Abstract Background Mesenchymal stem cells (MSCs) have potential for treating degenerative and immune diseases, but their clinical efficacy is limited by senescence, characterized by mitochondrial dysfunction, impaired mitophagy, and metabolic imbalance. The goal of this study was to investigate the effects of dimethyloxalylglycine (DMOG), a hypoxia-mimetic agent that stabilizes hypoxia-inducible factor 1 alpha (HIF-1α), on rejuvenating senescent MSCs by enhancing mitochondrial function, mitophagy, and metabolic reprogramming. Methods Two models of MSC senescence were established: oxidative stress-induced senescence using hydrogen peroxide and replicative senescence through serial passaging. Umbilical cord derived MSCs were treated with DMOG for 48 h under normoxic conditions. Mitochondrial function, mitophagy, and metabolism were assessed using assays that measured mitochondrial membrane potential, reactive oxygen species levels, ATP production, and mitophagy. Western blotting and real-time PCR were employed to analyze the expression changes of relevant molecules. RNA sequencing (RNA-seq) was performed to identify key genes and pathways regulated by DMOG. Additionally, to evaluate the therapeutic potential of rejuvenated MSCs, a co-culture system was established, where DMOG-treated senescent MSCs were co-cultured with IL-1β-treated chondrocytes. Results DMOG treatment significantly reduced key senescence markers, including senescence-associated beta-galactosidase, p53, and p21, in both senescence models. DMOG treatment restored mitochondrial morphology and function, improving mitochondrial membrane potential, reducing mitochondrial reactive oxygen species, and enhancing ATP production. DMOG also promoted mitophagy, as evidenced by increased colocalization of mitochondria with lysosomes. RNA-seq analysis revealed that DMOG activated key pathways, including HIF-1 signaling, calcium signaling, and mitophagy-related gene (BNIP3 and BNIP3L). Notably, BNIP3 knockdown greatly abolished DMOG-induced mitophagy and its anti-senescence effects. Furthermore, DMOG treatment improved metabolic flexibility by enhancing both mitochondrial respiration and glycolysis in senescent MSCs. Moreover, DMOG-treated senescent MSCs partially restored their therapeutic efficacy in an osteoarthritis model by improving extracellular matrix regulation in IL-1β-stimulated chondrocytes. Conclusions Short-term DMOG treatment rejuvenates senescent MSCs by enhancing mitochondrial function, promoting mitophagy via HIF-1α/BNIP3, and improving metabolic reprogramming. DMOG-treated MSCs also showed enhanced therapeutic efficacy in co-culture with IL-1β-treated chondrocytes, suggesting its potential to improve MSC-based therapies in regenerative medicine. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-025-04422-2. Keywords: Mesenchymal stem cells (MSCs), Senescence, Hypoxia-Mimetic agent, HIF-1α, BNIP3, Mitophagy Background Over the past two decades, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic tool for treating immune-related and degenerative diseases owing to their regenerative and immunomodulatory properties [[44]1, [45]2]. However, the clinical translation of MSC-based therapies is limited by their susceptibility to senescence, which reduces their proliferative potential, differentiation capacity, and therapeutic efficacy [[46]3, [47]4]. Senescence, induced by aging or prolonged in vitro expansion, is associated with the development of a senescence-associated secretory phenotype (SASP) that compromises the regenerative potential of MSCs and hinders their widespread clinical application [[48]3, [49]5]. Therefore, understanding and mitigating MSCs senescence is essential for maximizing their therapeutic potential. MSCs senescence is driven by multiple mechanisms, including telomere shortening, oxidative stress, and impaired autophagy, with mitochondrial dysfunction emerging as a critical hallmark [[50]4, [51]6]. Aging MSCs accumulate dysfunctional mitochondria characterized by reduced bioenergetic capacity, increased reactive oxygen species (ROS) production, and a shift in metabolic activity [[52]7, [53]8]. These mitochondrial impairments exacerbate oxidative damage and contribute to progressive loss of cellular function. Notably, mitophagy, a selective form of autophagy that eliminates damaged mitochondria, plays a vital role in maintaining mitochondrial quality and preventing senescence [[54]7–[55]9]. Impaired mitophagy has been implicated in the aging process; however, strategies to enhance mitophagy in senescent MSCs remain underexplored. Low oxygen levels in the stem cell niche are essential for preserving MSC function by limiting oxidative stress and maintaining cellular homeostasis [[56]10, [57]11]. Hypoxic conditions activate hypoxia-inducible factor-1α (HIF-1α), a master regulator of cellular responses to low oxygen levels [[58]12]. HIF-1α drives the expression of genes involved in metabolic adaptation, ROS detoxification, and autophagy [[59]13]. Numerous studies have demonstrated that hypoxia delays the onset of senescence and promotes stem cell function; however, culturing cells in hypoxic environments is technically challenging and costly [[60]14–[61]19]. Hypoxia-mimetic agents, such as dimethyloxalylglycine (DMOG), offer a practical alternative by stabilizing HIF-1α under normoxic conditions [[62]20]. Although hypoxia-mimetic agents are known to mimic some effects of physical hypoxia, their specific role in preventing MSCs senescence and modulating mitochondrial function remains unclear. In this study, we developed two models of MSCs senescence, oxidative stress-induced senescence (via hydrogen peroxide) and replicative senescence (via multi-generation expansion), to investigate the effects of short-term (48-hour) treatment with DMOG, a hypoxia-mimetic that stabilizes HIF-1α. We explored how DMOG modulates mitochondrial function, mitophagy, and metabolism to counteract MSCs senescence, highlighting its potential to enhance the effectiveness of MSCs based therapies. Materials and methods Culture and treatment of MSCs Passage 2 (P2) MSCs derived from the umbilical cord were purchased from GLABIOLIUS GROUP (Shenyang, China) and cultured in Mesenchymal Stem Cell Medium (MSCM) supplemented with 5% fetal bovine serum (FBS) (ScienCell, USA). To avoid interference from cytokines, the medium was replaced with α-MEM (Invitrogen) for 48 h prior to the experiments. Flow cytometry was conducted to confirm the expression of MSC surface markers, including CD14, CD34, CD45, CD73, and CD90. To examine the anti-aging effects of the hypoxia-mimetic agent Dimethyloxallyl Glycine (DMOG), MSCs at passage 5 (P5) and passage 15 (P15) were treated with 200 µM DMOG (MedChemExpress, China) for 48 h under standard culture conditions as previously described in our earlier studies [[63]21]. Establishment of MSCs senescence models Young MSC controls were defined as passage 5 (P5) cells with minimal aging characteristics. Oxidative stress-induced senescence was modeled by treating P5 MSCs with 200 µM hydrogen peroxide (H₂O₂) for 24 h. Replicative senescence was achieved by serial passaging. Passage 15 (P15) MSCs were used as the aged group. Both models were used for further analysis. Senescence-Associated β-Galactosidase (SA-β-Gal) staining SA-β-Gal staining was performed using an SA-β-Gal Staining Kit (Beyotime Biotechnology, China) following the manufacturer’s protocol. Cells were seeded into 6-well plates, fixed, and incubated with the staining solution overnight at 37 °C. Senescent cells were identified based on their blue-green coloration under a light microscope. All experiments were biologically replicated at least three times (n = 3). Western blot analysis Total proteins were extracted from MSCs using RIPA lysis buffer (Boster, China), separated via SDS-PAGE, and transferred to PVDF membranes. The primary antibodies used were p53 (1:1000, BM4309, Boster, China), p21 (1:1000, BM3990, Boster, China), HIF-1α (1:1000, BM4083, Boster, China), MFN1 (1:1000, BM4882, Boster, China), MFN2 (1:1000, BM4906, Boster, China), FIS1 (1:1000, A01932-3, Boster, China), BNIP3 (1:1000, BA4304-2, Boster, China), and GAPDH (1:5000, BM1632, Boster, China) as an internal control. After incubation with HRP-conjugated secondary antibodies, protein bands were visualized using an enhanced chemiluminescence system. Real-Time PCR Total RNA was extracted using TOROzol Reagent (TOROIVD, China) and reverse-transcribed into cDNA using a TOROBlue^® qPCR RT Kit. Real-time PCR was performed using TOROGreen^® qPCR Master Mix on an Applied Biosystems™ QuantStudio™ 5 System (Thermo Fisher, USA). Gene expression was normalized to that of GAPDH and the results were calculated using the 2 − ΔΔCt method. Each experiment was conducted in triplicates. Cell viability assay Cell viability was evaluated using the CCK-8 assay and Calcein/Propidium Iodide (PI) staining. For CCK-8, MSCs were seeded at 2,000 cells/well in 96-well plates, treated for 48 h, and incubated with the CCK-8 reagent (Boster, China). The absorbance was measured at 450 nm. For calcein/PI staining, cells were incubated with calcein-AM (2 µM) and PI (4 µM) (Beyotime, China) at 37 °C for 30 min Live (green) and dead (red) cells were visualized using fluorescence microscopy. Apoptosis assay Apoptosis was detected using an Annexin V-FITC/PI Apoptosis Detection Kit (Beyotime, China). After treatment, the cells were collected, stained with Annexin V-FITC and PI, and analyzed on a BD FACSCelesta™ Flow Cytometer. The percentage of apoptotic cells was quantified using the FACSDiva software. Mitochondrial functional assays (1) Membrane Potential: Mitochondrial membrane potential was measured using the JC-1 Assay Kit (MCE, USA). MSCs were incubated with JC-1 solution for 20 min, and fluorescence images were captured using a laser confocal microscope. (2) ROS Measurement: Mitochondrial ROS levels were assessed using MitoSOX Red (Thermo Fisher Scientific, USA). Cells were incubated with 5 µM MitoSOX for 10 min, washed, counterstained with Hoechst 33,342, and analyzed using the CellInsight CX7 Imaging System. (3) ATP Production: ATP levels were measured using an ATP Assay Kit (Beyotime, China). Luminescence intensity was normalized to the protein content and determined using the BCA Assay. Mitochondrial respiration analysis Mitochondrial respiration was analyzed using a Seahorse XFe96 Analyzer (Agilent, USA). MSCs were seeded in XF96 assay plates, and sequential additions of oligomycin, FCCP, antimycin A, and rotenone was performed to assess mitochondrial respiration. Protein normalization was performed after the assay. Mitophagy assay Mitophagy was evaluated by the colocalization of mitochondria and lysosomes using MitoTracker Green and LysoTracker Red (Beyotime, China). MSCs were stained sequentially, counterstained with Hoechst, and imaged using the CellInsight CX7 System. SiRNA transfection To knock down BNIP3 expression, MSCs were transfected with siBNIP3 or siControl using the jetPRIME^® Transfection Reagent (Polyplus, France) in Opti-MEM (Gibco, USA) following the manufacturer’s protocol. After 6 h, the medium was replaced, and the cells were collected 24 h later for analysis. Knockdown efficiency was confirmed using quantitative PCR. The siRNA sequences for BNIP3 (siBNIP3) were as follows: sense strand, 5’-GGAACACGAGCGUCAUGAA(dT)(dT)-3’ and antisense strand, 5’-UUCAUGACGCUCGUGUUCC(dT)(dT)-3’. Chondrocyte isolation and Co-Culture Articular cartilage samples were obtained with ethical approval (PLA General Hospital No. 2024KY0132-KS001). The cartilage was minced, digested with trypsin (15 min) and 0.2% collagenase II (Gibco, USA) overnight, and centrifuged to isolate the chondrocytes. Chondrocytes were seeded on sterilized coverslips in 24-well plates. For osteoarthritis (OA) modeling, 1 nM IL-1β (Genscript, USA) was added for 24 h and, then washed with PBS. MSCs were seeded directly into the upper compartments of Transwell inserts (Merck, USA) for 48 h of co-culture. Immunofluorescence staining Chondrocytes were fixed, permeabilized with 0.1% Triton X-100, blocked with 1% BSA, and incubated with primary antibodies against Col2a1, MMP13, and Aggrecan (1:200, Boster, China) overnight at 4 °C. FITC-conjugated secondary antibodies were applied, and the nuclei were stained with DAPI. The fluorescence images were captured using a NanoZoomer Scanner under standardized exposure conditions. For quantitative analysis, fluorescence intensity was measured using ImageJ software (version 1.53, NIH, USA) with the following parameters: (1) three independent biological replicates were analyzed per condition; (2) five representative fields were captured per replicate; and (3) a minimum of 50 cells were quantified per sample to ensure statistical robustness. All measurements were normalized to background fluorescence and DAPI-stained nuclei counts. Mitochondrial morphology analysis Mitochondrial morphology was analyzed using the Operetta CLS High-Content Imaging System (PerkinElmer, USA) with a 60×water-immersion objective. MitoTracker Red fluorescence was detected at 490 nm excitation and 516 nm emission, with z-stack imaging performed to ensure optimal focus. For each well, five representative regions (top, bottom, left, right, and center) were captured, and approximately 100–120 cells per group were analyzed across 15 regions. Morphological quantification was performed using Harmony 4.9 software (PerkinElmer, USA), assessing key parameters including mitochondrial area, perimeter, aspect ratio, and form factor. A fluorescence intensity threshold was applied to exclude background noise, and automated algorithms quantified fragmentation and elongation. This standardized, high-throughput approach ensured objective and reproducible analysis of mitochondria. Transmission Electron Microscopy (TEM) assay The adherent UC-MSC in the petri dishes from different experimental groups were carefully scraped using a cell scraper and collected by centrifugation at 1000 g for 5 min. The cell pellets were fixed overnight at 4 °C in 2.5% glutaraldehyde solution (Cat No. P1126, Solarbio, China). After fixation, the samples were post-fixed with 1% osmium tetroxide in phosphate buffer at room temperature for 1 h to enhance structural contrast. The fixed cell samples were then embedded in epoxy resin and polymerized at 60 °C for 48 h. Thin Sects. (50–100 nm) of the resin blocks were cut and stained with 2% uranyl acetate for 10 min, followed by staining with lead citrate for 5 min to further enhance contrast. Mitochondrial ultrastructure was examined using a transmission electron microscope (JEOL JEM-1400, JEOL Ltd., Japan) operated at 80 kV. The images were analyzed for mitochondrial morphology and structural integrity using ImageJ software (NIH, USA). RNA-seq analysis UC-MSCs from the young generation (P5), replicative senescence model (P24), and DMOG treatment group were cultured in 10 cm dishes for 48 h, and at least 1 × 10^6 cells each group were harvested for RNA extraction using TRIzol reagent. Each group has three biological replicates. Total RNA was purified via phenol-chloroform extraction, with sample purity assessed using a NanoPhotometer^® spectrophotometer (IMPLEN, USA). RNA integrity and concentration were determined using an Agilent 2100 RNA Nano 6000 Assay Kit (Agilent Technologies, USA). Sequencing libraries were prepared using the VAHTS Universal V6 RNA-seq Library Prep Kit for Illumina^® (NR604-01/02), incorporating unique index tags for multiplexing. High-throughput sequencing was performed on an Illumina NovaSeq 6000 platform (Illumina, USA). For bioinformatics analysis, clean reads were aligned to the reference genome using HISAT2 (v2.1.0) with Bowtie2 (v1.0.1)-generated indexes. Differentially expressed genes were identified based on|log2(fold change)| and adjusted P-value thresholds. RNA-seq and computational analysis were conducted by easyresearch (Beijing, China). Statistical analysis Statistical analyses were performed using GraphPad Prism version 8.0. Student’s t-test was used for comparisons between two groups, whereas one-way analysis of variance (ANOVA) or two-way ANOVA was applied for multiple-group comparisons. Data are presented as mean ± SD, and statistical significance was set at p < 0.05. Results Short-term DMOG treatment alleviates oxidative stress- and replication-induced MSCs senescence by activating HIF-1α and reduce apoptosis A replicative senescence model was established by serially passaging the human umbilical cord derived MSCs. No significant differences in the surface markers CD73 and CD90 were observed between P5 and P15 MSCs (Fig. [64]S1). In addition, the effect of different concentrations of hydrogen peroxide (H₂O₂) on cell viability was evaluated using the CCK-8 assay (Fig. [65]S2). Based on these results, we used 200 µM H₂O₂ to induce oxidative stress-mediated premature senescence in MSCs. SA-β-gal staining, a hallmark of cellular senescence, revealed a significant increase in the proportion of SA-β-gal-positive cells in H₂O₂-treated MSCs, which was markedly reduced after short-term DMOG treatment (Fig. [66]1A, B). Quantitative PCR analysis demonstrated that DMOG significantly reduced the p53 and p21 mRNA levels (Fig. [67]S3). Western blot analysis also demonstrated that H₂O₂ treatment significantly upregulated p53 and p21 protein levels, while DMOG treatment effectively decreased expression of P53 (Fig. [68]1C). Similarly, in the replicative senescence model, DMOG treatment significantly decreased the proportion of SA-β-gal-positive cells, with a more pronounced reduction observed in the P15 cells (Fig. [69]1D, E). Western blot analysis revealed that DMOG treatment effectively suppressed p53 and p21 expression, particularly in P15 cells (Fig. [70]1F). Furthermore, we assessed the expression levels of senescence-associated genes, including IL6, CXCL1, and MMP3, in both senescence models. These genes were markedly upregulated in senescent MSCs, whereas DMOG treatment significantly downregulated their expression (Fig. [71]1G). Because DMOG is a hypoxia mimetic that stabilizes HIF-1α, our Western blot analysis showed that short-term DMOG treatment significantly increased HIF-1α protein levels in both P5 and P15 cells, with a more pronounced effect under H₂O₂-induced oxidative stress conditions. Protein quantification and RNA analysis further confirmed that DMOG increased HIF-1α protein and mRNA levels (Fig. [72]1H). These results suggest that short-term DMOG treatment alleviates oxidative stress- and replication-induced senescence and activates HIF-1α in MSCs. Fig. 1. [73]Fig. 1 [74]Open in a new tab Short-term DMOG treatment reduces MSC senescence by activating HIF-1α and decreasing apoptosis. (A, B) Representative images of SA-β-gal staining showing the proportion of senescent cells in H₂O₂-treated MSCs before and after DMOG treatment. Scale bar = 500 μm. (C) Western blot analysis of p53 and p21 protein expression during oxidative stress-induced senescence. (D, E) SA-β-gal staining in replicative senescence (P15) MSCs, demonstrating the effect of DMOG in reducing senescence markers. Scale bar = 500 μm. (F) Western blot analysis of p53 and p21 protein levels in P5 (young) and P15 (senescent) MSCs. (G) Expression levels of senescence-associated genes (IL6, CXCL1, and MMP3) in both senescence models as assessed by qRT-PCR. (H) Western blotting and qPCR analysis showing increased HIF-1α protein and mRNA levels in both senescence models after DMOG treatment. (I) Calcein/PI live-dead staining revealed an increase in the proportion of live cells following DMOG treatment in both senescence models. (J, K) Flow cytometric analysis of apoptosis. Error bars represent the mean ± SD of three independent experiments. Statistical significance was set as p < 0.05. Full-length blots are presented in Supplementary Materials - WB Raw Data We further investigated the effects of DMOG on the apoptosis of senescent MSCs. Calcein/PI live-dead staining revealed an increase in the proportion of dead cells in both senescence models, while short-term DMOG treatment markedly increased the proportion of live cells (Fig. [75]1I and Fig. [76]S4). Flow cytometry analysis indicated that apoptosis levels were significantly elevated in both senescence models compared to the controls, whereas DMOG treatment significantly reduced the proportion of apoptotic cells (Fig. [77]1J, K). These findings demonstrate that short-term DMOG treatment effectively counteracted apoptosis in senescent MSCs. Short-term DMOG treatment significantly improved the mitochondrial morphology and reduced damage in aged MSCs To elucidate the role of mitochondria in DMOG-mediated rejuvenation of aged MSCs, we investigated the impact of DMOG treatment on the regulation of mitochondrial morphology and function in these cells. In this study, mitochondria were labeled with MitoTracker Green and analyzed using fluorescence microscopy and transmission electron microscopy (TEM) to assess the effects of various treatments on mitochondrial structure and function. Fluorescence microscopy revealed that mitochondria in both models of senescent MSCs exhibited a reduction in number, an increase in size, and morphological changes characterized by shortened length and increased diameter (Fig. [78]2A). Following DMOG treatment, mitochondrial length, diameter, and circularity were significantly improved in senescent MSCs, with a notable increase in mitochondrial density and reduction in the area of individual mitochondria (Fig. [79]2A-C). TEM analysis of the mitochondrial ultrastructure showed that senescent MSCs had larger mitochondrial volumes, fewer mitochondria, and reduced inner membrane surface area. DMOG treatment effectively reversed these changes, leading to a decrease in mitochondrial damage and the restoration of mitochondrial structural integrity (Fig. [80]2D, E). Our results demonstrated that aging induced alterations in the mitochondrial morphology of MSCs, whereas treatment with DMOG effectively reversed mitochondrial damage and morphological changes in aged MSCs. Fig. 2. [81]Fig. 2 [82]Open in a new tab Short-term DMOG treatment restores mitochondrial morphology and reduces damage in aged MSCs. (A) Representative fluorescence microscopy images of mitochondria labeled with Mitotracker Green in senescent MSCs (induced by oxidative stress or replicative aging) after DMOG treatment. The second row shows the mitochondrial morphology after software processing. Scale bar = 20 μm. The locally enlarged portion of this is a typical region that reflects the mitochondrial morphology of the group. (B, C) Quantitative analysis of mitochondrial morphological parameters (length, diameter, circularity, and density) s. (D) Transmission electron microscopy (TEM) images of the mitochondrial ultrastructure in aged MSCs. (Red box: mitochondrial autophagosomes; yellow arrow: dividing mitochondria; blue arrow: damaged and swollen mitochondria). (E) Quantitative TEM analysis showing reduced mitochondrial damage and restored structural integrity after DMOG treatment. Error bars represent the mean ± SD from three independent experiments. Statistical significance was set at p < 0.05 Short-term DMOG treatment enhances mitochondrial function and mitophagy in aged MSCs Previous studies have shown that HIF-1α promotes mitochondrial fission and regulate functions such as mitophagy [[83]22]. Here, we examined the effects of DMOG on mitochondrial function in aged MSCs, including mitochondrial membrane potential, ROS levels, mitophagy, ATP production, and the expression of fusion and fission-related genes. JC-1 fluorescence images revealed that, compared to the P5 group, both the H₂O₂-treated group and P15-aged MSCs exhibited significantly reduced mitochondrial membrane potential, whereas DMOG treatment significantly restored mitochondrial membrane potential in both MSCs senescence models (Fig. [84]3A). MitoSox Red staining demonstrated increased mitochondrial ROS generation in H₂O₂-treated and P15-aged MSCs, which was markedly reduced by DMOG treatment (Fig. [85]3B). Quantitative analysis further confirmed that DMOG treatment restored the mitochondrial membrane potential and reduced mitochondrial ROS production in aged MSCs (Fig. [86]3C, D). Western blot analysis revealed the expression levels of the mitochondrial fusion-related proteins MFN1 and MFN2, along with the mitochondrial fission-related protein Fis1. Notably, DMOG treatment led to significant upregulation of Fis1 expression, with the effect being particularly pronounced in replicative senescent P15 cells (Fig. [87]3E). Co-staining with LysTtracker Red and MitoTracker Green revealed a significant reduction in the number of mitochondrial autophagosomes in aged MSCs, whereas DMOG treatment notably enhanced the co-localization of mitochondria with lysosomes and increased the level of mitophagy in aged MSCs (Fig. [88]3F, G). Since the primary function of the mitochondria is ATP production, we observed a marked reduction in ATP levels in aging cells. However, DMOG treatment significantly restored ATP production in both MSC senescence models (Fig. [89]3H). Collectively, these findings demonstrate that DMOG restores mitochondrial function in aged MSCs by improving the membrane potential, reducing ROS levels, enhancing mitophagy, and boosting ATP production. Fig. 3. [90]Fig. 3 [91]Open in a new tab DMOG treatment ameliorates mitochondrial dysfunction and enhances mitophagy and ATP production in aged MSCs. (A) Representative JC-1 fluorescence images showing mitochondrial membrane potential in different groups: CCCP (positive control for mitochondrial depolarization), P5 MSCs, P5 + H₂O₂-treated MSCs, P5 + H₂O₂+DMOG-treated MSCs, P15 MSCs, and P15 + DMOG-treated MSCs. JC-1 red fluorescence indicates high mitochondrial membrane potential, whereas green fluorescence indicates depolarized mitochondria. Scale bar = 100 μm. (B) Representative MitoSox Red fluorescence images showing mitochondrial ROS levels in different treatment groups. Scale bar = 10 μm. (C, D) Quantitative analysis of the mitochondrial membrane potential (C) and mitochondrial ROS levels (D). (E) Western blot analysis showing the expression levels of mitophagy-related proteins (MFN1, MFN2, and Fis1) in P5 and P15 MSCs with or without H₂O₂ and DMOG treatment. GAPDH was used as a loading control. (F) Representative confocal images of co-staining with LysoTracker Red (lysosomes) and MitoTracker Green (mitochondria) in MSCs under different conditions. Scale bar = 10 μm. (G) Quantitative analysis of the number of mitophagosomes per cell in the different groups. (H) Quantitative analysis of ATP production per cell in the different groups. Data are expressed as the mean ± SEM (n = 3). *p < 0.05, **p < 0.01. ***p < 0.001. Full-length blots are presented in Supplementary Materials - WB Raw Data Short-term DMOG treatment partially restores mitochondrial respiration and glycolytic function in senescent MSCs We further investigated the impact of DMOG on the metabolic reprogramming of senescent MSCs. Mitochondrial respiration and glycolytic function in MSCs under various treatment conditions were assessed using oxygen consumption rate (OCR) and extracellular acidification rate (ECAR). The OCR results demonstrated that MSCs in the P5 group exhibited significantly higher baseline mitochondrial respiration, ATP production, and maximal respiratory capacity than those in the P15 group (Fig. [92]4A, B). Similarly, MSCs in the H₂O₂-induced premature senescence group showed markedly lower baseline mitochondrial respiration, ATP production, and maximal respiratory capacity than those in the P5 group (Fig. [93]4A, B). Notably, short-term DMOG treatment significantly enhanced baseline mitochondrial respiration and ATP production in the H₂O₂-induced premature senescence group, while it had no significant impact on the P15 replicative senescence group (Fig. [94]4A, B). In terms of glycolytic function, the P5 group showed substantially higher glycolysis, glycolytic capacity, and glycolytic reserve than both the P15 group and the H₂O₂-induced premature senescence group (Fig. [95]4C, D). This indicates that aged MSCs exhibit diminished glycolytic function, including significant reductions in basal glycolysis, glycolytic capacity, and glycolytic reserves. After DMOG treatment, a notable improvement in glycolysis and glycolytic capacity was observed in the H₂O₂-induced premature senescence group. The P15 replicative senescence group also experienced significant increases in glycolysis and glycolytic capacity, although these were still markedly lower than those in P5 cells under all treatment conditions (Fig. [96]4C, D). Our results indicated that MSCs in the P5 group exhibited superior metabolic activity, whereas aged MSCs (P15 and H₂O₂-treated P5 group) displayed significant impairments in both mitochondrial oxidative phosphorylation and glycolytic functions. Notably, short-term DMOG treatment partially restored the OCR and ECAR in senescent MSCs. Fig. 4. [97]Fig. 4 [98]Open in a new tab DMOG treatment improves mitochondrial respiration and glycolytic function in senescent MSCs. (A) Oxygen consumption rate (OCR) profiles representing mitochondrial respiration in MSCs under various conditions. (B) Quantitative analysis of mitochondrial respiration parameters, including baseline respiration, ATP production, and maximal respiratory capacity of MSCs. (C) Extracellular acidification rate (ECAR) profiles representing the glycolytic function in MSCs under various conditions. (D) Quantitative analysis of glycolytic parameters, including glycolysis, glycolytic capacity, and glycolytic reserves. Data are expressed as the mean ± SEM (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001 Transcriptomic analysis reveals key genes and signaling pathways involved in the rejuvenation of aged MSCs by short-term DMOG treatment To gain deeper insights into the molecular mechanisms underlying the rejuvenation of aged MSCs by short-term DMOG treatment, we cultured P5 MSCs through successive passages until P24, followed by DMOG treatment. RNA sequencing and subsequent bioinformatic analyses were conducted to investigate the transcriptional changes induced by DMOG. Volcano plots (Fig. [99]5A, C) show the gene expression profiles of P5 and P24 MSCs following DMOG treatment. Notably, the number of differentially expressed genes regulated by DMOG in P24-aged MSCs was significantly lower than that in P5 MSCs. Furthermore, key mitophagy-related genes, such as BNIP3 and BNIP3L, were consistently upregulated in P5 and P24 MSCs after DMOG treatment. KEGG pathway analysis (Fig. [100]5B, D) revealed that in P5 young MSCs, DMOG primarily activated the calcium, HIF-1, MAPK glycolysis/gluconeogenesis, and mitophagy-related pathways. In contrast, in P24-aged MSCs, DMOG predominantly activated the HIF-1 signaling, glycolysis/gluconeogenesis, fructose and mannose metabolism, and mitophagy-related pathways. Heatmaps and KEGG analysis of the four groups (Fig. [101]5E, F) further demonstrated that differentially expressed genes were enriched in the calcium, HIF-1, MAPK, and mitophagy-related pathways. The heatmap (Fig. [102]5G) highlights the expression levels of key mitophagy signaling genes, including HIF-1α, ATG9A, BNIP3, and BNIP3L, following DMOG treatment. A Venn diagram (Fig. [103]5H) highlights both shared and group-specific DEGs, revealing differences in gene expression between young and aged MSCs in response to DMOG treatment. GO analysis of the 79 genes co-regulated by DMOG in both P5 and P24 MSCs showed significant enrichment in processes such as cellular response to hypoxia, metabolic reprogramming, and mitophagy-related pathways (Fig. [104]5I-K). Pathway analysis confirmed their involvement in HIF-1 signaling, glycolysis/gluconeogenesis, and arginine and proline metabolism. GO analysis further emphasized terms, such as cellular response to decreased oxygen levels, peptidyl-proline hydroxylation, and metabolic processes. These findings suggest that DMOG rejuvenates aged MSCs by activating hypoxia- and mitophagy-related genes such as HIF-1α, BNIP3, and BNIP3L, while enhancing metabolic reprogramming and adaptive stress responses. Fig. 5. [105]Fig. 5 [106]Open in a new tab DMOG treatment activates hypoxia- and mitophagy-related pathways in aged MSCs. (A, C) Volcano plots of differentially expressed genes (DEGs) in P5 (A) and P24 (C) MSCs after DMOG treatment. Key mitophagy-related genes, BNIP3 and BNIP3L, were consistently upregulated in both groups. (B, D) KEGG pathway enrichment analysis in P5 (B) and P24 (D) MSCs showing that DMOG activated HIF-1 signaling, glycolysis/gluconeogenesis, and mitophagy-related pathways in both young and aged MSCs. (E) Heatmap of DEGs in the P5, P5 + DMOG, P24, and P24 + DMOG groups. Each group has three biological replicates. (F) KEGG pathway analysis of all groups highlights the key pathways activated by DMOG, including HIF-1 signaling and glycolysis. (G) Heatmap showing the expression levels of key mitophagy-related genes (e.g., BNIP3, BNIP3L, HIF-1 A) upregulated by DMOG treatment. (H) Venn diagram illustrating the shared and unique DEGs across comparisons. A total of 79 genes were co-regulated by DMOG in both P5 and P24 MSCs. (I-K) GO and pathway analyses of co-regulated genes revealed enrichment in hypoxia response, glycolysis, and other metabolic processes BNIP3-mediated mitophagy is essential for the rejuvenation effects of DMOG on senescent MSCs RNA-seq analysis revealed that DMOG significantly upregulated the expression of mitophagy-related genes, BNIP3 and BNIP3L, which is essential for the rejuvenating effects of DMOG on senescent MSCs. Western blotting results confirmed that the expression of BNIP3 and BNIP3L was markedly downregulated in both H₂O₂-induced premature senescence and replicative senescence models, whereas DMOG treatment effectively restored their expression in senescent MSCs (Fig. [107]6A, B). Previous studies have identified BNIP3 as a direct target of HIF-1α. Given that BNIP3 is a known direct target of HIF-1α, we investigated whether BNIP3-mediated mitophagy was critical for the rejuvenating effects of DMOG on senescent MSCs. To this end, BNIP3 was knocked down in a P15 replicative senescence model using BNIP3-specific siRNA. Western blot analysis demonstrated that DMOG significantly increased BNIP3 expression in senescent MSCs; however, this upregulation was notably suppressed upon BNIP3 knockdown (Fig. [108]6C, D). Consistently, real-time PCR results corroborated that BNIP3 knockdown abolished DMOG-induced upregulation of BNIP3 (Fig. [109]6E). Functional assessments using SA-β-gal staining revealed that BNIP3 knockdown increased the proportion of senescent cells in P15 BMSCs, whereas DMOG treatment significantly reduced the percentage of SA-β-gal-positive cells. The anti-senescence effect of DMOG was markedly attenuated by BNIP3 knockdown (Fig. [110]6F, G). Furthermore, colocalization analyses using LysoTracker Red and MitoTracker Green staining indicated that DMOG significantly enhanced the interaction between mitochondria and lysosomes, reflecting increased mitophagy, an effect that was substantially suppressed by BNIP3 knockdown (Fig. [111]6H, I). Collectively, these findings highlight the pivotal role of BNIP3-mediated mitophagy in the rejuvenating effects of DMOG on senescent MSCs, providing novel insights into the underlying mechanisms. Fig. 6. [112]Fig. 6 [113]Open in a new tab DMOG rejuvenates senescent MSCs through BNIP3-dependent mitophagy. (A, B) Western blot analysis (A) and quantification (B) showing the expression of mitophagy-related proteins BNIP3 and BNIP3L in P5, P15, and H₂O₂-treated P5 MSCs with or without DMOG treatment. (C, D) Western blot analysis (C) and quantification (D) of BNIP3 expression in P15 MSCs after BNIP3 knockdown with siRNA. (E) Real-time PCR analysis of BNIP3 expression in P15 MSCs with or without DMOG treatment and BNIP3 knockdown. (F, G) SA-β-gal staining (F) and quantification (G) showing the percentage of senescent cells in P15 MSCs. Scale bar = 500 μm. (H, I) Confocal images (H) and quantification (I) of colocalization between mitochondria (Mitotracker Green) and lysosomes (Lysotracker Red) in P15 MSCs. Scale bar = 10 μm. Data are expressed as mean ± SEM (n = 3). **p < 0.01, ***p < 0.001. Full-length blots are presented in Supplementary Materials - WB Raw Data DMOG restores the therapeutic potential of senescent MSCs to alleviate metabolic dysfunction in OA chondrocytes To evaluate whether DMOG treatment could restore the therapeutic potential of senescent MSCs for osteoarthritis (OA), we established an in vitro co-culture system to investigate the effects of DMOG on MSCs in modulating the expression of cartilage extracellular matrix (ECM)-related genes and proteins under IL-1β stimulation (Fig. [114]7A). OA-like conditions were induced in isolated human articular chondrocytes through IL-1β treatment, and these cells were then co-cultured with MSCs from different experimental groups using a Transwell system. Immunofluorescence staining and quantitative analysis revealed a substantial reduction in the expression of key ECM molecules, including Collagen II (Col2a1) and aggrecan (AGC), in IL-1β-stimulated OA chondrocytes, along with significant upregulation of MMP13 expression (Fig. [115]7B, C). Young MSCs at passage 5 (P5) effectively ameliorated the anabolic and catabolic imbalance induced by IL-1β in OA chondrocytes. However, both H₂O₂-induced prematurely senescent MSCs and replicatively senescent MSCs failed to mitigate metabolic dysregulation in OA chondrocytes (Fig. [116]7B, C). Notably, short-term DMOG treatment partially restored the therapeutic efficacy of both H₂O₂-induced prematurely senescent and replicatively senescent MSCs. Although the level of improvement did not fully match that of P5 young MSCs, the observed effects were statistically significant compared with the untreated senescent MSC groups (Fig. [117]7B, C). Quantitative real-time PCR (qRT-PCR) analysis further confirmed that DMOG-treated senescent MSCs significantly increased the mRNA expression of Col2a1 and AGC in OA chondrocytes, while concurrently suppressing MMP13 mRNA expression (Fig. [118]7D). In addition to restoring the paracrine regulatory function of senescent MSCs in OA chondrocytes, DMOG treatment also ameliorated their compromised multipotent differentiation capacity—effectively suppressing aberrant adipogenic differentiation while partially rescuing osteogenic and chondrogenic potential in hydrogen peroxide-induced aged MSCs (Fig. [119]S5). These findings suggest that DMOG may enhance the therapeutic potential of senescent MSCs, possibly by partially restoring their multipotent differentiation capacity and modulating their ability to regulate ECM-related gene and protein expression, which could contribute to mitigating the metabolic dysfunction in OA chondrocytes. Fig. 7. [120]Fig. 7 [121]Open in a new tab DMOG enhances the therapeutic potential of senescent MSCs to mitigate metabolic dysfunction in OA chondrocytes. (A) Schematic diagram of the co-culture system. Human articular chondrocytes were treated with IL-1β to induce OA-like conditions and co-cultured with MSCs from various experimental groups using a Transwell system. (B) Representative immunofluorescence staining of ECM-related proteins, including Collagen II (Col2a1), Aggrecan (AGC), and MMP13, in OA chondrocytes co-cultured with MSCs from different experimental groups. Scale bar = 50 μm. (C) Quantitative analysis of immunofluorescence intensity for Col2a1, AGC, and MMP13. (D) Quantitative real-time PCR (qRT-PCR) analysis of mRNA levels for Col2a1, AGC, and MMP13 in OA chondrocytes co-cultured with MSCs. Data are expressed as mean ± SEM (n = 3). *p < 0.05, **p < 0.01, ***p < 0.001 Discussion This study demonstrates that short-term DMOG treatment effectively rejuvenates senescent MSCs by targeting mitochondrial dysfunction and enhancing mitophagy possibly through the HIF-1α/BNIP3 signaling pathway. Hypoxia plays a critical role in maintaining the stem cell niche by reducing oxidative stress and preserving the properties [[122]10, [123]23, [124]24]. Studies have shown that hypoxic preconditioning can delay MSC senescence and improve their proliferation and differentiation capacities [[125]11, [126]13, [127]14, [128]19, [129]25, [130]26]. However, the effects of hypoxia on MSC senescence remain inconsistent. While most studies have suggested that hypoxia has anti-aging benefits, some have reported potential adverse effects [[131]27, [132]28]. These inconsistencies in previous findings may be attributed to differences in experimental conditions, such as hypoxic environment settings, oxygen concentrations, and MSC sources. As a hypoxia-mimetic agent, DMOG provides a reproducible and practical alternative by stabilizing HIF-1α under normoxic conditions, thereby eliminating the need for complex hypoxic culture systems [[133]16, [134]29]. This study is for the first to demonstrate that short-term DMOG treatment reverses aging-associated phenotypes in umbilical cord-derived MSCs in both oxidative stress-induced and replicative senescence models. It significantly reduces aging markers such as SA-β-gal, p53, and p21, while decreasing apoptosis and enhancing cell viability. These findings underscore the translational potential of DMOG as a robust strategy for delaying MSC senescence and restoring their functionality. A key mechanism underlying DMOG-induced rejuvenation is its ability to restore mitochondrial function, which is a hallmark of cellular aging and a primary driver of MSC senescence. Senescent MSCs exhibit increased ROS production, loss of mitochondrial membrane potential, and impaired ATP generation [[135]8, [136]30]. This study demonstrated that DMOG can reverse structural and functional mitochondrial damage. Transmission electron microscopy and functional analyses showed that DMOG improved the mitochondrial membrane potential, reduced ROS levels, enhanced ATP production, increased mitochondrial respiratory efficiency, and promoted mitophagy. These findings highlight the importance of mitochondrial quality control in combating senescence, consistent with previous studies linking mitochondrial homeostasis to stem cell rejuvenation [[137]7, [138]8]. Moreover, the role of mitochondrial dysfunction in aging is further underscored by previous studies showing that Ndufs6 deficiency in the mitochondria accelerates stem cell aging through excessive ROS accumulation and activation of the p53/p21 pathway [[139]31]. Previous studies have suggested that senescent MSCs exhibit significant declines in both oxidative phosphorylation and glycolytic capacity [[140]32]. This study demonstrated that DMOG partially restores oxidative phosphorylation and glycolytic capacity, improves the metabolic flexibility of senescent MSCs and supports the energy requirements of rejuvenated cells. Another key mechanism by which DMOG induces cellular rejuvenation is the activation of the HIF-1α/BNIP3 pathway, thereby regulating mitophagy. Mitophagy, a selective process for the removal of damaged mitochondria to maintain mitochondrial quality, protects bone marrow-derived mesenchymal stem cells (MSCs) against oxidative damage [[141]7, [142]33]. Previous studies have demonstrated that DMOG regulates the HIF-1α signaling pathway and enhances the biological functions of MSCs [[143]29]. Hypoxia protects nucleus pulposus-derived stem cells (NPSCs) under compression by increasing macroautophagy/autophagy levels, whereas HIF-1α alleviates compression-induced apoptosis in NPSCs through autophagy upregulation [[144]34]. BNIP3, a hypoxia-responsive gene regulated by HIF-1α, is critical for maintaining metabolic homeostasis and mitochondrial function in nucleus pulposus cells of intervertebral discs [[145]35]. In vivo animal experiments further confirmed that DMOG delays osteoarthritis (OA) progression in mice by stabilizing HIF-1α and enhancing mitophagy [[146]36]. This study demonstrated that DMOG significantly upregulates BNIP3 expression by stabilizing HIF-1α, thereby promoting mitophagy, improving mitochondrial quality, and reducing ROS accumulation. Transcriptomic and functional analyses further revealed that DMOG activates the HIF-1α/BNIP3 signaling pathway, which enhances mitophagy and improves the structural and functional integrity of the mitochondria. Notably, BNIP3 knockdown markedly attenuated the anti-aging effects and pro-mitophagy activities of DMOG, confirming the indispensable role of BNIP3 in the anti-aging mechanisms of DMOG. This finding aligns with previous studies, further establishing the critical regulatory role of the HIF-1α/BNIP3 axis in mitochondrial function and stress responses in senescent cells. Moreover, this highlights the potential of mitophagy as a therapeutic target for anti-aging interventions. The translational relevance of effects of DMOG on MSCs was further validated in an osteoarthritis (OA) co-culture model. Untreated senescent MSCs failed to restore the catabolic-anabolic balance in OA chondrocytes, whereas DMOG-treated MSCs showed significantly enhanced therapeutic efficacy. DMOG-treated MSCs promoted the expression of anabolic markers, including collagen II and aggrecan, while suppressing the catabolic marker MMP13, demonstrating their capacity to restore the therapeutic potential of senescent MSCs in degenerative disease contexts. Consistent with previous reports, hypoxia and hypoxia mimetic agents are promising priming approaches to enhance the potential of mesenchymal stem cells [[147]16]. Meanwhile, HIF-1α was stabilized and its nuclear localization enhanced by releasing DMOG from mesenchymal stem cell (MSC)-loaded alginate hydrogels, which was associated with both enhanced chondrogenesis and reduced expression of chondrocyte hypertrophy-related markers [[148]37]. However, the long-term effects of DMOG treatment and its potential for off-target effects require further investigation. Additionally, the observed differences in responses between replicative and oxidative stress-induced senescence emphasize the complexity of cellular aging and the need for tailored therapeutic strategies. Conclusions This study demonstrated for the first time that DMOG rejuvenates the senescence-associated phenotype of MSCs via HIF-1α/BNIP3-mediated mitophagy. By enhancing mitochondrial function, promoting mitophagy, and reprogramming cellular metabolism, DMOG has emerged as a practical and effective hypoxia-mimetic strategy to delay MSC senescence and preserve their functional capacity. This mechanistic study lays a theoretical foundation for developing innovative stem cell rejuvenation strategies and improving the therapeutic efficacy of MSCs in regenerative medicine. Electronic supplementary material Below is the link to the electronic supplementary material. [149]Supplementary Material 1^ (3.2MB, tif) [150]Supplementary Material 2^ (1.1MB, tif) [151]Supplementary Material 3^ (1.1MB, tif) [152]Supplementary Material 4^ (6.8MB, tif) [153]Supplementary Material 5^ (8.8MB, tif) [154]Supplementary Material 6^ (13.5MB, tif) [155]Supplementary Material 7^ (4MB, xlsx) [156]Supplementary Material 8^ (4.7MB, xlsx) [157]Supplementary Material 9^ (1.6MB, docx) Acknowledgements