Skip to main content

The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration

Abstract

It is widely acknowledged that aging, mitochondrial dysfunction, and cellular phenotypic abnormalities are intricately associated with the degeneration of bone and cartilage. Consequently, gaining a comprehensive understanding of the regulatory patterns governing mitochondrial function and its underlying mechanisms holds promise for mitigating the progression of osteoarthritis, intervertebral disc degeneration, and osteoporosis. Mitochondrial hormesis, referred to as mitohormesis, represents a cellular adaptive stress response mechanism wherein mitochondria restore homeostasis and augment resistance capabilities against stimuli by generating reactive oxygen species (ROS), orchestrating unfolded protein reactions (UPRmt), inducing mitochondrial-derived peptides (MDP), instigating mitochondrial dynamic changes, and activating mitophagy, all prompted by low doses of stressors. The varying nature, intensity, and duration of stimulus sources elicit divergent degrees of mitochondrial stress responses, subsequently activating one or more signaling pathways to initiate mitohormesis. This review focuses specifically on the effector molecules and regulatory networks associated with mitohormesis, while also scrutinizing extant mechanisms of mitochondrial dysfunction contributing to bone and cartilage degeneration through oxidative stress damage. Additionally, it underscores the potential of mechanical stimulation, intermittent dietary restrictions, hypoxic preconditioning, and low-dose toxic compounds to trigger mitohormesis, thereby alleviating bone and cartilage degeneration.

Background

Osteoarthritis (OA), intervertebral disc degeneration (IVDD), and osteoporosis (OP) stand out as significant degenerative diseases affecting bone and cartilage, with cellular senescence serving as the primary pathogenic factor. Extant research underscores that the molecular underpinnings of cellular senescence encompass telomere shortening, mitochondrial dysfunction, oxidative stress, DNA damage, a pervasive inflammatory response, radiation, and aberrant mechanical stimulation [1,2,3,4,5]. Mitochondrial dysfunction, identified as a pivotal contributor to age-related phenotypic anomalies, demonstrates a close association with the onset, progression, and prognosis of degenerative bone and cartilage diseases. OA, IVDD, and OP share analogous pathological features, including disturbances in energy metabolism, mitochondrial dysfunction, oxidative stress, inflammation, and abnormal protein aggregation. Targeted enhancement of mitochondrial function, spanning aspects such as metabolism, respiration, redox regulation, calcium signaling, biogenesis, dynamics, and quality control, has shown feasibility in numerous cellular and animal models. Consequently, the current research focus involves the prospect of alleviating or reversing the advancement of aging-related bone and cartilage degenerative diseases through the restoration or enhancement of mitochondrial quality.

In recent years, mitochondrial hormesis (mitohormesis), recognized as an adaptive protective mechanism of mitochondria, has garnered increasing attention within the biomedical research realm. This phenomenon specifically denotes an inherent adaptive stress response exhibited by mitochondria in the face of low doses of stressors that do not reach the damage threshold. Such a response serves to bolster mitochondrial function and augment stress resistance through the activation of pertinent protective molecules and signal transduction pathways [6, 7]. Mitohormesis engenders a spectrum of adaptive defense mechanisms within cells, encompassing enhancements in antioxidant capacity, fortification of the cellular protective response, augmentation of exogenous biological detoxification, and elevation of cellular metabolic flux [8, 9]. Consequently, this review endeavors to delve into the pathogenesis of mitochondrial dysfunction in aging-related OA, IVDD, and OP. Furthermore, it aims to scrutinize current research concerning the mitigation of bone and cartilage degeneration through the stimulation of mitochondrial hormesis and to delineate existing research gaps, laying the groundwork for future breakthroughs.

Mitochondrial hormesis

Hormesis pertains to beneficial biological responses resulting from exposure to low continuous or moderate intermittent doses of stress, which may prove harmful at higher or chronic levels. When organisms encounter lower stress levels, it confers protection against more severe stress in subsequent exposures. The concept of mitohormesis, a term combining mitochondria and hormesis, was originally evidenced in simpler organisms [8]. Exposure to mild mitochondrial stressors has been shown to extend lifespan and mitigate aging-related diseases. This phenomenon has now been documented in mammals [10].

At present, there is a prevailing consensus that stressors capable of inducing mitohormesis primarily fall into distinct categories, including moderate exercise, dietary restrictions, hypoxic preconditioning, ionizing radiation, and exposure to low-toxic compounds [11,12,13,14,15]. Cells uphold mitochondrial homeostasis through various mechanisms, encompassing reactive oxygen species (ROS) regulation, unfolded protein reaction (UPRmt), mitochondrial-derived peptides (MDP), dynamic alterations in mitochondrial morphology (fission and fusion), and the process of mitophagy (Fig. 1). These intricate processes collectively serve to sustain cell survival and uphold biological efficacy [16,17,18,19,20,21].

Fig. 1
figure 1

Schematic representation of the overall understanding of mitohormesis. ATF4: activating transcription factor 4, ATF5: activating transcription factor 5, CHOP: C/EBP homologous protein, Drp1/2: dynamin-related protein 1/2, FIS1: mitochondrial fission 1, Mfn1/2: mitofusin-1/2, MOTS-c: mitochondrial ORF of the 12 S rRNA Type-C, Nrf2: NF-E2 p45-related factor 2, OPA1: optic atrophy-1, PINK1: PTEN induced putative kinase 1, SHLPs 1–6: small humanin-like peptides 1–6

Oxidative stress

The ROS cascade stands as the principal signaling system for mitohormesis, as illustrated in Fig. 2. Within the mitochondrial oxidative phosphorylation electron transfer chain, electron spillovers (1-5%) give rise to ROS such as superoxide anions (O2•−), hydroxyl radicals (•OH), and hydrogen peroxide (H2O2) [22, 23]. Ionizing radiation predominantly generates ROS, including •OH, through the ionizing decomposition of water in tissues and cells [14]. ROS elicit a mitohormic effect by activating downstream transcription factors, fostering communication between mitochondria and the nucleus. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a pivotal transcription factor responsive to ROS oxidative stress, orchestrates the transcriptional activation of genes involved in antioxidants, antioxidant biosynthesis, and metabolic flux transitions [24]. Under normal nonstress conditions, Nrf2 undergoes ubiquitination by the Keap1-E3 ubiquitin-ligase complex, involving Cullin 3, and subsequent degradation by the 26 S proteasome. Upon the mitohormic impact of ROS, mitochondrial ROS induce Nrf2 phosphorylation by PKCδ, facilitating its translocation into the nucleus [25]. Additionally, ROS prompt the relocation of Mst1/2 from the cytoplasm to mitochondria. Keap1 is translocated to mitochondria and phosphorylated by Mst1/2 at four Ser/Thr residues at its N-terminus, leading to Keap1 dimerization and subsequent release of Nrf2 [26, 27]. Hence, these findings imply that mitochondrial ROS activate Nrf2 in an indirect and kinase-dependent manner. Subsequent to gene induction in the nucleus, Nrf2 undergoes phosphorylation by Fyn, a member of the SRC family of tyrosine kinases, culminating in ubiquitination and degradation of Nrf2 [28]. Nrf2 orchestrates the regulation of a cadre of cytoprotective enzymes, including heme oxygenase-1 (HO-1), NAD(P)H-quinone oxidoreductase (NQO1), thioredoxin, peroxiredoxin, and glutathione peroxidase, via the antioxidant response element (ARE), in response to diverse oxidative stresses [29]. Furthermore, ROS-activated Nrf2 amplifies the expression of nuclear respiratory factor 1/2 (NRF-1/2), PGC-1α, and mitochondrial transcription factor A (TFAM), thereby promoting mitochondrial biogenesis [30]. Evidence indicates direct binding of Nrf2 to the promoter region of PINK1 [31]. The mitohormetic effect on promoting fission appears logical, as fission facilitates the removal of damaged mitochondria through mitophagy and enhances mitochondrial membrane potential [32]. However, ROS may induce mitophagy by disrupting mitochondrial membrane potential, enabling the recruitment of PINK1 to the mitochondrial outer membrane. The membrane-bound PINK1 protein stabilizes and undergoes autophosphorylation, initiating the recruitment of Parkin, a soluble E3 ubiquitin ligase. Parkin ubiquitinates various mitochondrial outer proteins, culminating in subsequent autophagy, phagocytosis, and lysosomal degradation [33]. Recent investigations emphasize the PINK1/Parkin pathway’s involvement solely in stress-activated mitophagy in a mitochondrial potential-dependent manner [34]. Nevertheless, some studies propose that the Keap1-Nrf2 stress response pathway diminishes mitochondrial fission and encourages excessive mitochondrial fusion by degrading the mitochondrial fission protein Drp1 [35]. This dynamic shift results in an over fused mitochondrial network with potential cellular protective effects against oxidative stress. In summary, we posit that slightly elevated ROS establish a positive feedback loop influencing mitochondrial kinetics and mitophagy. ROS can induce mitochondrial dynamic changes by disrupting the membrane potential and regulate PINK1/Parkin-mediated mitophagy activity in an Nrf2-dependent manner, thereby achieving mitochondrial quality control.

Fig. 2
figure 2

Oxidative stress signaling system of mitohormesis. There are synergistic and positive feedback regulation between oxidative stress and mitophagy. AKT: serine/threonine kinase proteins, AMPK: adenosine 5’-monophosphate (AMP)-activated protein kinase, ARE: antioxidant response element, FOXO3: forkhead box class O3a, HSF1: heat shock factor 1, Keap1: Kelch-like ECH-associated protein 1, MAPK: mitogen-activated protein kinase, Mst1/2: mammalian Ste20-like kinases 1/2, NFκB: nuclear factor kappa-B, PGC-1α: proliferator‐activated receptor gamma coactivator 1‐alpha, PKCδ: protein kinase Cδ, SESN2: Sestrin2, sMaf: small Maf

Beyond Nrf2, several other key regulators, including PGC-1α, FOXO3a, HSF, and NFκB, assume crucial roles in ROS-induced mitohormesis. Activation of PGC-1α occurs through AMPK or p38-MAPK phosphorylation [36, 37] and sirtuin 1 (SIRT1)-mediated deacetylation [38], markedly amplifying the transcription of proteins vital for mitochondrial biogenesis and respiration. Noteworthy is the finding that sestrin2, a stress-inducing protein in brown adipose tissue, fosters mitochondrial biogenesis by activating AMPK and subsequently enhancing PGC-1α activity. Sestrin2 also exhibits antioxidant functions when stimulated by ROS at non-toxic levels [39]. FOXO3a, akin to PGC-1α, undergoes activation via AMPK phosphorylation [40] and deacetylation mediated by SIRT1 [41] and SIRT3 [42]. While FOXO3a augments TFAM expression, its predominant role lies in enhancing the expression of proteins endowed with antioxidant and repair functions. Additionally, HSF1 and NFκB are acknowledged as downstream effector molecules in ROS-induced mitohormetic effects [43, 44]. However, investigations in this realm are currently limited, and the involvement of the HSF signal regulated by ROS may be partially linked to the unfolded protein response (UPR), necessitating further exploration.

UPRmt

Mild stimulation-induced protein imbalance triggers adaptive responses within mitochondria, activating distinct mitochondrial cytoplasmic and mitochondrial nuclear stress reactions. These responses play a pivotal role in regulating gene transcription, translation, posttranslational modification, and protein degradation to sustain protein stability, as depicted in Fig. 3. The UPRmt predominantly hinges on the subsequent regulation of three bZIP transcription factors, namely CHOP, ATF4, and ATF5, to facilitate the degradation of unfolded or misfolded proteins by mitochondrial chaperone proteins (Hsp10/Hsp27/Hsp60/mtHsp70) and proteases (ClpP/LONP1), thereby reconstructing mitochondrial protein homeostasis [18, 45,46,47]. Throughout the mitochondrial stress response, the activation of UPRmt predominantly relies on the transcription factors ATF4 and ATF5, which typically accumulate and degrade within mitochondria under physiological conditions. In response to stimulation, ATF5 translocates from mitochondria to the nucleus, thus regulating downstream protein expression [48,49,50]. Current understanding posits that the expression of these three transcription factors necessitates eIF2α phosphorylation [51, 52]. The sensing of stresses involves four kinases—GCN2, HRI, PKR, and PERK—which collectively contribute to the phosphorylation of a specific serine on the eukaryotic translation initiation factor eIF2α [53]. Furthermore, Haynes et al. postulated that the combination of CHOP, ATF4, and eIF2α phosphorylation is adequate to induce ATF5 expression [46]. Notably, c-jun and JNK have been identified as the upstream mechanism in the UPRmt signaling system, responsible for regulating CHOP. Upon JNK activation, the transcription factor c-Jun binds to the activator protein-1 (AP-1) consensus sequence, thereby promoting CHOP expression [54, 55]. It is crucial to highlight that, aside from CHOP, ATF4, and ATF5, UPRmt activation necessitates the involvement of the nuclear receptor ERα when disturbances occur within the mitochondria intermembrane space (IMS) [56]. ERα-mediated signaling pathways are activated through AKT phosphorylation, leading to increased expression of the IMS protease HTRA2. This, in turn, eliminates dislocated or defective IMS proteins and concurrently upregulates the expression of transcription factors NRF1 and Nrf1. NRF1 and Nrf1 play distinct roles in promoting mitochondrial biogenesis and protease expression, respectively [7, 46, 56, 57].

Fig. 3
figure 3

Mechanism of mitochondrial unfolded protein reaction (UPRmt) in mitohormesis. c-Jun: jun proto-oncogene, ClpP: caseinolytic protease P, eIF2α: eukaryotic translation initiation factor 2, Erα: estrogen receptor alpha, FGF21: fibroblast growth factor 21, GCN2: general control nonderepressible 2, GDF15: growth differentiation factor 15, HRI: heme-regulated inhibitor, Hsp: heat shock protein, HTRA2: high temperature requirement protein A2, JNK: c-Jun N-terminal kinase, LONP1: Lon protease 1, NRF1: nuclear respiratory factor1, Nrf1: NF-E2 p45-related factor 1, PERK: PKR‐like endoplasmic reticulum kinase, Perk: PKR‐like endoplasmic reticulum kinase, PKR: protein kinase R

MDP

MDP refer to short bioactive peptides (< 100–150 amino acids) encoded by the short open reading frame of mitochondrial DNA. Notable examples include humanin, MOTS-c, and SHLPs 1–6 [58], which cells express in response to repeated stressor stimuli to activate mitohormesis, as illustrated in Fig. 4. Through the mitochondria-nuclear retrograde signal transduction pathway, mitochondria possess the capability to regulate the expression of nuclear genes. Humanin, among these MDPs, plays a pivotal role in mediating a diverse array of intracellular and extracellular signaling pathways, exerting multiple cellular protective functions. Wang et al. suggested that humanin may induce the degradation of Keap1 and the subsequent release of Nrf2, thereby promoting antioxidant gene expression and facilitating the recovery of mitochondrial function [59]. Humanin induces the phosphorylation of AMPK, contributing to the induction of PGC-1α, NRF1, and TFAM, thereby significantly enhancing mitochondrial biogenesis [60]. Furthermore, Humanin elevates the phosphorylation levels of AKT, ERK1/2, and STAT3, with the involvement of PI3K, MEK, and JAK in the activation of these signaling pathways [61]. This activation, in turn, promotes the expression of antioxidant proteins, resulting in reduced ROS levels and mitigated oxidative stress damage. Humanin also exerts an inhibitory effect on calcium overload, consequently suppressing MAPK (JNK/p38) signaling, and enhancing mitochondrial oxidative phosphorylation and biosynthesis [62]. Notably, Humanin activates autophagy signaling mediated by Hsp90, playing a crucial role in stress resistance [63]. MOTS-c, another mitochondrial polypeptide, assumes a pivotal role in the retrograde signal transduction pathway [64]. In an AMPK-dependent manner, MOTS-c translocates to the nucleus, where it interacts with ARE-regulating stress-responsive transfer factors, including Nrf2, thereby enhancing cellular stress resistance [65, 66]. MOTS-c further contributes to metabolic reprogramming by regulating downstream effectors such as HSF1 and HO-1, ultimately restoring mitochondrial function [58, 67]. MOTS-c inhibits oophorectomy-induced bone loss through AMPK activation [68].

Fig. 4
figure 4

Schematic illustration of mitochondrial derived peptide (MDP) regulation of mitohormesis. ERK1/2: extracellular signal-related kinases 1 and 2, JAK: janus kinase, MEK: mitogen-activated extracellular signal-regulated kinase, MOTS-c: mitochondrial ORF of the twelve S c, PI3K: phosphoinositide 3-kinase, STAT3: signal transducer and activator of transcription 3

In a broader context, mitochondrial-derived peptides encompass not only peptides encoded by mitochondrial DNA but also cytokines encoded by nuclear genes. These cytokines are induced when mitochondria are stimulated by stress, acting as endocrine or paracrine factors. FGF21 and GDF15 stand out as the two pivotal factors in mitohormesis, regulating the adaptive responses of the entire body [69, 70]. The upregulation and secretion of FGF21 have been demonstrated to depend on PI3K/AKT signaling [71]. The autocrine activity of FGF21 induces the upregulation of PGC-1α, enhancing mitochondrial oxidation function by activating AMPK and SIRT1 [72, 73]. GDF15, categorized as a stress response cytokine of the TGF-β superfamily, is regulated by the transcription factor CHOP in the UPRmt pathway. CHOP can directly bind to the promoter of GDF15 and activate its transcription under stress conditions [74]. The mitohormesis effect results from the synergistic action of multiple networks, where signal transduction and positive feedback loops between mitochondria and the nucleus collaboratively enhance the stress resistance of cells.

Mitochondrial dynamic changes and mitophagy

Mitohormesis is contingent upon alterations in mitochondrial dynamics and mitochondrial autophagy. Mitochondrial dynamics, involving fission and fusion processes, plays a crucial role in maintaining mitochondrial function and morphology [75]. Mitochondrial fission relies on the induction of cytoplasmic dynamics-related proteins (Drp1 and Drp2) and FIS1 located in the outer membrane, while mitochondrial fusion is mediated by Mfn1 and Mfn2 in the outer membrane and OPA1 in the inner membrane [76]. The elimination of defective mitochondria is ensured through fission, fusion, and mitophagy, aiming to retain mitochondria with complete mitochondrial DNA copy number, membrane composition, and optimal metabolic capacity [21, 77, 78].

Under stress stimulation, the inner membrane depolarizes, leading to the accumulation of PINK1 in the outer membrane, promoting Parkin recruitment to the mitochondrial surface and inducing autophagy for the clearance of damaged mitochondria [21]. Despite potential oxidative stress-induced mutations in mitochondrial DNA (mtDNA), mtDNA with mutations may still fuse with other mitochondria in the same cell through Mfn2 mediation when the mutation load is mild [19, 79]. Additionally, migrasome-mediated mitochondrial exocytosis plays a crucial role in cases of mild mitochondrial damage, assisting in the progress of mitohormesis. During mitochondrial exocytosis, damaged mitochondria are transported outward by the motor protein kinesin family member 5B (KIF5B) and subsequently attached to cortactin by myosin [20]. Following Drp1-mediated mitochondrial division, dysfunctional mitochondria are packaged as migrasomes and excluded from the cell [80]. Studies by Zhang et al. indicate that moderate mechanical stress stimulation enhances the efficiency of mitochondrial dynamics conversion by upregulating the expression of Mfn1/2, OPA1, and the translocation of Drp1 from the cytoplasm to mitochondria [81]. Similarly, Yao et al. observed an upregulation in the expression of Mfn2, FIS1, and Parkin within 24 h in IL-1β-treated chondrocytes, speculating that short-term IL‐1β stimulation promotes mitochondrial dynamics activity and mitophagy [82]. Mitophagy functions to engulf selectively separated mitochondrial fragments resulting from fusion and fission processes, contributing to the reconstruction of mitochondrial homeostasis.

Therefore, the equilibrium between mitochondrial dynamic changes and mitophagy activity stands as the pivotal determinant of mitochondrial structure and function. In essence, mitohormesis predominantly relies on a convergence of multiple signaling pathways, encompassing ROS, metabolites, UPRmt, mitochondrial-cytoplasmic stress responses, MDP secretion, and mitochondrial structural alterations.

Mitochondrial dysfunction is involved in aging-related bone and cartilage degeneration

Within the intracellular microenvironment, mitochondrial dysfunction stands out as a central mechanism in cellular aging injury. Anomalies in oxidative phosphorylation result in reduced ATP synthesis and stimulate the generation of excessive ROS. Oxidative stress injury is regarded as the paramount pathogenic factor associated with cellular senescence phenotypes observed in OA, IVDD, and OP [83,84,85]. The mechanism of cell damage induced by ROS exhibits a high degree of uniformity, primarily involving two aspects: direct oxidative damage to lipids, proteins, and DNA; and participation in signal cascade amplification as a second messenger [86].

OA is a gradual synovial joint disease characterized by focal articular cartilage destruction and a hypertrophic response in adjacent bone. This process results in subchondral bone remodeling, osteophyte formation, and varying degrees of synovial inflammation, joint capsule thickening, and structural destruction of soft tissues [87]. Predominantly, cartilage degeneration is ascribed to extracellular matrix abnormalities, encompassing the deposition of advanced glycosylation end products, proteoglycan deficiency, and collagen hydrolysis [88]. Aging and mechanical overload are recognized as the most significant risk factors for OA [89, 90]. The aging microenvironment of osteoarthritis extends beyond chondrocytes to include synovial fibroblasts and synovial macrophages [91].

IVDD is characterized by dehydration of the nucleus pulposus, annulus fibrosus tear, and loss of extracellular matrix components such as collagen fibers and proteoglycan [92]. Aging emerges as a major risk factor for IVDD development [93]. The interplay of aging and continuous mechanical stress disrupts intervertebral disc metabolism, resulting in an imbalance between the expression of catabolic factors (e.g., proinflammatory cytokines and matrix metalloproteinases) and anabolic molecules (e.g., growth factors). This imbalance ultimately confuses extracellular matrix homeostasis, promoting intervertebral disc degeneration [94, 95]. The aging microenvironment of intervertebral discs encompasses nucleus pulposus cells, annulus fibrosus cells, and chondrocytes [96].

OP is characterized by decreased bone density, microstructure destruction of bone tissue, increased bone fragility, and susceptibility to fractures. The fundamental pathological mechanism of osteoporosis involves an imbalance in bone remodeling homeostasis between osteoblast bone formation and osteoclast bone resorption [97]. Aging and a lack of mechanical stimulation are closely linked to the occurrence and development of osteoporosis [98,99,100]. The aging microenvironments of osteoporosis encompass bone marrow mesenchymal stem cells (BMSCs), osteoblasts, osteoclasts, and osteocytes [5].

Excessive ROS instigate lipid peroxidation and glycosylation, leading to an augmented production of endogenous active aldehydes and their byproducts (glyoxal, methyl glyoxal, malondialdehyde, and 4-hydroxy-2-nonenal), advanced lipoxidation end products (ALE), and advanced glycation end products (AGE) [101]. Elevated levels of ALE and AGE serve as robust indicators of age-related oxidative damage in cartilage and intervertebral discs, potentially hastening the degeneration process by promoting apoptosis and inhibiting extracellular matrix metabolism [102,103,104,105]. Notably, the levels of carbonyl protein and advanced oxidation protein products (AOPPs) in degenerative intervertebral discs exhibit a significant increase [106]. Proteins within the discs of older mice manifest a higher content of oxidized amino acids compared to those of younger mice [106, 107].Moreover, oxidative modification of intervertebral disc collagen induces collagen crosslinking and aggregation, leading to conformational changes in proteins that result in collagen cleavage. This ultimately impacts the anatomical integrity and biomechanical properties of the intervertebral disc [107,108,109]. ROS-induced chondrocyte apoptosis occurs through the reduction of mtDNA integrity and repair capability due to oxidative damage [110]. MtDNA damage and mutation yield respiratory chain subunits with impaired mitochondrial function, hindering effective chondrocyte differentiation. This, in turn, exacerbates ROS production and promotes apoptosis [111]. Similarly, DNA damage induced by ROS in nucleus pulposus cells and osteoblasts has been observed in age-related disc degeneration and osteoporosis [112, 113].Furthermore, ROS regulate cell signaling through the posttranslational oxidative modification of specific mercaptan groups in proteins containing active cysteine [114]. ROS induce sulfonylation of various chondrocyte proteins, including tyrosine kinase (SRC), thereby promoting the expression of matrix metalloproteinase 13 (MMP-13) [115]. These findings suggest that ROS-induced sulfonylation of chondrocyte proteins may activate signaling pathways contributing to cartilage matrix degradation. Additionally, aging-associated ROS-induced hyperoxidation of cysteine reduces the activity of redox-sensitive proteins, ultimately leading to chondrocyte death [116].

Studies have demonstrated that oxidative stress in the mitochondria of aged chondrocytes differentially inhibits the phosphorylation of AKT mediated by insulin-like growth factor-1 (IGF-1) while activating the MEK-ERK MAPK signaling pathway. This alteration affects chondrocyte metabolism, resulting in a decrease in extracellular matrix gene expression and protein synthesis [117, 118]. ROS can activate various signaling pathways, including the p38-MAPK, ERK, and JNK pathways, triggering nuclear translocation of NFκB and Nrf2. This activation promotes the senescence phenotype in chondrocytes, nucleus pulposus cells, and osteoblasts [119,120,121]. Mitochondrial dysfunction and ROS production further stimulate the release of cytokines involved in the regulation of aging-related secretory phenotypes through mechanisms such as the activation of IL-1, IL-6, and inflammasomes assembled from NOD-like receptor family pyrin domain-containing 3 (NLRP3). These processes exacerbate apoptosis and contribute to the development of osteoarthritis and disc degeneration [122,123,124,125]. It is evident from the above that oxidative stress damage caused by mitochondrial dysfunction in senescent cells establishes a positive feedback loop, intensifying the senescence-related phenotype and promoting the progression of bone and cartilage degeneration.

Activation of mitohormesis to combat degenerative changes in bone and cartilage

Aging-related degenerative diseases affecting bone and cartilage entail prolonged chronic progression, with the diminished vitality and stress resistance of cells in the bone microenvironment representing key pathogenic factors. Illustrated in Fig. 5 are current stimuli with the potential to augment the physiological anti-damage mechanism and impede the progression of these degenerative diseases by activating mitohormesis effects. These stimuli encompass mechanical stimulation, intermittent dietary restriction, hypoxic preconditioning, and exposure to certain toxic compounds.

Fig. 5
figure 5

Activation of mitohormesis to relieve bone and cartilage degeneration. IDT: intermittent dietary restriction, IVD: intervertebral disc degeneration, NP: nucleus pulposus, OA: osteoarthritis, OP: osteoporosis, SM; smooth muscle

Mechanical stimulation

The fundamental characteristics of cells in the bone and cartilage microenvironment include sensitivity and an adaptive response to mechanical stimulation [126]. Exercise serves as an effective stimulus for inducing mitohormesis [11, 127]. Varied intensity, duration, and frequency of exercise prompt diverse mitochondrial changes, encompassing biogenesis, oxidative stress, UPRmt, secretion of MDP, fusion and fission, and mitophagy [128, 129]. Physiological-level mechanical stimulation can sustain normal energy metabolism and intracellular homeostasis. It achieves this by inducing moderate ROS stress and the UPRmt response through mitochondria-dependent pathways, ultimately enhancing cell tolerance [130]. When chondrocytes experience physiological circulatory loads, mitochondria generate beneficial levels of ROS that support metabolism, homeostasis, and matrix synthesis [131, 132].

Research indicates that various forms of exercise, including nontraditional activities like tai chi and yoga, can mitigate degenerative injuries, enhance pain relief, improve limb function, and elevate the quality of life in osteoarthritis patients [133]. Notably, low-intensity activity significantly inhibits the onset and progression of osteoarthritis [134]. For instance, walking can prevent subchondral cyst formation, cartilage degeneration, and inhibit osteoclast activity and osteocyte apoptosis [135]. Moderate-intensity exercise hinders cartilage degeneration, extracellular matrix loss, chondrocyte apoptosis, inflammation, and ROS-mediated positive feedback loops [136]. Furthermore, exercise effectively improves bone mass and strength in senile osteoporosis [137, 138]. Dynamic mechanical loading and sufficiently high strain (> 5 microstrain) are essential for inducing bone remodeling [136, 139]. Recent studies have demonstrated that high-intensity progressive resistance exercise enhances bone mass in older individuals [140,141,142]. Niehoff et al. found that exercise protects the bones of mice deficient in superoxide dismutase (SOD2) from oxidative stress [143]. Similarly, tai chi and other exercises can reduce oxidative stress levels in postmenopausal women with osteopenia [144]. However, research on intervertebral disc degeneration has primarily focused on direct exogenous antioxidant interventions, with limited evidence demonstrating exercise’s regulation of ROS levels to activate mitohormesis in nucleus pulposus cells. In conclusion, the interplay between exercise, oxidative stress, and mitochondrial function regulation is complex in the intervertebral disc microenvironment, with no strict criteria for determining the intensity and duration of motor and mechanical stimuli. Excessive mechanical stimulation can cause stress damage that surpasses mitochondrial adaptive flux, leading to dysfunction. Conversely, moderate exercise stimulation may enhance antioxidant response efficiency by activating mitohormesis.

In conclusion, our contention is that ROS, stemming from mitochondrial dysfunction, escalate with age. Excessive ROS levels induce damage to chondrocytes, rendering them more susceptible to the aging process. Furthermore, an excess accumulation of ROS within the bone marrow niche alters the presentation of cell signaling cascades, impedes the osteogenic differentiation of BMSCs, and amplifies osteoclast activity. Mechanical stimulation has the capacity to directly engage cells in exercise, stimulating the mitohormesis effect. This, in turn, prevents ROS-mediated crosstalk between cells and ultimately manifests a protective effect on bone and cartilage.

Intermittent dietary restriction (IDT)

Limiting calorie intake to 60–70% of normal adult weight maintenance requirements has been demonstrated to extend life by 30–50% and confer nearly optimal health [145]. Ketone bodies, generated through the β-oxidation of fatty acids and released as supplementary fuel during intermittent dietary restriction (IDT), induce oxidative stress that, paradoxically, offers long-term benefits by triggering hormetic responses. Initially, ketone bodies stimulate the mitochondrial production of ROS, activating key regulators of cellular protective mechanisms, including Nrf2, AMPK, SIRT1, PGC-1α, and FOXO3a. This activation enhances antioxidant activity, DNA repair, and autophagy [12, 146].

IDT has exhibited noteworthy health benefits, including a discernible impact on osteoarthritis, manifesting within a brief two-week period [145, 147]. Clinical investigations have discerned that ketogenic diets exert a significant improvement in both knee pain and functional outcomes among patients grappling with type 2 diabetes [148]. In a study conducted by Stephen et al., where the effects of dietary restriction and/or exercise were scrutinized in a cohort of 450 knee osteoarthritis patients, it was observed that the combination of IDT and exercise yielded markedly superior positive effects compared to exercise in isolation [149]. The documented influence of IDT extends to pivotal aspects such as inflammation, insulin resistance, and cardiovascular disease. Furthermore, there is suggestive evidence that IDT may enhance bone health by mitigating recognized risk factors associated with osteoporosis. A study investigating the effects of fasting during Ramadan on bone metabolism concluded that dietary practices characteristic of Muslim festivals may confer benefits for bone turnover [150, 151]. The imperative for well-designed studies to thoroughly elucidate the role of IDT in bone metabolism is underscored.

Currently, numerous published studies have explored the favorable outcomes of IDT in the context of bone and cartilage degenerative diseases. The majority of these studies have predominantly focused on systemic regulation, specifically in terms of reducing obesity and inflammation. However, limited research has investigated the mechanistic aspects of IDT in terms of its impact on cellular mitochondrial metabolism. Our hypothesis posits that the activated mitochondrial hormesis effect serves as the fundamental mechanism underpinning the protective effects of IDT on bone and cartilage. Subsequent studies should aim to delineate the intricacies of cell-specific and species-specific nuances in the personalized design of IDT protocols.

Hypoxic preconditioning

An extreme anoxic oxygen tension of 0.5% leads to heightened oxidative stress and cellular apoptosis [152]. In contrast, mild hypoxia within the range of 0.5–3% O2 prompts the release of mitochondrial ROS that function as signaling messengers, initiating various biological processes and mitohormesis. This includes the activation of the transcription factor hypoxia-inducible factor (HIF), which supports the survival of stem cells in hypoxic conditions [7]. Mesenchymal stem cells (MSCs) cultured under 1% O2 hypoxic conditions for approximately 2 weeks exhibit enhanced proliferation and viability, thereby retarding phenotypic changes such as increased cell size, altered morphology, and the expression of senescence-associated β-galactosidase [153].

Physiologically, the microenvironments of bone and cartilage are naturally exposed to hypoxia, with levels ranging from 1% in cartilage to 1–7% in bone marrow. The activated HIF-1α signal in response to hypoxia enhances glycogen storage, preventing energy deficiency and contributing to cell survival. Pathological disruption of the HIF cascade is implicated in bone and cartilage degenerative diseases [154,155,156]. Hence, even a slight decrease in ambient oxygen concentration or the use of hypoxia simulators activates downstream target genes, initiating adaptive responses to hypoxia. Hypoxia preconditioning activates the mitohormesis effect in bone and cartilage microenvironment cells, enhancing stress resistance and cellular activity. Under 1% O2 hypoxic conditions, mouse chondrocytes showed increased HIF expression, triggering a stable autophagy protection mechanism that prevents mitochondrial dysfunction, apoptosis, and senescence, thereby improving cartilage degradation in OA mice [157]. Chronic intermittent hypoxia preconditioning (simulating 3000 m altitude, 5 h/day for 28 days, PO2 = 108.8 mmHg) demonstrated protective effects against collagen-induced osteoarthritis in rats [158]. In addition, Idrus et al. comprehensively analyzed studies on OA treatment involving the creation of anoxic microenvironments [159]. Current research on osteoporosis targets HIF signal activation or inhibition of HIF protein degradation (using deferoxamine, cobalt chloride, and dimethyloxaloylglycine) to simulate hypoxic conditions for therapeutic effects [160, 161]. Furthermore, MSCs cultured with 1% O2 for 7 days exhibited increased proliferation, migration, and promoted osteogenic differentiation [162]. Periodic hypoxia (3% O2 for 1, 2, or 4 h, 4 days/week) regulates MSC differentiation, improving bone health in aging [163]. A randomized clinical trial found that 24 weeks of normobaric cyclic hypoxic exposure combined with resistance circuit training could potentially yield positive effects on bone in older individuals [164]. Timon et al. discovered that 18 weeks of whole-body vibration training with hypoxic stimuli positively affected bone density in elderly individuals [165]. The combination of moderate mechanical stimulation with intermittent hypoxia treatment demonstrated significant benefits on bone metabolism. The nucleus pulposus, characterized by an anoxic and avascular environment, is protected against intervertebral disc degeneration through chronic periodic hypoxic preconditioning (3000 m altitude, 5 h/day for 28 days, PO2 = 108.8 mmHg) in rats [166]. Do et al. assessed phenotype changes in nucleus pulposus cells at two oxygen tension levels (5% and 20%) up to serial passage 20, finding that hypoxia significantly increased autophagosome numbers, activating autophagic flux and concurrently reducing apoptotic protein expression to maintain nucleus homeostasis [167]. In conclusion, hypoxia proves beneficial in delaying the progression of bone and cartilage degenerative diseases, but a deeper exploration of the underlying mechanisms between hypoxia and mitohormesis is warranted.

Toxic compounds

Toxic compounds do not invariably result in pathological outcomes, as mitochondria can function as an adaptive mechanism, inducing diverse stress responses, particularly when stressors are transient. In fact, exposure to mitochondrial toxins, causing transient mitochondrial stress, can be beneficial by fostering mitochondrial hormesis [168]. Resveratrol, a small-molecule compound, effectively emulates the impacts of dietary restrictions. The chronic intermittent application of resveratrol (10 µM) not only enhances bone formation and mitigates accelerated bone loss but also improves osteogenic differentiation in aged BMSCs by stimulating mitochondrial autonomous gene transcription and facilitating mitochondrial functional recovery [169]. Moreover, rotenone, an inhibitor of the electron transport chain, seems to replicate the effects of hypoxic preconditioning. Low concentrations of rotenone (12.5 nM) may induce mitohormesis by modestly elevating ROS levels, thereby increasing SIRT1 expression and subsequent deacetylation of PGC-1α [170]. Additionally, various compounds, including As2O3, arsenite, N-acetyloxyfenicine, 3-bromopyruvate, N-acetylcysteine, and L-lactate, at low concentrations, have demonstrated the capacity to induce mitohormesis in cells. All these compounds have shown that instantaneous ROS production improves the stress resistance of cells [171,172,173,174,175,176]. However, research on the preventive or therapeutic potential of low-dose toxic compounds that trigger mitohormesis in OA, IVDD, and OP remains limited.

Future perspectives

  • The excitatory impact of ROS on mitochondrial hormesis lacks accurate quantitative studies. Many investigations primarily describe the appropriate quantity, medium, and moderation, providing insufficient guidance for subsequent in-depth examinations of exogenous interventions to activate mitochondrial hormesis.

  • The influence of ROS on stress resistance relies not only on ROS concentration but also on their cellular localization. Notably, increased mitochondrial ROS levels significantly enhance cell longevity and stress resistance, while elevated cytoplasmic ROS levels have the opposite effect.

  • Exploring hypoxia preconditioning as a research direction to enhance cellular anti-stress capabilities holds promise. The low perfusion characteristics of bone and cartilage microenvironments introduce complexity to the relationship between hypoxia preconditioning and the enhancement of mitochondrial function.

  • Dietary restrictions exhibit the potential to decelerate the progression of aging-related diseases. It is imperative to investigate, at the cellular level, the role of nutritional restriction in mitigating bone and cartilage degeneration. However, the impact of dietary restrictions on bone and cartilage health is contentious, with specific dietary restrictions (type, proportion, and duration of food intake) becoming the focal point for future research.

  • The synergistic effects of mechanisms in mitohormesis, signal coupling, and positive feedback loops involving ROS, mitochondrial UPRmt, MDP, mitochondrial dynamics, and mitophagy require further elucidation.

Conclusion

Mitochondrial hormesis, serving as a cellular adaptive mechanism to diverse stressors, has garnered significant attention in unraveling disease pathogenesis. In recent years, scholars have delved into the therapeutic potential of mitochondrial hormesis, particularly in cardiovascular and neurodegenerative diseases. The degenerative changes in bone and cartilage associated with aging are intricately linked to mitochondrial dysfunction. However, prevailing studies predominantly concentrate on the interplay between mitochondrial hormesis induced by skeletal muscle exercise and aging. Notably, there is a dearth of research addressing direct stimulation to instigate mitohormetic effects and restore the functionality of chondrocytes, nucleus pulposus cells, BMSCs, and osteoblasts. In the broader physiological context, bone and muscle exhibit systemic regulation. Age-related muscle loss significantly correlates with the onset and progression of OA, IVDD, and OP. ROS production in skeletal muscle cells, arising as a byproduct of mitochondrial oxidative phosphorylation, predominantly governs mitochondrial hormesis during exercise or muscle contraction. Presently, aerobic exercise stands as a partial remedy for age-related muscle decline, with moderate exercise potentially mitigating the advancement of age-related degenerative bone and cartilage conditions by enhancing muscle quality and function.

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

AGE:

advanced glycation end products

ALE:

advanced lipoxidation end products

AOPPs:

advanced oxidation protein products

ARE:

antioxidant response element

FGF21:

fibroblast growth factor 21

GDF15:

growth differentiation factor 15

HIF:

hypoxia-inducible factor

HO-1:

heme oxygenase-1

IDT:

intermittent dietary restriction

IGF-1:

insulin-like growth factor-1

IVDD:

intervertebral disc degeneration

KIF5B:

motor protein kinesin family member 5B

MDP:

mitochondrial-derived peptides

NLRP3:

NOD-like receptor family pyrin domain-containing 3

NQO1:

NAD(P)H-quinone oxidoreductase

NRF-1/2:

nuclear respiratory factor 1/2

Nrf2:

nuclear factor (erythroid-derived 2)-like 2

OA:

osteoarthritis

OP:

osteoporosis

ROS:

reactive oxygen species

TFAM:

mitochondrial transcription factor A

UPRmt:

mitochondrial unfolded protein reaction

References

  1. McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell. 2017;16(2):210–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Toh WS, Brittberg M, Farr J, Foldager CB, Gomoll AH, Hui JH, et al. Cellular senescence in aging and osteoarthritis. Acta Orthop. 2016;87(sup363):6–14.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Zhang XX, He SH, Liang X, Li W, Li TF, Li DF. Aging, cell senescence, the Pathogenesis and targeted therapies of Osteoarthritis. Front Pharmacol. 2021;12:728100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: eliminating senescent cells and alleviating intervertebral disc degeneration. Front Bioeng Biotechnol. 2022;10:823945.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Fang H, Deng Z, Liu J, Chen S, Deng Z, Li W. The mechanism of bone remodeling after bone aging. Clin Interv Aging. 2022;17:405–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bordon Y. Protect the species with mitohormesis? Nat Rev Immunol. 2021;21(7):407.

    Article  CAS  PubMed  Google Scholar 

  7. Barzegari A, Aaboulhassanzadeh S, Landon R, Gueguen V, Meddahi-Pellé A, Parvizpour S, et al. Mitohormesis and mitochondrial dynamics in the regulation of stem cell fate. J Cell Physiol. 2022;237(9):3435–48.

    Article  CAS  PubMed  Google Scholar 

  8. Yun J, Finkel T, Mitohormesis. Cell Metab. 2014;19(5):757–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ristow M. Unraveling the truth about antioxidants: mitohormesis explains ROS-induced health benefits. Nat Med. 2014;20(7):709–11.

    Article  CAS  PubMed  Google Scholar 

  10. Cox CS, McKay SE, Holmbeck MA, Christian BE, Scortea AC, Tsay AJ, et al. Mitohormesis in mice via sustained basal activation of mitochondrial and antioxidant signaling. Cell Metab. 2018;28(5):776–e865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yoon TK, Lee CH, Kwon O, Kim MS. Exercise, Mitohormesis, and mitochondrial ORF of the 12S rRNA Type-C (MOTS-c). Diabetes Metab J. 2022;46(3):402–13.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kolb H, Kempf K, Röhling M, Lenzen-Schulte M, Schloot NC, Martin S. Ketone bodies: from enemy to friend and guardian angel. BMC Med. 2021;19(1):313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Burtscher J, Mallet RT, Pialoux V, Millet GP, Burtscher M. Adaptive responses to Hypoxia and/or Hyperoxia in humans. Antioxid Redox Signal. 2022.

  14. Pohjoismäki JLO, Goffart S. Adaptive and pathological outcomes of Radiation stress-Induced Redox Signaling. Antioxid Redox Signal. 2022;37(4–6):336–48.

    Article  PubMed  Google Scholar 

  15. Meyer JN, Leung MC, Rooney JP, Sendoel A, Hengartner MO, Kisby GE, et al. Mitochondria as a target of environmental toxicants. Toxicol Sci. 2013;134(1):1–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ristow M, Schmeisser K, Mitohormesis. Promoting Health and Lifespan by increased levels of reactive oxygen species (ROS). Dose Response. 2014;12(2):288–341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Woodhead JST, Merry TL. Mitochondrial-derived peptides and exercise. Biochim Biophys Acta Gen Subj. 2021;1865(12):130011.

    Article  CAS  PubMed  Google Scholar 

  18. Qureshi MA, Haynes CM, Pellegrino MW. The mitochondrial unfolded protein response: signaling from the powerhouse. J Biol Chem. 2017;292(33):13500–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Youle RJ, van der Bliek AM. Mitochondrial fission, fusion, and stress. Volume 337. New York, NY: Science; 2012. pp. 1062–5. 6098.

    Google Scholar 

  20. Nielsen J, Gejl KD, Hey-Mogensen M, Holmberg HC, Suetta C, Krustrup P, et al. Plasticity in mitochondrial cristae density allows metabolic capacity modulation in human skeletal muscle. J Physiol. 2017;595(9):2839–47.

    Article  CAS  PubMed  Google Scholar 

  21. Yoo SM, Jung YK. A Molecular Approach to Mitophagy and mitochondrial dynamics. Mol Cells. 2018;41(1):18–26.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Shadel GS, Horvath TL. Mitochondrial ROS signaling in organismal homeostasis. Cell. 2015;163(3):560–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation responses: epigenetic, metabolic, and signaling impacts. Int J Mol Sci. 2021;22(20).

  24. Mitsuishi Y, Motohashi H, Yamamoto M. The Keap1-Nrf2 system in cancers: stress response and anabolic metabolism. Front Oncol. 2012;2:200.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Zamponi E, Zamponi N, Coskun P, Quassollo G, Lorenzo A, Cannas SA, et al. Nrf2 stabilization prevents critical oxidative damage in Down syndrome cells. Aging Cell. 2018;17(5):e12812.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang P, Geng J, Gao J, Zhao H, Li J, Shi Y, et al. Macrophage achieves self-protection against oxidative stress-induced ageing through the Mst-Nrf2 axis. Nat Commun. 2019;10(1):755.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tsushima M, Liu J, Hirao W, Yamazaki H, Tomita H, Itoh K. Emerging evidence for crosstalk between Nrf2 and mitochondria in physiological homeostasis and in heart disease. Arch Pharm Res. 2020;43(3):286–96.

    Article  CAS  PubMed  Google Scholar 

  28. Jain AK, Jaiswal AK. GSK-3beta acts upstream of fyn kinase in regulation of nuclear export and degradation of NF-E2 related factor 2. J Biol Chem. 2007;282(22):16502–10.

    Article  CAS  PubMed  Google Scholar 

  29. Kasai S, Shimizu S, Tatara Y, Mimura J, Itoh K. Regulation of Nrf2 by mitochondrial reactive oxygen species in Physiology and Pathology. Biomolecules. 2020;10(2).

  30. Piantadosi CA, Carraway MS, Babiker A, Suliman HB. Heme oxygenase-1 regulates cardiac mitochondrial biogenesis via Nrf2-mediated transcriptional control of nuclear respiratory factor-1. Circ Res. 2008;103(11):1232–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Murata H, Takamatsu H, Liu S, Kataoka K, Huh NH, Sakaguchi M. NRF2 regulates PINK1 expression under oxidative stress conditions. PLoS ONE. 2015;10(11):e0142438.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Palmeira CM, Teodoro JS, Amorim JA, Steegborn C, Sinclair DA, Rolo AP. Mitohormesis and metabolic health: the interplay between ROS, cAMP and sirtuins. Free Radic Biol Med. 2019;141:483–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wei H, Liu L, Chen Q. Selective removal of mitochondria via mitophagy: distinct pathways for different mitochondrial stresses. Biochim Biophys Acta. 2015;1853(10 Pt B):2784–90.

    Article  CAS  PubMed  Google Scholar 

  34. McWilliams TG, Prescott AR, Montava-Garriga L, Ball G, Singh F, Barini E, et al. Basal mitophagy occurs independently of PINK1 in mouse tissues of high metabolic demand. Cell Metab. 2018;27(2):439–e495.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sabouny R, Fraunberger E, Geoffrion M, Ng AC, Baird SD, Screaton RA, et al. The Keap1-Nrf2 stress response pathway promotes mitochondrial hyperfusion through degradation of the mitochondrial fission protein Drp1. Antioxid Redox Signal. 2017;27(18):1447–59.

    Article  CAS  PubMed  Google Scholar 

  36. Irrcher I, Ljubicic V, Hood DA. Interactions between ROS and AMP kinase activity in the regulation of PGC-1alpha transcription in skeletal muscle cells. Am J Physiol Cell Physiol. 2009;296(1):C116–23.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang Y, Uguccioni G, Ljubicic V, Irrcher I, Iqbal S, Singh K et al. Multiple signaling pathways regulate contractile activity-mediated PGC-1α gene expression and activity in skeletal muscle cells. Physiol Rep. 2014;2(5).

  38. Nemoto S, Fergusson MM, Finkel T. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem. 2005;280(16):16456–60.

    Article  CAS  PubMed  Google Scholar 

  39. Ro SH, Semple I, Ho A, Park HW, Lee JH. Sestrin2, a Regulator of Thermogenesis and mitohormesis in Brown Adipose tissue. Front Endocrinol (Lausanne). 2015;6:114.

    Article  PubMed  Google Scholar 

  40. Li XN, Song J, Zhang L, LeMaire SA, Hou X, Zhang C, et al. Activation of the AMPK-FOXO3 pathway reduces fatty acid-induced increase in intracellular reactive oxygen species by upregulating thioredoxin. Diabetes. 2009;58(10):2246–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Olmos Y, Sánchez-Gómez FJ, Wild B, García-Quintans N, Cabezudo S, Lamas S, et al. SirT1 regulation of antioxidant genes is dependent on the formation of a FoxO3a/PGC-1α complex. Antioxid Redox Signal. 2013;19(13):1507–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tseng AH, Shieh SS, Wang DL. SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative damage. Free Radic Biol Med. 2013;63:222–34.

    Article  CAS  PubMed  Google Scholar 

  43. Esparza-Moltó PB, Nuevo-Tapioles C, Cuezva JM. Regulation of the H(+)-ATP synthase by IF1: a role in mitohormesis. Cell Mol Life Sci. 2017;74(12):2151–66.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Formentini L, Sánchez-Aragó M, Sánchez-Cenizo L, Cuezva JM. The mitochondrial ATPase inhibitory factor 1 triggers a ROS-mediated retrograde prosurvival and proliferative response. Mol Cell. 2012;45(6):731–42.

    Article  CAS  PubMed  Google Scholar 

  45. Pellegrino MW, Nargund AM, Haynes CM. Signaling the mitochondrial unfolded protein response. Biochim Biophys Acta. 2013;1833(2):410–6.

    Article  CAS  PubMed  Google Scholar 

  46. Shpilka T, Haynes CM. The mitochondrial UPR: mechanisms, physiological functions and implications in ageing. Nat Rev Mol Cell Biol. 2018;19(2):109–20.

    Article  CAS  PubMed  Google Scholar 

  47. Zhao Q, Wang J, Levichkin IV, Stasinopoulos S, Ryan MT, Hoogenraad NJ. A mitochondrial specific stress response in mammalian cells. Embo j. 2002;21(17):4411–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dey S, Baird TD, Zhou D, Palam LR, Spandau DF, Wek RC. Both transcriptional regulation and translational control of ATF4 are central to the integrated stress response. J Biol Chem. 2010;285(43):33165–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Fiorese CJ, Schulz AM, Lin YF, Rosin N, Pellegrino MW, Haynes CM. The transcription factor ATF5 mediates a mammalian mitochondrial UPR. Curr Biol. 2016;26(15):2037–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sears TK, Angelastro JM. The transcription factor ATF5: role in cellular differentiation, stress responses, and cancer. Oncotarget. 2017;8(48):84595–609.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Kim KH, Jeong YT, Oh H, Kim SH, Cho JM, Kim YN, et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat Med. 2013;19(1):83–92.

    Article  CAS  PubMed  Google Scholar 

  52. Yan W, Frank CL, Korth MJ, Sopher BL, Novoa I, Ron D, et al. Control of PERK eIF2alpha kinase activity by the endoplasmic reticulum stress-induced molecular chaperone P58IPK. Proc Natl Acad Sci USA. 2002;99(25):15920–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Costa-Mattioli M, Walter P. The integrated stress response: from mechanism to disease. Sci (New York NY). 2020;368(6489).

  54. Horibe T, Hoogenraad NJ. The chop gene contains an element for the positive regulation of the mitochondrial unfolded protein response. PLoS ONE. 2007;2(9):e835.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Weiss C, Schneider S, Wagner EF, Zhang X, Seto E, Bohmann D. JNK phosphorylation relieves HDAC3-dependent suppression of the transcriptional activity of c-Jun. Embo j. 2003;22(14):3686–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Papa L, Germain D. Estrogen receptor mediates a distinct mitochondrial unfolded protein response. J Cell Sci. 2011;124(Pt 9):1396–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Koizumi S, Hamazaki J, Murata S. Transcriptional regulation of the 26S proteasome by Nrf1. Proc Jpn Acad Ser B Phys Biol Sci. 2018;94(8):325–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yang Y, Gao H, Zhou H, Liu Q, Qi Z, Zhang Y, et al. The role of mitochondria-derived peptides in cardiovascular disease: recent updates. Biomed Pharmacother. 2019;117:109075.

    Article  CAS  PubMed  Google Scholar 

  59. Wang Y, Li N, Zeng Z, Tang L, Zhao S, Zhou F et al. Humanin regulates oxidative stress in the ovaries of polycystic ovary syndrome patients via the Keap1/Nrf2 pathway. Mol Hum Reprod. 2021;27(2).

  60. Qin Q, Jin J, He F, Zheng Y, Li T, Zhang Y, et al. Humanin promotes mitochondrial biogenesis in pancreatic MIN6 β-cells. Biochem Biophys Res Commun. 2018;497(1):292–7.

    Article  CAS  PubMed  Google Scholar 

  61. Kim SJ, Guerrero N, Wassef G, Xiao J, Mehta HH, Cohen P, et al. The mitochondrial-derived peptide humanin activates the ERK1/2, AKT, and STAT3 signaling pathways and has age-dependent signaling differences in the hippocampus. Oncotarget. 2016;7(30):46899–912.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Yang X, Zhang H, Wu J, Yin L, Yan LJ, Zhang C. Humanin attenuates NMDA-Induced Excitotoxicity by inhibiting ROS-dependent JNK/p38 MAPK pathway. Int J Mol Sci. 2018;19(10).

  63. Gong Z, Tasset I, Diaz A, Anguiano J, Tas E, Cui L, et al. Humanin is an endogenous activator of chaperone-mediated autophagy. J Cell Biol. 2018;217(2):635–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Yong CQY, Tang BL. A mitochondrial encoded Messenger at the Nucleus. Cells. 2018;7(8).

  65. Zheng Y, Wei Z, Wang T. MOTS-c: a promising mitochondrial-derived peptide for therapeutic exploitation. Front Endocrinol (Lausanne). 2023;14:1120533.

    Article  PubMed  Google Scholar 

  66. Kim KH, Son JM, Benayoun BA, Lee C. The mitochondrial-encoded peptide MOTS-c translocates to the Nucleus to regulate Nuclear Gene expression in response to metabolic stress. Cell Metab. 2018;28(3):516–e247.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Alves de Souza RW, Gallo D, Lee GR, Katsuyama E, Schaufler A, Weber J, et al. Skeletal muscle heme oxygenase-1 activity regulates aerobic capacity. Cell Rep. 2021;35(3):109018.

    Article  CAS  PubMed  Google Scholar 

  68. Ming W, Lu G, Xin S, Huanyu L, Yinghao J, Xiaoying L, et al. Mitochondria related peptide MOTS-c suppresses ovariectomy-induced bone loss via AMPK activation. Biochem Biophys Res Commun. 2016;476(4):412–9.

    Article  PubMed  Google Scholar 

  69. Kim KH, Kim SH, Min YK, Yang HM, Lee JB, Lee MS. Acute exercise induces FGF21 expression in mice and in healthy humans. PLoS ONE. 2013;8(5):e63517.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Johann K, Kleinert M, Klaus S. The role of GDF15 as a myomitokine. Cells. 2021;10(11).

  71. Izumiya Y, Bina HA, Ouchi N, Akasaki Y, Kharitonenkov A, Walsh K. FGF21 is an akt-regulated myokine. FEBS Lett. 2008;582(27):3805–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Chau MD, Gao J, Yang Q, Wu Z, Gromada J. Fibroblast growth factor 21 regulates energy metabolism by activating the AMPK-SIRT1-PGC-1alpha pathway. Proc Natl Acad Sci USA. 2010;107(28):12553–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fisher FM, Kleiner S, Douris N, Fox EC, Mepani RJ, Verdeguer F, et al. FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 2012;26(3):271–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Li D, Zhang H, Zhong Y. Hepatic GDF15 is regulated by CHOP of the unfolded protein response and alleviates NAFLD progression in obese mice. Biochem Biophys Res Commun. 2018;498(3):388–94.

    Article  CAS  PubMed  Google Scholar 

  75. Giacomello M, Pyakurel A, Glytsou C, Scorrano L. The cell biology of mitochondrial membrane dynamics. Nat Rev Mol Cell Biol. 2020;21(4):204–24.

    Article  CAS  PubMed  Google Scholar 

  76. Tilokani L, Nagashima S, Paupe V, Prudent J. Mitochondrial dynamics: overview of molecular mechanisms. Essays Biochem. 2018;62(3):341–60.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Eisner V, Picard M, Hajnóczky G. Mitochondrial dynamics in adaptive and maladaptive cellular stress responses. Nat Cell Biol. 2018;20(7):755–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Seo AY, Joseph AM, Dutta D, Hwang JC, Aris JP, Leeuwenburgh C. New insights into the role of mitochondria in aging: mitochondrial dynamics and more. J Cell Sci. 2010;123(Pt 15):2533–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Meyer JN, Leuthner TC, Luz AL. Mitochondrial fusion, fission, and mitochondrial toxicity. Toxicology. 2017;391:42–53.

    Article  CAS  PubMed  Google Scholar 

  80. Jiao H, Jiang D, Hu X, Du W, Ji L, Yang Y, et al. Mitocytosis, a migrasome-mediated mitochondrial quality-control process. Cell. 2021;184(11):2896–e91013.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang J, Hao X, Chi R, Qi J, Xu T. Moderate mechanical stress suppresses the IL-1β-induced chondrocyte apoptosis by regulating mitochondrial dynamics. J Cell Physiol. 2021;236(11):7504–15.

    Article  CAS  PubMed  Google Scholar 

  82. Yao X, Zhang J, Jing X, Ye Y, Guo J, Sun K, et al. Fibroblast growth factor 18 exerts anti-osteoarthritic effects through PI3K-AKT signaling and mitochondrial fusion and fission. Pharmacol Res. 2019;139:314–24.

    Article  CAS  PubMed  Google Scholar 

  83. Blanco FJ, Rego I, Ruiz-Romero C. The role of mitochondria in osteoarthritis. Nat Rev Rheumatol. 2011;7(3):161–9.

    Article  CAS  PubMed  Google Scholar 

  84. Wang DK, Zheng HL, Zhou WS, Duan ZW, Jiang SD, Li B, et al. Mitochondrial dysfunction in oxidative stress-mediated intervertebral disc degeneration. Orthop Surg. 2022;14(8):1569–82.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Altindag O, Erel O, Soran N, Celik H, Selek S. Total oxidative/anti-oxidative status and relation to bone mineral density in osteoporosis. Rheumatol Int. 2008;28(4):317–21.

    Article  CAS  PubMed  Google Scholar 

  86. Nathan C, Ding A, SnapShot. Reactive oxygen intermediates (ROI). Cell. 2010;140(6):951–e2.

    Article  PubMed  Google Scholar 

  87. Tong L, Yu H, Huang X, Shen J, Xiao G, Chen L, et al. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022;10(1):60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Lotz M, Loeser RF. Effects of aging on articular cartilage homeostasis. Bone. 2012;51(2):241–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Loeser RF, Collins JA, Diekman BO. Ageing and the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016;12(7):412–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Buckwalter JA, Anderson DD, Brown TD, Tochigi Y, Martin JA. The roles of mechanical stresses in the pathogenesis of Osteoarthritis: implications for treatment of Joint injuries. Cartilage. 2013;4(4):286–94.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Jeon OH, David N, Campisi J, Elisseeff JH. Senescent cells and osteoarthritis: a painful connection. J Clin Invest. 2018;128(4):1229–37.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Raj PP. Intervertebral disc: anatomy-physiology-pathophysiology-treatment. Pain Pract. 2008;8(1):18–44.

    Article  PubMed  Google Scholar 

  93. Wang F, Cai F, Shi R, Wang XH, Wu XT. Aging and age related stresses: a senescence mechanism of intervertebral disc degeneration. Osteoarthritis Cartilage. 2016;24(3):398–408.

    Article  CAS  PubMed  Google Scholar 

  94. Romaniyanto MF, Sigit Prakoeswa CR, Notobroto HB, Tinduh D, Ausrin R, et al. An update of current therapeutic approach for intervertebral disc degeneration: a review article. Ann Med Surg (Lond). 2022;77:103619.

    CAS  PubMed  Google Scholar 

  95. Saberi M, Zhang X, Mobasheri A. Targeting mitochondrial dysfunction with small molecules in intervertebral disc aging and degeneration. Geroscience. 2021;43(2):517–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang Y, Yang B, Wang J, Cheng F, Shi K, Ying L, et al. Cell senescence: a Nonnegligible Cell State under survival stress in Pathology of Intervertebral Disc Degeneration. Oxid Med Cell Longev. 2020;2020:9503562.

    PubMed  PubMed Central  Google Scholar 

  97. Liang B, Burley G, Lin S, Shi YC. Osteoporosis pathogenesis and treatment: existing and emerging avenues. Cell Mol Biol Lett. 2022;27(1):72.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Föger-Samwald U, Kerschan-Schindl K, Butylina M, Pietschmann P. Age related osteoporosis: Targeting Cellular Senescence. Int J Mol Sci. 2022;23(5).

  99. Pignolo RJ, Law SF, Chandra A. Bone aging, Cellular Senescence, and osteoporosis. JBMR Plus. 2021;5(4):e10488.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Zhang L, Zheng YL, Wang R, Wang XQ, Zhang H. Exercise for osteoporosis: a literature review of pathology and mechanism. Front Immunol. 2022;13:1005665.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Vistoli G, De Maddis D, Cipak A, Zarkovic N, Carini M, Aldini G. Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): an overview of their mechanisms of formation. Free Radic Res. 2013;47(Suppl 1):3–27.

    Article  CAS  PubMed  Google Scholar 

  102. Abusarah J, Bentz M, Benabdoune H, Rondon PE, Shi Q, Fernandes JC, et al. An overview of the role of lipid peroxidation-derived 4-hydroxynonenal in osteoarthritis. Inflamm Res. 2017;66(8):637–51.

    Article  CAS  PubMed  Google Scholar 

  103. Jitjumnong M, Chalermkitpanit P, Suantawee T, Dechsupa S, Vajarintarangoon L, Honsawek S. Telomere Shortening and increased oxidative stress in lumbar disc degeneration. Int J Mol Sci. 2022;23(17).

  104. Sivan SS, Tsitron E, Wachtel E, Roughley P, Sakkee N, van der Ham F, et al. Age-related accumulation of pentosidine in aggrecan and collagen from normal and degenerate human intervertebral discs. Biochem J. 2006;399(1):29–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Feng C, Yang M, Lan M, Liu C, Zhang Y, Huang B, et al. ROS: crucial intermediators in the pathogenesis of intervertebral disc degeneration. Oxid Med Cell Longev. 2017;2017:5601593.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Hou G, Lu H, Chen M, Yao H, Zhao H. Oxidative stress participates in age-related changes in rat lumbar intervertebral discs. Arch Gerontol Geriatr. 2014;59(3):665–9.

    Article  CAS  PubMed  Google Scholar 

  107. Scharf B, Clement CC, Yodmuang S, Urbanska AM, Suadicani SO, Aphkhazava D, et al. Age-related carbonylation of fibrocartilage structural proteins drives tissue degenerative modification. Chem Biol. 2013;20(7):922–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Elgawish A, Glomb M, Friedlander M, Monnier VM. Involvement of hydrogen peroxide in collagen cross-linking by high glucose in vitro and in vivo. J Biol Chem. 1996;271(22):12964–71.

    Article  CAS  PubMed  Google Scholar 

  109. Pokharna HK, Phillips FM. Collagen crosslinks in human lumbar intervertebral disc aging. Spine (Phila Pa 1976). 1998;23(15):1645–8.

    Article  CAS  PubMed  Google Scholar 

  110. Grishko VI, Ho R, Wilson GL, Pearsall, AWt. Diminished mitochondrial DNA integrity and repair capacity in OA chondrocytes. Osteoarthritis Cartilage. 2009;17(1):107–13.

    Article  CAS  PubMed  Google Scholar 

  111. Blanco FJ, Valdes AM, Rego-Pérez I. Mitochondrial DNA variation and the pathogenesis of osteoarthritis phenotypes. Nat Rev Rheumatol. 2018;14(6):327–40.

    Article  CAS  PubMed  Google Scholar 

  112. Guo Q, Zhu D, Wang Y, Miao Z, Chen Z, Lin Z, et al. Targeting STING attenuates ROS induced intervertebral disc degeneration. Osteoarthritis Cartilage. 2021;29(8):1213–24.

    Article  CAS  PubMed  Google Scholar 

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

  114. Klomsiri C, Karplus PA, Poole LB. Cysteine-based redox switches in enzymes. Antioxid Redox Signal. 2011;14(6):1065–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Wood ST, Long DL, Reisz JA, Yammani RR, Burke EA, Klomsiri C, et al. Cysteine-mediated Redox Regulation of Cell Signaling in chondrocytes stimulated with fibronectin fragments. Arthritis Rheumatol. 2016;68(1):117–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Collins JA, Wood ST, Nelson KJ, Rowe MA, Carlson CS, Chubinskaya S, et al. Oxidative stress promotes Peroxiredoxin Hyperoxidation and attenuates Pro-survival Signaling in Aging chondrocytes. J Biol Chem. 2016;291(13):6641–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Yin W, Park JI, Loeser RF. Oxidative stress inhibits insulin-like growth factor-I induction of chondrocyte proteoglycan synthesis through differential regulation of phosphatidylinositol 3-Kinase-akt and MEK-ERK MAPK signaling pathways. J Biol Chem. 2009;284(46):31972–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Loeser RF, Gandhi U, Long DL, Yin W, Chubinskaya S. Aging and oxidative stress reduce the response of human articular chondrocytes to insulin-like growth factor 1 and osteogenic protein 1. Arthritis Rheumatol. 2014;66(8):2201–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Minguzzi M, Cetrullo S, D’Adamo S, Silvestri Y, Flamigni F, Borzì RM. Emerging players at the intersection of chondrocyte loss of Maturational Arrest, oxidative stress, senescence and low-Grade inflammation in Osteoarthritis. Oxid Med Cell Longev. 2018;2018:3075293.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Dimozi A, Mavrogonatou E, Sklirou A, 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. 2015;30:89–102. discussion 3.

    Article  CAS  PubMed  Google Scholar 

  121. Domazetovic V, Marcucci G, Iantomasi T, Brandi ML, Vincenzini MT. Oxidative stress in bone remodeling: role of antioxidants. Clin Cases Min Bone Metab. 2017;14(2):209–16.

    Article  Google Scholar 

  122. Portal-Núñez S, Esbrit P, Alcaraz MJ, Largo R. Oxidative stress, autophagy, epigenetic changes and regulation by miRNAs as potential therapeutic targets in osteoarthritis. Biochem Pharmacol. 2016;108:1–10.

    Article  PubMed  Google Scholar 

  123. Lepetsos P, Papavassiliou AG. ROS/oxidative stress signaling in osteoarthritis. Biochim Biophys Acta. 2016;1862(4):576–91.

    Article  CAS  PubMed  Google Scholar 

  124. Tang P, Gu JM, Xie ZA, Gu Y, Jie ZW, Huang KM, et al. Honokiol alleviates the degeneration of intervertebral disc via suppressing the activation of TXNIP-NLRP3 inflammasome signal pathway. Free Radic Biol Med. 2018;120:368–79.

    Article  CAS  PubMed  Google Scholar 

  125. Loeser RF. The role of aging in the development of Osteoarthritis. Trans Am Clin Climatol Assoc. 2017;128:44–54.

    PubMed  PubMed Central  Google Scholar 

  126. He Y, Makarczyk MJ, Lin H. Role of mitochondria in mediating chondrocyte response to mechanical stimuli. Life Sci. 2020;263:118602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Musci RV, Hamilton KL, Linden MA. Exercise-Induced Mitohormesis for the Maintenance of Skeletal Muscle and Healthspan Extension. Sports (Basel). 2019;7(7).

  128. Hood DA, Memme JM, Oliveira AN, Triolo M. Maintenance of Skeletal Muscle Mitochondria in Health, Exercise, and aging. Annu Rev Physiol. 2019;81:19–41.

    Article  CAS  PubMed  Google Scholar 

  129. Lundby C, Jacobs RA. Adaptations of skeletal muscle mitochondria to exercise training. Exp Physiol. 2016;101(1):17–22.

    Article  CAS  PubMed  Google Scholar 

  130. Merry TL, Ristow M. Mitohormesis in exercise training. Free Radic Biol Med. 2016;98:123–30.

    Article  CAS  PubMed  Google Scholar 

  131. Jiang W, Liu H, Wan R, Wu Y, Shi Z, Huang W. Mechanisms linking mitochondrial mechanotransduction and chondrocyte biology in the pathogenesis of osteoarthritis. Ageing Res Rev. 2021;67:101315.

    Article  CAS  PubMed  Google Scholar 

  132. Wolff KJ, Ramakrishnan PS, Brouillette MJ, Journot BJ, McKinley TO, Buckwalter JA, et al. Mechanical stress and ATP synthesis are coupled by mitochondrial oxidants in articular cartilage. J Orthop Res. 2013;31(2):191–6.

    Article  CAS  PubMed  Google Scholar 

  133. Wellsandt E, Golightly Y. Exercise in the management of knee and hip osteoarthritis. Curr Opin Rheumatol. 2018;30(2):151–9.

    Article  PubMed  Google Scholar 

  134. Dunlop DD, Song J, Semanik PA, Sharma L, Bathon JM, Eaton CB, et al. Relation of physical activity time to incident disability in community dwelling adults with or at risk of knee arthritis: prospective cohort study. BMJ. 2014;348:g2472.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Iijima H, Aoyama T, Ito A, Yamaguchi S, Nagai M, Tajino J, et al. Effects of short-term gentle treadmill walking on subchondral bone in a rat model of instability-induced osteoarthritis. Osteoarthritis Cartilage. 2015;23(9):1563–74.

    Article  CAS  PubMed  Google Scholar 

  136. Little-Letsinger SE, Rubin J, Diekman B, Rubin CT, McGrath C, Pagnotti GM, et al. Exercise to mend aged-tissue crosstalk in bone targeting osteoporosis & osteoarthritis. Semin Cell Dev Biol. 2022;123:22–35.

    Article  CAS  PubMed  Google Scholar 

  137. Chen X, Li L, Guo J, Zhang L, Yuan Y, Chen B, et al. Treadmill running exercise prevents senile osteoporosis and upregulates the wnt signaling pathway in SAMP6 mice. Oncotarget. 2016;7(44):71072–86.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Buhl KM, Jacobs CR, Turner RT, Evans GL, Farrell PA, Donahue HJ. Aged bone displays an increased responsiveness to low-intensity resistance exercise. J Appl Physiol (1985). 2001;90(4):1359–64.

    Article  CAS  PubMed  Google Scholar 

  139. Rubin CT, Lanyon LE. Regulation of bone mass by mechanical strain magnitude. Calcif Tissue Int. 1985;37(4):411–7.

    Article  CAS  PubMed  Google Scholar 

  140. Harding AT, Weeks BK, Lambert C, Watson SL, Weis LJ, Beck BR. A comparison of bone-targeted Exercise strategies to reduce fracture risk in Middle-aged and older men with Osteopenia and osteoporosis: LIFTMOR-M semi-randomized controlled trial. J Bone Min Res. 2020;35(8):1404–14.

    Article  Google Scholar 

  141. Lambert C, Beck BR, Harding AT, Watson SL, Weeks BK. Regional changes in indices of bone strength of upper and lower limbs in response to high-intensity impact loading or high-intensity resistance training. Bone. 2020;132:115192.

    Article  PubMed  Google Scholar 

  142. Watson S, Weeks B, Weis L, Harding A, Horan S, Beck B. High-intensity resistance and impact training improves bone Mineral density and physical function in Postmenopausal Women with Osteopenia and osteoporosis: the LIFTMOR Randomized Controlled Trial. J Bone Min Res. 2019;34(3):572.

    Article  Google Scholar 

  143. Baur A, Henkel J, Bloch W, Treiber N, Scharffetter-Kochanek K, Brüggemann GP, et al. Effect of exercise on bone and articular cartilage in heterozygous manganese superoxide dismutase (SOD2) deficient mice. Free Radic Res. 2011;45(5):550–8.

    Article  CAS  PubMed  Google Scholar 

  144. Qian G, Xue K, Tang L, Wang F, Song X, Chyu MC, et al. Mitigation of oxidative damage by green tea polyphenols and Tai Chi exercise in postmenopausal women with osteopenia. PLoS ONE. 2012;7(10):e48090.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Johnson JB, Laub DR, John S. The effect on health of alternate day calorie restriction: eating less and more than needed on alternate days prolongs life. Med Hypotheses. 2006;67(2):209–11.

    Article  PubMed  Google Scholar 

  146. Miller VJ, Villamena FA, Volek JS. Nutritional ketosis and mitohormesis: potential implications for mitochondrial function and Human Health. J Nutr Metab. 2018;2018:5157645.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Radakovich LB, Marolf AJ, Culver LA, Santangelo KS. Calorie restriction with regular chow, but not a high-fat diet, delays onset of spontaneous osteoarthritis in the Hartley guinea pig model. Arthritis Res Ther. 2019;21(1):145.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Lyman KS, Athinarayanan SJ, McKenzie AL, Pearson CL, Adams RN, Hallberg SJ, et al. Continuous care intervention with carbohydrate restriction improves physical function of the knees among patients with type 2 diabetes: a non-randomized study. BMC Musculoskelet Disord. 2022;23(1):297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Messier SP, Legault C, Mihalko S, Miller GD, Loeser RF, DeVita P, et al. The intensive Diet and Exercise for Arthritis (IDEA) trial: design and rationale. BMC Musculoskelet Disord. 2009;10:93.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Veronese N, Reginster JY. The effects of calorie restriction, intermittent fasting and vegetarian diets on bone health. Aging Clin Exp Res. 2019;31(6):753–8.

    Article  PubMed  Google Scholar 

  151. Bahijri SM, Ajabnoor GM, Borai A, Al-Aama JY, Chrousos GP. Effect of Ramadan fasting in Saudi Arabia on serum bone profile and immunoglobulins. Ther Adv Endocrinol Metab. 2015;6(5):223–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Barzegari A, Nouri M, Gueguen V, Saeedi N, Pavon-Djavid G, Omidi Y. Mitochondria-targeted antioxidant mito-TEMPO alleviate oxidative stress induced by antimycin A in human mesenchymal stem cells. J Cell Physiol. 2020;235(7–8):5628–36.

    Article  CAS  PubMed  Google Scholar 

  153. Kim DS, Ko YJ, Lee MW, Park HJ, Park YJ, Kim DI, et al. Effect of low oxygen tension on the biological characteristics of human bone marrow mesenchymal stem cells. Cell Stress Chaperones. 2016;21(6):1089–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Chen W, Wu P, Yu F, Luo G, Qing L, Tang J. HIF-1α regulates bone homeostasis and angiogenesis, participating in the occurrence of bone metabolic diseases. Cells. 2022;11(22).

  155. Wang J, Zhao B, Che J, Shang P. Hypoxia pathway in osteoporosis: Laboratory Data for clinical prospects. Int J Environ Res Public Health. 2023;20(4).

  156. Chen JW, Li B, Yang YH, Jiang SD, Jiang LS. Significance of hypoxia in the physiological function of intervertebral disc cells. Crit Rev Eukaryot Gene Expr. 2014;24(3):193–204.

    Article  CAS  PubMed  Google Scholar 

  157. Hu S, Zhang C, Ni L, Huang C, Chen D, Shi K, et al. Stabilization of HIF-1α alleviates osteoarthritis via enhancing mitophagy. Cell Death Dis. 2020;11(6):481.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Shi M, Cui F, Liu AJ, Ma HJ, Cheng M, Song SX, et al. The protective effects of chronic intermittent hypobaric hypoxia pretreatment against collagen-induced arthritis in rats. J Inflamm (Lond). 2015;12:23.

    Article  PubMed  Google Scholar 

  159. Mohd Yunus MH, Lee Y, Nordin A, Chua KH. Bt Hj Idrus R. Remodeling osteoarthritic articular cartilage under hypoxic conditions. Int J Mol Sci. 2022;23(10).

  160. Zhao Q, Shen X, Zhang W, Zhu G, Qi J, Deng L. Mice with increased angiogenesis and osteogenesis due to conditional activation of HIF pathway in osteoblasts are protected from ovariectomy induced bone loss. Bone. 2012;50(3):763–70.

    Article  CAS  PubMed  Google Scholar 

  161. Liu X, Tu Y, Zhang L, Qi J, Ma T, Deng L. Prolyl hydroxylase inhibitors protect from the bone loss in ovariectomy rats by increasing bone vascularity. Cell Biochem Biophys. 2014;69(1):141–9.

    Article  CAS  PubMed  Google Scholar 

  162. Hung SP, Ho JH, Shih YR, Lo T, Lee OK. Hypoxia promotes proliferation and osteogenic differentiation potentials of human mesenchymal stem cells. J Orthop Res. 2012;30(2):260–6.

    Article  PubMed  Google Scholar 

  163. Camacho-Cardenosa M, Quesada-Gómez JM, Camacho-Cardenosa A, Leal A, Dorado G, Torrecillas-Baena B, et al. Effects of normobaric cyclic hypoxia exposure on mesenchymal stem-cell differentiation-pilot study on bone parameters in elderly. World J Stem Cells. 2020;12(12):1667–90.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Camacho-Cardenosa A, Camacho-Cardenosa M, Martínez-Guardado I, Leal A, Andrada JMV, Timón R. Resistance circuit training combined with hypoxia stimulates bone system of older adults: a randomized trial. Exp Gerontol. 2022;169:111983.

    Article  CAS  PubMed  Google Scholar 

  165. Camacho-Cardenosa M, Camacho-Cardenosa A, Burtscher M, Brazo-Sayavera J, Tomas-Carus P, Olcina G, et al. Effects of whole-body vibration training combined with cyclic hypoxia on bone Mineral Density in Elderly people. Front Physiol. 2019;10:1122.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Liu SR, Ren D, Wu HT, Yao SQ, Song ZH, Geng LD et al. Reparative effects of chronic intermittent hypobaric hypoxia pre–treatment on intervertebral disc degeneration in rats. Mol Med Rep. 2022;25(5).

  167. Kim HJ, Lee HR, Kim H, Do SH. Hypoxia helps maintain Nucleus Pulposus Homeostasis by Balancing Autophagy and apoptosis. Oxid Med Cell Longev. 2020;2020:5915481.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Lee YM, Lee DH. Mitochondrial toxins and healthy Lifestyle Meet at the crossroad of Hormesis. Diabetes Metab J. 2019;43(5):568–77.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Lv YJ, Yang Y, Sui BD, Hu CH, Zhao P, Liao L, et al. Resveratrol counteracts bone loss via mitofilin-mediated osteogenic improvement of mesenchymal stem cells in senescence-accelerated mice. Theranostics. 2018;8(9):2387–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Yuyun X, Jinjun Q, Minfang X, Jing Q, Juan X, Rui M, et al. Effects of low concentrations of Rotenone upon mitohormesis in SH-SY5Y cells. Dose Response. 2013;11(2):270–80.

    Article  PubMed  Google Scholar 

  171. Bironaite D, Westberg JA, Andersson LC, Venalis A. A variety of mild stresses upregulate stanniocalcin-1 (STC-1) and induce mitohormesis in neural crest-derived cells. J Neurol Sci. 2013;329(1–2):38–44.

    Article  CAS  PubMed  Google Scholar 

  172. Schmeisser S, Schmeisser K, Weimer S, Groth M, Priebe S, Fazius E, et al. Mitochondrial hormesis links low-dose arsenite exposure to lifespan extension. Aging Cell. 2013;12(3):508–17.

    Article  CAS  PubMed  Google Scholar 

  173. Matsumura T, Ono M, Osada S, Matsuhisa F, Ochiai M, Hayakawa Y. N-acetyloxfenicine strongly induces mitohormesis in mice as well as in insects. FEBS Lett. 2023;597(2):288–97.

    Article  CAS  PubMed  Google Scholar 

  174. Arya JK, Kumar R, Tripathi SS, Rizvi SI. 3-Bromopyruvate, a caloric restriction mimetic, exerts a mitohormetic effect to provide neuroprotection through activation of autophagy in rats during aging. Biogerontology. 2022;23(5):641–52.

    Article  CAS  PubMed  Google Scholar 

  175. Singh F, Charles AL, Schlagowski AI, Bouitbir J, Bonifacio A, Piquard F, et al. Reductive stress impairs myoblasts mitochondrial function and triggers mitochondrial hormesis. Biochim Biophys Acta. 2015;1853(7):1574–85.

    Article  CAS  PubMed  Google Scholar 

  176. Zelenka J, Dvořák A, Alán L. L-Lactate protects skin fibroblasts against Aging-Associated mitochondrial dysfunction via Mitohormesis. Oxid Med Cell Longev. 2015;2015:351698.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (82272561 and 81974347), the Natural Science Foundation of Sichuan Province (2023NSFSC1544), the Key Project of Sichuan Provincial Department of Science and Technology (2022YFS0050), the China Postdoctoral Science Foundation (2023M732437), the Post-Doctor Research Project, West China Hospital, Sichuan University (2023HXBH035) and the 1.3.5 Program for Disciplines of Excellence, West China Hospital, Sichuan University (ZYJC18002, 2018HXFH040).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection and analysis were performed by DWCL and CQ. The first draft of the manuscript was written by DWCL and edited by SB. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Bin Shen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors approved this version of the manuscript to publish.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Da, W., Chen, Q. & Shen, B. The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration. Biol Res 57, 37 (2024). https://doi.org/10.1186/s40659-024-00494-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40659-024-00494-1

Keywords