Urban JP and Luk KD: Intervertebral disc regeneration: Do nutrients lead the way? Nat Rev Rheumatol. 10:561–566. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Zhang H, Zhu T, Zhang L and Wu Q: Stromal cell-derived factor-1 induces matrix metalloproteinase expression in human endplate chondrocytes, cartilage endplate degradation in explant culture, and the amelioration of nucleus pulposus degeneration in vivo. Int J Mol Med. 41:969–976. 2018.

65 

Hiyama A, Sakai D and Mochida J: Cell signaling pathways related to pain receptors in the degenerated disk. Global Spine J. 3:165–174. 2013. View Article : Google Scholar :

66 

Kondo N, Yuasa T, Shimono K, Tung W, Okabe T, Yasuhara R, Pacifici M, Zhang Y, Iwamoto M and Enomoto-Iwamoto M: Intervertebral disc development is regulated by Wnt/β-catenin signaling. Spine (Phila Pa 1976). 36:E513–E518. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Lotz JC and Ulrich JA: Innervation, inflammation, and hypermobility may characterize pathologic disc degeneration: review of animal model data. J Bone Joint Surg Am. 88(Suppl 2): S76–S82. 2006.

68 

Finkel T and Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature. 408:239–247. 2000. View Article : Google Scholar : PubMed/NCBI

69 

Olovnikov AM: A theory of marginotomy. The incomplete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenomenon. J Theor Biol. 41:181–190. 1973. View Article : Google Scholar : PubMed/NCBI

70 

Stein GH, Beeson M and Gordon L: Failure to phosphorylate the retinoblastoma gene product in senescent human fibroblasts. Science. 249:666–669. 1990. View Article : Google Scholar : PubMed/NCBI

71 

Feng C, Yang M, Lan M, Liu C, Zhang Y, Huang B, Liu H and Zhou Y: ROS: Crucial intermediators in the pathogenesis of intervertebral disc degeneration. Oxid Med Cell Longev. 2017:56015932017. View Article : Google Scholar : PubMed/NCBI

72 

Dimozi A, Mavrogonatou E, Sklirou A and Kletsas D: Oxidative stress inhibits the proliferation, induces premature senescence and promotes a catabolic phenotype in human nucleus pulposus intervertebral disc cells. Eur Cell Mater. 30:89–103. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Feng C, Liu H, Yang M, Zhang Y, Huang B and Zhou Y: Disc cell senescence in intervertebral disc degeneration: Causes and molecular pathways. Cell Cycle. 15:1674–1684. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Wang D, Hu Z, Hao J, He B, Gan Q, Zhong X, Zhang X, Shen J, Fang J and Jiang W: SIRT1 inhibits apoptosis of degenerative human disc nucleus pulposus cells through activation of Akt pathway. Age (Dordr). 35:1741–1753. 2013. View Article : Google Scholar

75 

Chen JW, Ni BB, Zheng XF, Li B, Jiang SD and Jiang LS: Hypoxia facilitates the survival of nucleus pulposus cells in serum deprivation by down-regulating excessive autophagy through restricting ROS generation. Int J Biochem Cell Biol. 59:1–10. 2015. View Article : Google Scholar

76 

Kim J, Kundu M, Viollet B and Guan KL: AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 13:132–141. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Xu HG, Yu YF, Zheng Q, Zhang W, Wang CD, Zhao XY, Tong WX, Wang H, Liu P and Zhang XL: Autophagy protects end plate chondrocytes from intermittent cyclic mechanical tension induced calcification. Bone. 66:232–239. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Wada T and Penninger JM: Mitogen-activated protein kinases in apoptosis regulation. Oncogene. 23:2838–2849. 2004. View Article : Google Scholar : PubMed/NCBI

79 

Deosarkar SD, Arsule AD and Kalyankar TM: Effect of antidiabetic metformin hydrochloride on physicochemical properties of cationic surfactant cetyltrimethylammonium bromide in aqueous solutions. Colloids Surf A: Physicochem Eng Asp. 613:1260522021. View Article : Google Scholar

80 

Dołowy M, Jampilek J and Bober-Majnusz K: A comparative study of the lipophilicity of metformin and phenformin. Molecules. 26:66132021. View Article : Google Scholar :

81 

Ismail AH, Al-Garawi ZS, Al-Shamari K and Salman AT: Metformin compounds: A review on the importance and the possible applications. J Phys Conf Ser. 1853:0120602021. View Article : Google Scholar

82 

He M, Lu B, Opoku M, Zhang L, Xie W, Jin H, Chen S, Li Y and Deng Z: Metformin prevents or delays the development and progression of osteoarthritis: New insight and mechanism of action. Cells. 11:30122022. View Article : Google Scholar : PubMed/NCBI

83 

Graham GG, Punt J, Arora M, Day RO, Doogue MP, Duong JK, Furlong TJ, Greenfield JR, Greenup LC, Kirkpatrick CM, et al: Clinical pharmacokinetics of metformin. Clin Pharmacokinet. 50:81–98. 2011. View Article : Google Scholar : PubMed/NCBI

84 

Bailey CJ: Metformin: Historical overview. Diabetologia. 60:1566–1576. 2017. View Article : Google Scholar : PubMed/NCBI

85 

He L: Metformin and systemic metabolism. Trends Pharmacol Sci. 41:868–881. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Tucker GT, Casey C, Phillips PJ, Connor H, Ward JD and Woods HF: Metformin kinetics in healthy subjects and in patients with diabetes mellitus. Br J Clin Pharmacol. 12:235–246. 1981. View Article : Google Scholar : PubMed/NCBI

87 

Wilcock C and Bailey CJ: Accumulation of metformin by tissues of the normal and diabetic mouse. Xenobiotica. 24:49–57. 1994. View Article : Google Scholar : PubMed/NCBI

88 

Sambol NC, Brookes LG, Chiang J, Goodman AM, Lin ET, Liu CY and Benet LZ: Food intake and dosage level, but not tablet vs solution dosage form, affect the absorption of metformin HCl in man. Br J Clin Pharmacol. 42:510–512. 1996. View Article : Google Scholar : PubMed/NCBI

89 

Song Y, Wu Z and Zhao P: The function of metformin in aging-related musculoskeletal disorders. Front Pharmacol. 13:8655242022. View Article : Google Scholar : PubMed/NCBI

90 

Chen S, Ruan G, Zeng M, Chen T, Cao P, Zhang Y, Li J, Wang X, Li S, Tang S, et al: Association between metformin use and risk of total knee arthroplasty and degree of knee pain in knee osteoarthritis patients with diabetes and/or obesity: A retrospective study. J Clin Med. 11:47962022. View Article : Google Scholar : PubMed/NCBI

91 

El-Arabey AA, Abdalla M and Ali Eltayb W: Metformin: Ongoing journey with superdrug revolution. Adv Pharm Bull. 9:1–4. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Mazor M, Best TM, Cesaro A, Lespessailles E and Toumi H: Osteoarthritis biomarker responses and cartilage adaptation to exercise: A review of animal and human models. Scand J Med Sci Sports. 29:1072–1082. 2019. View Article : Google Scholar : PubMed/NCBI

93 

Vinatier C, Merceron C and Guicheux J: Osteoarthritis: From pathogenic mechanisms and recent clinical developments to novel prospective therapeutic options. Drug Discov Today. 21:1932–1937. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Williams S, Alkhatib B and Serra R: Development of the axial skeleton and intervertebral disc. Curr Top Dev Biol. 133:49–90. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Li X, Lee JP, Balian G and Greg Anderson D: Modulation of chondrocytic properties of fat-derived mesenchymal cells in co-cultures with nucleus pulposus. Connect Tissue Res. 46:75–82. 2005. View Article : Google Scholar : PubMed/NCBI

96 

Jørgensen SB, Richter EA and Wojtaszewski JF: Role of AMPK in skeletal muscle metabolic regulation and adaptation in relation to exercise. J Physiol. 574:17–31. 2006. View Article : Google Scholar : PubMed/NCBI

97 

Lee WJ, Kim M, Park HS, Kim HS, Jeon MJ, Oh KS, Koh EH, Won JC, Kim MS, Oh GT, et al: AMPK activation increases fatty acid oxidation in skeletal muscle by activating PPARalpha and PGC-1. Biochem Biophys Res Commun. 340:291–295. 2006. View Article : Google Scholar

98 

Lee-Young RS, Canny BJ, Myers DE and McConell GK: AMPK activation is fiber type specific in human skeletal muscle: effects of exercise and short-term exercise training. J Appl Physiol (1985). 107:283–289. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Bahrambeigi S, Yousefi B, Rahimi M and Shafiei-Irannejad V: Metformin; an old antidiabetic drug with new potentials in bone disorders. Biomed Pharmacother. 109:1593–1601. 2019. View Article : Google Scholar

100 

Qin X, Jiang T, Liu S, Tan J, Wu H, Zheng L and Zhao J: Effect of metformin on ossification and inflammation of fibroblasts in ankylosing spondylitis: An in vitro study. J Cell Biochem. 119:1074–1082. 2018. View Article : Google Scholar

101 

Ma T, Tian X, Zhang B, Li M, Wang Y, Yang C, Wu J, Wei X, Qu Q, Yu Y, et al: Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature. 603:159–165. 2022. View Article : Google Scholar : PubMed/NCBI

102 

Sakamoto K and Jessen N: PEN2: Metformin's new partner at lysosome. Cell Res. 32:507–508. 2022. View Article : Google Scholar : PubMed/NCBI

103 

Liu C, Zhang S, Xue J, Zhang H and Yin J: Evaluation of PEN2-ATP6AP1 axis as an antiparasitic target for metformin based on phylogeny analysis and molecular docking. Mol Biochem Parasitol. 255:1115802023. View Article : Google Scholar : PubMed/NCBI

104 

Dong J, Xu X, Zhang Q, Yuan Z and Tan B: The PI3K/AKT pathway promotes fracture healing through its crosstalk with Wnt/β-catenin. Exp Cell Res. 394:1121372020. View Article : Google Scholar

105 

Watabe H, Furuhama T, Tani-Ishii N and Mikuni-Takagaki Y: Mechanotransduction activates α5β1 integrin and PI3K/Akt signaling pathways in mandibular osteoblasts. Exp Cell Res. 317:2642–2649. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Wang Z, Ma F, Wang J, Zhou Z, Liu B, He X, Fu L, He W and Cooper PR: Extracellular signal-regulated kinase mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt signaling are required for lipopolysaccharide-mediated mineralization in murine odontoblast-like cells. J Endod. 41:871–876. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Tang Y, Mo Y, Xin D, Xiong Z, Zeng L, Luo G and Cao Y: Regulation of osteoblast autophagy based on PI3K/AKT/mTOR signaling pathway study on the effect of β-ecdysterone on fracture healing. J Orthop Surg Res. 16:7192021. View Article : Google Scholar

108 

Lee SH, Lee JH, Lee HY and Min KJ: Sirtuin signaling in cellular senescence and aging. BMB Rep. 52:24–34. 2019. View Article : Google Scholar :

109 

Hu X, Feng G, Meng Z, Ma L and Jin Q: The protective mechanism of SIRT1 on cartilage through regulation of LEF-1. BMC Musculoskelet Disord. 22:6422021. View Article : Google Scholar : PubMed/NCBI

110 

Wang C, Yang Y, Zhang Y, Liu J, Yao Z and Zhang C: Protective effects of metformin against osteoarthritis through upregulation of SIRT3-mediated PINK1/Parkin-dependent mitophagy in primary chondrocytes. Biosci Trends. 12:605–612. 2019. View Article : Google Scholar

111 

Chen Y, Wu YY, Si HB, Lu YR and Shen B: Mechanistic insights into AMPK-SIRT3 positive feedback loop-mediated chondrocyte mitochondrial quality control in osteoarthritis pathogenesis. Pharmacol Res. 166:1054972021. View Article : Google Scholar : PubMed/NCBI

112 

Lin S, Guo H, You X, Zhang Z and Ye H: SND1 aggravates mitochondrial damage, apoptosis and extracellular matrix degradation in IL-1β-stimulated chondrocytes via PINK1/BECN1 pathway. Eur J Med Res. 28:3712023. View Article : Google Scholar

113 

Hu D, Xie F, Xiao Y, Lu C, Zhong J, Huang D, Chen J, Wei J, Jiang Y and Zhong T: Metformin: A potential candidate for targeting aging mechanisms. Aging Dis. 12:480–493. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Xing H, Liang C, Wang C, Xu X, Hu Y and Qiu B: Metformin mitigates cholesterol accumulation via the AMPK/SIRT1 pathway to protect osteoarthritis chondrocytes. Biochem Biophys Res Commun. 632:113–121. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Kang W, Wang T, Hu Z, Liu F, Sun Y and Ge S: Metformin inhibits porphyromonas gingivalis lipopolysaccharide-influenced inflammatory response in human gingival fibroblasts via regulating activating transcription factor-3 expression. J Periodontol. 88:e169–e178. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Saluja M, Pareek KK and Swami YK: Study of diversity of metformin related gastrointestinal side effects. J Assoc Physicians India. 68:36–38. 2020.PubMed/NCBI

117 

Lamos EM, Stein SA and Davis SN: Combination of glibenclamide-metformin HCl for the treatment of type 2 diabetes mellitus. Expert Opin Pharmacother. 13:2545–2554. 2012. View Article : Google Scholar : PubMed/NCBI

118 

Stades AME, Heikens JT, Erkelens DW, Holleman F and Hoekstra JBL: Metformin and lactic acidosis: Cause or coincidence? A review of case reports. J Intern Med. 255:179–187. 2004. View Article : Google Scholar : PubMed/NCBI

119 

Eppenga WL, Lalmohamed A, Geerts AF, Derijks HJ, Wensing M, Egberts A, De Smet PA and de Vries F: Risk of lactic acidosis or elevated lactate concentrations in metformin users with renal impairment: A population-based cohort study. Diabetes Care. 37:2218–2224. 2014. View Article : Google Scholar : PubMed/NCBI

120 

O CK, Siu BW, Leung VW, Lin YY, Ding CZ, Lau ES, Luk AO, Chow EY, Ma RC, Chan JC, et al: Association of insomnia with incident chronic cognitive impairment in older adults with type 2 diabetes mellitus: A prospective study of the Hong Kong diabetes register. J Diabetes Complications. 37:1085982023. View Article : Google Scholar : PubMed/NCBI

121 

Wiwanitkit S and Wiwanitkit V: Metformin and sleep disorders. Indian J Endocrinol Metab. 16(Suppl 1): S63–S64. 2012. View Article : Google Scholar : PubMed/NCBI

122 

Rosekind MR and Gregory KB: Insomnia risks and costs: health, safety, and quality of life. Am J Manag Care. 16:617–626. 2010.PubMed/NCBI

123 

Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Mannerås-Holm L, Ståhlman M, Olsson LM, Serino M, Planas-Fèlix M, et al: Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med. 23:850–858. 2017. View Article : Google Scholar : PubMed/NCBI

124 

Salvatore T, Pafundi PC, Marfella R, Sardu C, Rinaldi L, Monaco L, Ricozzi C, Imbriani S, Nevola R, Adinolfi LE and Sasso FC: Metformin lactic acidosis: Should we still be afraid? Diabetes Res Clin Pract. 157:1078792019. View Article : Google Scholar : PubMed/NCBI

125 

Rahman F and Tuba S: Lactic acidosis associated with metformin in patients with diabetic kidney disease. Med Arch. 76:297–300. 2022. View Article : Google Scholar : PubMed/NCBI

126 

Hardie DG, Ross FA and Hawley SA: AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 13:251–262. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Du J, Xu M, Kong F, Zhu P, Mao Y, Liu Y, Zhou H, Dong Z, Yu Z, Du T, et al: CB2R attenuates intervertebral disc degeneration by delaying nucleus pulposus cell senescence through AMPK/GSK3β pathway. Aging Dis. 13:552–267. 2022. View Article : Google Scholar : PubMed/NCBI

128 

Zhang Z, Wu J, Teng C, Wang J, Yu J, Jin C, Wang L, Wu L and Lin Z, Yu Z and Lin Z: Orientin downregulating oxidative stress-mediated endoplasmic reticulum stress and mitochondrial dysfunction through AMPK/SIRT1 pathway in rat nucleus pulposus cells in vitro and attenuated intervertebral disc degeneration in vivo. Apoptosis. 27:1031–1048. 2022. View Article : Google Scholar : PubMed/NCBI

129 

Harada M, Tadevosyan A, Qi X, Xiao J, Liu T, Voigt N, Karck M, Kamler M, Kodama I, Murohara T, et al: Atrial fibrillation activates AMP-dependent protein kinase and its regulation of cellular calcium handling: Potential role in metabolic adaptation and prevention of progression. J Am Coll Cardiol. 66:47–58. 2015. View Article : Google Scholar : PubMed/NCBI

130 

Zhang GZ, Deng YJ, Xie QQ, Ren EH, Ma ZJ, He XG, Gao YC and Kang XW: Sirtuins and intervertebral disc degeneration: Roles in inflammation, oxidative stress, and mitochondrial function. Clin Chim Acta. 508:33–42. 2020. View Article : Google Scholar : PubMed/NCBI

131 

Liu Y, Lin J, Wu X, Guo X, Sun H, Yu B, Shen J, Bai J, Chen Z, Yang H, et al: Aspirin-mediated attenuation of intervertebral disc degeneration by ameliorating reactive oxygen species in vivo and in vitro. Oxid Med Cell Longev. 2019:71898542019. View Article : Google Scholar : PubMed/NCBI

132 

Dai X, Chen Y, Yu Z, Liao C, Liu Z, Chen J and Wu Q: Advanced oxidation protein products induce annulus fibrosus cell senescence through a NOX4-dependent, MAPK-mediated pathway and accelerate intervertebral disc degeneration. PeerJ. 10:e138262022. View Article : Google Scholar : PubMed/NCBI

133 

Chen JW, Ni BB, Li B, Yang YH, Jiang SD and Jiang LS: The responses of autophagy and apoptosis to oxidative stress in nucleus pulposus cells: Implications for disc degeneration. Cell Physiol Biochem. 34:1175–1189. 2014. View Article : Google Scholar : PubMed/NCBI

134 

Brennan MÁ, Layrolle P and Mooney DJ: Biomaterials functionalized with MSC secreted extracellular vesicles and soluble factors for tissue regeneration. Adv Funct Mater. 30:19091252020. View Article : Google Scholar : PubMed/NCBI

135 

Varderidou-Minasian S and Lorenowicz MJ: Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: Challenges and opportunities. Theranostics. 10:5979–5997. 2020. View Article : Google Scholar : PubMed/NCBI

136 

Rani S, Ryan AE, Griffin MD and Ritter T: Mesenchymal stem cell-derived extracellular vesicles: Toward cell-free therapeutic applications. Mol Ther. 23:812–823. 2015. View Article : Google Scholar : PubMed/NCBI

137 

Latifkar A, Ling L, Hingorani A, Johansen E, Clement A, Zhang X, Hartman J, Fischbach C, Lin H, Cerione RA and Antonyak MA: Loss of sirtuin 1 alters the secretome of breast cancer cells by impairing lysosomal integrity. Dev Cell. 49:393–408.e7. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Minakaki G, Menges S, Kittel A, Emmanouilidou E, Schaeffner I, Barkovits K, Bergmann A, Rockenstein E, Adame A, Marxreiter F, et al: Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid autophagosome-exosome-like phenotype. Autophagy. 14:98–119. 2018. View Article : Google Scholar :

139 

Wang Y, Xu W, Yan Z, Zhao W, Mi J, Li J and Yan H: Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. J Exp Clin Cancer Res. 37:632018. View Article : Google Scholar : PubMed/NCBI

140 

Shen M, Yu H, Jin Y, Mo J, Sui J, Qian X and Chen T: Metformin facilitates osteoblastic differentiation and M2 macrophage polarization by PI3K/AKT/mTOR pathway in human umbilical cord mesenchymal stem cells. Stem Cells Int. 2022:94988762022. View Article : Google Scholar : PubMed/NCBI

141 

Han X, Chen H, Gong H, Tang X, Huang N, Xu W, Tai H, Zhang G, Zhao T, Gong C, et al: Autolysosomal degradation of cytosolic chromatin fragments antagonizes oxidative stress-induced senescence. J Biol Chem. 295:4451–4463. 2020. View Article : Google Scholar : PubMed/NCBI

142 

Tang Z, Hu B, Zang F, Wang J, Zhang X and Chen H: Nrf2 drives oxidative stress-induced autophagy in nucleus pulposus cells via a Keap1/Nrf2/p62 feedback loop to protect intervertebral disc from degeneration. Cell Death Dis. 10:5102019. View Article : Google Scholar : PubMed/NCBI

143 

Wang J, Huang C, Lin Z, Pan X, Chen J, Zheng G, Tian N, Yan Y, Zhang Z, Hu J, et al: Polydatin suppresses nucleus pulposus cell senescence, promotes matrix homeostasis and attenuates intervertebral disc degeneration in rats. J Cell Mol Med. 22:5720–5731. 2018. View Article : Google Scholar : PubMed/NCBI

144 

Kang L, Zhang H, Jia C, Zhang R and Shen C: Targeting oxidative stress and inflammation in intervertebral disc degeneration: Therapeutic perspectives of phytochemicals. Front Pharmacol. 13:9563552022. View Article : Google Scholar : PubMed/NCBI

145 

Glück S, Guey B, Gulen MF, Wolter K, Kang TW, Schmacke NA, Bridgeman A, Rehwinkel J, Zender L and Ablasser A: Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence. Nat Cell Biol. 19:1061–1070. 2017. View Article : Google Scholar : PubMed/NCBI

146 

Cheng Z, Dai T, He X, Zhang Z, Xie F, Wang S, Zhang L and Zhou F: The interactions between cGAS-STING pathway and pathogens. Signal Transduct Target Ther. 5:912020. View Article : Google Scholar : PubMed/NCBI

147 

Chen Q, Sun L and Chen ZJ: Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 17:1142–1149. 2016. View Article : Google Scholar : PubMed/NCBI

148 

Hu MM and Shu HB: Innate immune response to cytoplasmic DNA: Mechanisms and DIseases. Annu Rev Immunol. 38:79–98. 2020. View Article : Google Scholar

149 

Motwani M, Pesiridis S and Fitzgerald KA: DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet. 20:657–674. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Kim J, Kim HS and Chung JH: Molecular mechanisms of mitochondrial DNA release and activation of the cGAS-STING pathway. Exp Mol Med. 55:510–519. 2023. View Article : Google Scholar : PubMed/NCBI

151 

Guo Y, Gu R, Gan D, Hu F, Li G and Xu G: Mitochondrial DNA drives noncanonical inflammation activation via cGAS-STING signaling pathway in retinal microvascular endothelial cells. Cell Commun Signal. 18:1722020. View Article : Google Scholar : PubMed/NCBI

152 

Ritchie C, Carozza JA and Li L: Biochemistry, cell biology, and pathophysiology of the innate immune cGAS-cGAMP-STING pathway. Annu Rev Biochem. 91:599–628. 2022. View Article : Google Scholar : PubMed/NCBI

153 

Lawrence T: The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 1:a0016512009. View Article : Google Scholar

154 

Vo NV, Sowa GA, Kang JD, Seidel C and Studer RK: Prostaglandin E2 and prostaglandin F2α differentially modulate matrix metabolism of human nucleus pulposus cells. J Orthop Res. 28:1259–1266. 2010. View Article : Google Scholar : PubMed/NCBI

155 

Tisherman R, Coelho P, Phillibert D, Wang D, Dong Q, Vo N, Kang J and Sowa G: NF-κB signaling pathway in controlling intervertebral disk cell response to inflammatory and mechanical stressors. Phys Ther. 96:704–711. 2016. View Article : Google Scholar : PubMed/NCBI

156 

Ren H, Shao Y, Wu C, Ma X, Lv C and Wang Q: Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol Cell Endocrinol. 500:1106282020. View Article : Google Scholar

157 

Chen Q, Guan X, Zuo X, Wang J and Yin W: The role of high mobility group box 1 (HMGB1) in the pathogenesis of kidney diseases. Acta Pharm Sin B. 6:183–188. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Tang D, Kang R, Van Houten B, Zeh HJ, Billiar TR and Lotze MT: High mobility group box 1 (HMGB1) phenotypic role revealed with stress. Mol Med. 20:359–362. 2014. View Article : Google Scholar : PubMed/NCBI

159 

Leclerc P, Wähämaa H, Idborg H, Jakobsson PJ, Harris HE and Korotkova M: IL-1β/HMGB1 complexes promote The PGE2 biosynthesis pathway in synovial fibroblasts. Scand J Immunol. 77:350–360. 2013. View Article : Google Scholar : PubMed/NCBI

160 

Appavoo E, Hajam IA, Muneeswaran NS, Kondabattula G, Bhanuprakash V and Kishore S: Synergistic effect of high-mobility group box-1 and lipopolysaccharide on cytokine induction in bovine peripheral blood mononuclear cells. Microbiol Immunol. 60:196–202. 2016. View Article : Google Scholar

161 

Wang Y, Che M, Xin J, Zheng Z, Li J and Zhang S: The role of IL-1β and TNF-α in intervertebral disc degeneration. Biomed Pharmacother. 131:1106602020. View Article : Google Scholar

162 

Song Y, Wang Y, Zhang Y, Geng W, Liu W, Gao Y, Li S, Wang K, Wu X, Kang L and Yang C: Advanced glycation end products regulate anabolic and catabolic activities via NLRP3-inflammasome activation in human nucleus pulposus cells. J Cell Mol Med. 21:1373–1387. 2017. View Article : Google Scholar : PubMed/NCBI

163 

Dong HC, Li PN, Chen CJ, Xu X, Zhang H, Liu G, Zheng LJ and Li P: Sinomenine attenuates cartilage degeneration by regulating miR-223-3p/NLRP3 inflammasome signaling. Inflammation. 42:1265–1275. 2019. View Article : Google Scholar : PubMed/NCBI

164 

Xu H, Mei Q, Xu B, Liu G and Zhao J: Expression of matrix metalloproteinases is positively related to the severity of disc degeneration and growing age in the East Asian lumbar disc herniation patients. Cell Biochem Biophys. 70:1219–1225. 2014. View Article : Google Scholar : PubMed/NCBI

165 

Vo NV, Hartman RA, Yurube T, Jacobs LJ, Sowa GA and Kang JD: Expression and regulation of metalloproteinases and their inhibitors in intervertebral disc aging and degeneration. Spine J. 13:331–341. 2013. View Article : Google Scholar : PubMed/NCBI

166 

Liu Q, Jin L, Shen FH, Balian G and Li XJ: Fullerol nanoparticles suppress inflammatory response and adipogenesis of vertebral bone marrow stromal cells-a potential novel treatment for intervertebral disc degeneration. Spine J. 13:1571–1580. 2013. View Article : Google Scholar : PubMed/NCBI

167 

Xian H, Liu Y, Rundberg Nilsson A, Gatchalian R, Crother TR, Tourtellotte WG, Zhang Y, Aleman-Muench GR, Lewis G, Chen W, et al: Metformin inhibition of mitochondrial ATP and DNA synthesis abrogates NLRP3 inflammasome activation and pulmonary inflammation. Immunity. 54:1463–1477.e11. 2021. View Article : Google Scholar : PubMed/NCBI

168 

Jiang Y, Xie Z, Yu J and Fu L: Resveratrol inhibits IL-1β-mediated nucleus pulposus cell apoptosis through regulating the PI3K/Akt pathway. Biosci Rep. 39:BSR201900432019. View Article : Google Scholar

169 

Isoda K, Young JL, Zirlik A, MacFarlane LA, Tsuboi N, Gerdes N, Schönbeck U and Libby P: Metformin inhibits proinflammatory responses and nuclear factor-kappaB in human vascular wall cells. Arterioscler Thromb Vasc Biol. 26:611–617. 2006. View Article : Google Scholar

170 

Carafa V, Rotili D, Forgione M, Cuomo F, Serretiello E, Hailu GS, Jarho E, Lahtela-Kakkonen M, Mai A and Altucci L: Sirtuin functions and modulation: From chemistry to the clinic. Clin Epigenetics. 8:612016. View Article : Google Scholar : PubMed/NCBI

171 

Dai H, Sinclair DA, Ellis JL and Steegborn C: Sirtuin activators and inhibitors: Promises, achievements, and challenges. Pharmacol Ther. 188:140–154. 2018. View Article : Google Scholar : PubMed/NCBI

172 

Ohguchi K, Itoh T, Akao Y, Inoue H, Nozawa Y and Ito M: SIRT1 modulates expression of matrix metalloproteinases in human dermal fibroblasts. Br J Dermatol. 163:689–694. 2010. View Article : Google Scholar : PubMed/NCBI

173 

Dvir-Ginzberg M, Gagarina V, Lee EJ and Hall DJ: Regulation of cartilage-specific gene expression in human chondrocytes by SirT1 and nicotinamide phosphoribosyltransferase. J Biol Chem. 283:36300–36310. 2008. View Article : Google Scholar : PubMed/NCBI

174 

Qu L, Yu Y, Qiu L, Yang D, Yan L, Guo J and Jahan R: Sirtuin 1 regulates matrix metalloproteinase-13 expression induced by Porphyromonas endodontalis lipopolysaccharide via targeting nuclear factor-κB in osteoblasts. J Oral Microbiol. 9:13175782017. View Article : Google Scholar

175 

Abdelmawgoud H and El Awady RR: Effect of Sirtuin 1 inhibition on matrix metalloproteinase 2 and Forkhead box O3a expression in breast cancer cells. Genes Dis. 4:240–246. 2017. View Article : Google Scholar

176 

Gagarina V, Gabay O, Dvir-Ginzberg M, Lee EJ, Brady JK, Quon MJ and Hall DJ: SirT1 enhances survival of human osteoarthritic chondrocytes by repressing protein tyrosine phosphatase 1B and activating the insulin-like growth factor receptor pathway. Arthritis Rheum. 62:1383–1392. 2010. View Article : Google Scholar : PubMed/NCBI

177 

Song YM, Lee YH, Kim JW, Ham DS, Kang ES, Cha BS, Lee HC and Lee BW: Metformin alleviates hepatosteatosis by restoring SIRT1-mediated autophagy induction via an AMP-activated protein kinase-independent pathway. Autophagy. 11:46–59. 2015. View Article : Google Scholar :

178 

Rogacka D, Audzeyenka I, Rychłowski M, Rachubik P, Szrejder M, Angielski S and Piwkowska A: Metformin overcomes high glucose-induced insulin resistance of podocytes by pleiotropic effects on SIRT1 and AMPK. Biochim Biophys Acta Mol Basis Dis. 1864:115–125. 2018. View Article : Google Scholar

179 

Shen J, Lan Y, Ji Z and Liu H: Sirtuins in intervertebral disc degeneration: Current understanding. Mol Med. 30:442024. View Article : Google Scholar : PubMed/NCBI

180 

Xu J, Shao T, Lou J, Zhang J and Xia C: Aging, cell senescence, the pathogenesis and targeted therapies of intervertebral disc degeneration. Front Pharmacol. 14:11729202023. View Article : Google Scholar : PubMed/NCBI

181 

Song Y, Li S, Geng W, Luo R, Liu W, Tu J, Wang K, Kang L, Yin H, Wu X, et al: Sirtuin 3-dependent mitochondrial redox homeostasis protects against AGEs-induced intervertebral disc degeneration. Redox Biol. 19:339–353. 2018. View Article : Google Scholar : PubMed/NCBI

182 

Lin J, Du J, Wu X, Xu C, Liu J, Jiang L, Cheng X, Ge G, Chen L, Pang Q, et al: SIRT3 mitigates intervertebral disc degeneration by delaying oxidative stress-induced senescence of nucleus pulposus cells. J Cell Physiol. 236:6441–6456. 2021. View Article : Google Scholar : PubMed/NCBI

183 

Hu B, Wang P, Zhang S, Liu W, Lv X, Shi D, Zhao L, Liu H, Wang B, Chen S and Shao Z: HSP70 attenuates compression-induced apoptosis of nucleus pulposus cells by suppressing mitochondrial fission via upregulating the expression of SIRT3. Exp Mol Med. 54:309–323. 2022. View Article : Google Scholar : PubMed/NCBI

184 

Zhou TY, Wu YG, Zhang YZ, Bao YW and Zhao Y: SIRT3 retards intervertebral disc degeneration by anti-oxidative stress by activating the SIRT3/FOXO3/SOD2 signaling pathway. Eur Rev Med Pharmacol Sci. 23:9180–9188. 2019.PubMed/NCBI

185 

Karnewar S, Neeli PK, Panuganti D, Kotagiri S, Mallappa S, Jain N, Jerald MK and Kotamraju S: Metformin regulates mitochondrial biogenesis and senescence through AMPK mediated H3K79 methylation: Relevance in age-associated vascular dysfunction. Biochim Biophys Acta Mol Basis Dis. 1864:1115–1128. 2018. View Article : Google Scholar : PubMed/NCBI

186 

Du Y and Zhang J, Fang F, Wei X, Zhang H, Tan H and Zhang J: Metformin ameliorates hypoxia/reoxygenation-induced cardio-myocyte apoptosis based on the SIRT3 signaling pathway. Gene. 626:182–188. 2017. View Article : Google Scholar : PubMed/NCBI

187 

Chen J, Xie JJ, Jin MY, Gu YT, Wu CC, Guo WJ, Yan YZ, Zhang ZJ, Wang JL, Zhang XL, et al: Sirt6 overexpression suppresses senescence and apoptosis of nucleus pulposus cells by inducing autophagy in a model of intervertebral disc degeneration. Cell Death Dis. 9:562018. View Article : Google Scholar : PubMed/NCBI

188 

Shan L, Wang F, Zhai D, Meng X, Liu J and Lv X: Matrix metalloproteinases induce extracellular matrix degradation through various pathways to alleviate hepatic fibrosis. Biomed Pharmacother. 161:1144722023. View Article : Google Scholar : PubMed/NCBI

189 

Valenti MT, Dalle Carbonare L, Zipeto D and Mottes M: Control of the autophagy pathway in osteoarthritis: Key regulators, therapeutic targets and therapeutic strategies. Int J Mol Sci. 22:27002021. View Article : Google Scholar : PubMed/NCBI

190 

Ryter SW, Cloonan SM and Choi AM: Autophagy: A critical regulator of cellular metabolism and homeostasis. Mol Cells. 36:7–16. 2013. View Article : Google Scholar : PubMed/NCBI

191 

Mu W and Wang Z, Ma C, Jiang Y, Zhang N, Hu K, Li L and Wang Z: Metformin promotes the proliferation and differentiation of murine preosteoblast by regulating the expression of sirt6 and oct4. Pharmacol Res. 129:462–474. 2018. View Article : Google Scholar

192 

Yang M, Peng Y, Liu W, Zhou M, Meng Q and Yuan C: Sirtuin 2 expression suppresses oxidative stress and senescence of nucleus pulposus cells through inhibition of the p53/p21 pathway. Biochem Biophys Res Commun. 513:616–622. 2019. View Article : Google Scholar : PubMed/NCBI

193 

Tang X, Chen XF, Wang NY, Wang XM, Liang ST, Zheng W, Lu YB, Zhao X, Hao DL, Zhang ZQ, et al: SIRT2 acts as a cardioprotective deacetylase in pathological cardiac hypertrophy. Circulation. 136:2051–2067. 2017. View Article : Google Scholar : PubMed/NCBI

194 

Dutta S, Shah RB, Singhal S, Dutta SB, Bansal S, Sinha S and Haque M: Metformin: A review of potential mechanism and therapeutic utility beyond diabetes. Drug Des Devel Ther. 17:1907–1932. 2023. View Article : Google Scholar : PubMed/NCBI

195 

Yanovski JA, Krakoff J, Salaita CG, McDuffie JR, Kozlosky M, Sebring NG, Reynolds JC, Brady SM and Calis KA: Effects of metformin on body weight and body composition in obese insulin-resistant children: A randomized clinical trial. Diabetes. 60:477–485. 2011. View Article : Google Scholar : PubMed/NCBI

196 

Seifarth C, Schehler B and Schneider HJ: Effectiveness of metformin on weight loss in non-diabetic individuals with obesity. Exp Clin Endocrinol Diabetes. 121:27–31. 2013.

197 

Piskovatska V, Storey KB, Vaiserman AM and Lushchak O: The use of metformin to increase the human healthspan. Adv Exp Med Biol. 1260:319–332. 2020. View Article : Google Scholar : PubMed/NCBI

198 

Valencia WM, Palacio A, Tamariz L and Florez H: Metformin and ageing: Improving ageing outcomes beyond glycaemic control. Diabetologia. 60:1630–1638. 2017. View Article : Google Scholar : PubMed/NCBI

199 

Espinoza SE, Musi N, Wang CP, Michalek J, Orsak B, Romo T, Powers B, Conde A, Moris M, Bair-Kelps D, et al: Rationale and study design of a randomized clinical trial of metformin to prevent frailty in older adults with prediabetes. J Gerontol A Biol Sci Med Sci. 75:102–109. 2020. View Article : Google Scholar :

200 

Kulkarni AS, Brutsaert EF, Anghel V, Zhang K, Bloomgarden N, Pollak M, Mar JC, Hawkins M, Crandall JP and Barzilai N: Metformin regulates metabolic and nonmetabolic pathways in skeletal muscle and subcutaneous adipose tissues of older adults. Aging Cell. 17:e127232018. View Article : Google Scholar : PubMed/NCBI

201 

Wang C, Chen B, Feng Q, Nie C and Li T: Clinical perspectives and concerns of metformin as an anti-aging drug. Aging Med (Milton). 3:266–275. 2020. View Article : Google Scholar

202 

World's first anti-aging TAME trial gets green-light-Longevity. Technology. https://www.longevity.technology/worlds-first-anti-aging-trial-gets-green-light/. Accessed July 1, 2020

203 

American Diabetes Association: Erratum: Standards of Care in diabetes-2023 abridged for primary care providers. Clin diabetes. 2023.41:4–31. View Article : Google Scholar

Clin Diabetes. 41:3282023. View Article : Google Scholar

204 

Sun R, Feng J and Wang J: Underlying mechanisms and treatment of cellular senescence-induced biological barrier interruption and related diseases. Aging Dis. 15:612–639. 2024. View Article : Google Scholar :

205 

Zhao JY, Sheng XL, Li CJ, Qin T, He RD, Dai GY, Cao Y, Lu HB, Duan CY and Hu JZ: Metformin promotes angiogenesis and functional recovery in aged mice after spinal cord injury by adenosine monophosphate-activated protein kinase/endothelial nitric oxide synthase pathway. Neural Regen Res. 18:1553–1562. 2023. View Article : Google Scholar :

206 

Chen Q, Xie D, Yao Q and Yang L: Effect of metformin on locomotor function recovery in rat spinal cord injury model: A meta-analysis. Oxid Med Cell Longev. 2021:19480032021. View Article : Google Scholar : PubMed/NCBI

207 

Chao-Yang G, Peng C and Hai-Hong Z: Roles of NLRP3 inflammasome in intervertebral disc degeneration. Osteoarthritis Cartilage. 29:793–801. 2021. View Article : Google Scholar : PubMed/NCBI

208 

Vergroesen PP, Kingma I, Emanuel KS, Hoogendoorn RJ, Welting TJ, van Royen BJ, van Dieën JH and Smit TH: Mechanics and biology in intervertebral disc degeneration: a vicious circle. Osteoarthritis Cartilage. 23:1057–1070. 2015. View Article : Google Scholar : PubMed/NCBI

209 

Chen WK, Yu XH, Yang W, Wang C, He WS, Yan YG, Zhang J and Wang WJ: lncRNAs: Novel players in intervertebral disc degeneration and osteoarthritis. Cell Prolif. 50:e123132017. View Article : Google Scholar

210 

Fujiwara A, Lim TH, An HS, Tanaka N, Jeon CH, Andersson GB and Haughton VM: The effect of disc degeneration and facet joint osteoarthritis on the segmental flexibility of the lumbar spine. Spine (Phila Pa 1976). 25:3036–3044. 2000. View Article : Google Scholar

211 

Fine N, Lively S, Séguin CA, Perruccio AV, Kapoor M and Rampersaud R: Intervertebral disc degeneration and osteoarthritis: A common molecular disease spectrum. Nat Rev Rheumatol. 19:136–152. 2023. View Article : Google Scholar : PubMed/NCBI

212 

Lyu FJ, Cui H, Pan H, Mc Cheung K, Cao X, Iatridis JC and Zheng Z: Painful intervertebral disc degeneration and inflammation: from laboratory evidence to clinical interventions. Bone Res. 9:72021. View Article : Google Scholar : PubMed/NCBI

213 

Mojtahedzadeh M, Jafarieh A, Najafi A, Khajavi MR and Khalili N: Comparison of metformin and insulin in the control of hyperglycaemia in non-diabetic critically ill patients. Endokrynol Pol. 63:206–211. 2012.PubMed/NCBI

214 

Farkhondeh T, Amirabadizadeh A, Aramjoo H, Llorens S, Roshanravan B, Saeedi F, Talebi M, Shakibaei M and Samarghandian S: Impact of metformin on cancer biomarkers in non-diabetic cancer patients: A systematic review and meta-analysis of clinical trials. Curr Oncol. 28:1412–1423. 2021. View Article : Google Scholar : PubMed/NCBI

215 

Ko KP, Ma SH, Yang JJ, Hwang Y, Ahn C, Cho YM, Noh DY, Park BJ, Han W and Park SK: Metformin intervention in obese non-diabetic patients with breast cancer: Phase II randomized, double-blind, placebo-controlled trial. Breast Cancer Res Treat. 153:361–370. 2015. View Article : Google Scholar : PubMed/NCBI

216 

Lalau JD, Arnouts P, Sharif A and De Broe ME: Metformin and other antidiabetic agents in renal failure patients. Kidney Int. 87:308–322. 2015. View Article : Google Scholar

217 

Cetin M and Sahin S: Microparticulate and nanoparticulate drug delivery systems for metformin hydrochloride. Drug Deliv. 23:2796–2805. 2016. View Article : Google Scholar

218 

Adikwu MU, Yoshikawa Y and Takada K: Bioadhesive delivery of metformin using prosopis gum with antidiabetic potential. Biol Pharm Bull. 26:662–666. 2003. View Article : Google Scholar : PubMed/NCBI

219 

Varde NK and Pack DW: Microspheres for controlled release drug delivery. Expert Opin Biol Ther. 4:35–51. 2004. View Article : Google Scholar

220 

Alexis F, Pridgen EM, Langer R and Farokhzad OC: Nanoparticle technologies for cancer therapy. Handb Exp Pharmacol. 55–86. 2010. View Article : Google Scholar : PubMed/NCBI

221 

Feng Y, Egan B and Wang J: Genetic factors in intervertebral disc degeneration. Genes Dis. 3:178–185. 2016. View Article : Google Scholar : PubMed/NCBI

222 

Cosamalón-Gan I, Cosamalón-Gan T, Mattos-Piaggio G, Villar-Suárez V, García-Cosamalón J and Vega-Álvarez JA: Inflammation in the intervertebral disc herniation. Neurocirugia (Astur : Engl Ed). 32:21–35. 2021.In English, Spanish. View Article : Google Scholar

Related Articles

Journal Cover

August-2024
Volume 54 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang W, Yang Y, Wang Y, Gao Z, Zhang J, Gao W, Chen Y, Lu Y, Wang H, Zhou L, Zhou L, et al: Metformin prevents the onset and progression of intervertebral disc degeneration: New insights and potential mechanisms (Review). Int J Mol Med 54: 71, 2024
APA
Yang, W., Yang, Y., Wang, Y., Gao, Z., Zhang, J., Gao, W. ... Tao, H. (2024). Metformin prevents the onset and progression of intervertebral disc degeneration: New insights and potential mechanisms (Review). International Journal of Molecular Medicine, 54, 71. https://doi.org/10.3892/ijmm.2024.5395
MLA
Yang, W., Yang, Y., Wang, Y., Gao, Z., Zhang, J., Gao, W., Chen, Y., Lu, Y., Wang, H., Zhou, L., Wang, Y., Li, J., Tao, H."Metformin prevents the onset and progression of intervertebral disc degeneration: New insights and potential mechanisms (Review)". International Journal of Molecular Medicine 54.2 (2024): 71.
Chicago
Yang, W., Yang, Y., Wang, Y., Gao, Z., Zhang, J., Gao, W., Chen, Y., Lu, Y., Wang, H., Zhou, L., Wang, Y., Li, J., Tao, H."Metformin prevents the onset and progression of intervertebral disc degeneration: New insights and potential mechanisms (Review)". International Journal of Molecular Medicine 54, no. 2 (2024): 71. https://doi.org/10.3892/ijmm.2024.5395