Abstract Background Doxorubicin (DOX), a classical chemotherapeutic agent, faces significant limitations because of its well-documented risk of inducing cardiotoxicity. Effective prevention of DOX-induced cardiotoxicity is urgently needed. Given that alterations in metabolic pathways have been observed in both cardiovascular diseases and cancer, targeting specific metabolic pathways may offer dual benefits by mitigating DOX-induced cardiotoxicity while simultaneously enhancing its antitumor efficacy. Objectives This study sought to explore the characteristic metabolic alterations associated with early DOX-induced cardiotoxicity and identify a therapeutic target that simultaneously inhibits cancer and protects the myocardium. Methods Metabolomic and transcriptomic analyses were performed on heart tissues from murine models of DOX-induced cardiotoxicity to identify the most significantly altered metabolic pathway. The most altered metabolite involved in the candidate pathway was chosen and verified in both mice and humans. The protective effects of the chosen metabolite against DOX-induced cardiotoxicity and its antitumor effects were evaluated. Potential mechanisms were explored using C57BL/6J mice, OPLAH global knockout mice, BALB/c nude mice and NSG mice. Results The glutathione metabolic pathway was identified as the most altered pathway in heart tissues with DOX-induced cardiotoxicity. The 5-oxoproline/OPLAH (5-oxoprolinase) axis was the most critical node. Downregulation of 5-oxoproline was observed in serum samples from both humans and mice. Exogenous 5-oxoproline supplementation effectively restored myocardial 5-oxoproline levels, subsequently mitigating DOX-induced cardiac dysfunction. Interestingly, we observed an additional inhibitory effect of 5-oxoproline on tumor proliferation in tumor-bearing mice treated with DOX. Mechanistically, 5-oxoproline exerts its cardioprotective effects by restoring glutathione metabolic homeostasis through the modulation of its downstream enzyme OPLAH while simultaneously suppressing tumor proliferation by inhibiting its upstream enzyme, gamma-glutamyl cyclotransferase (GGCT). Conclusions This study reveals a previously unrecognized dual role of 5-oxoproline, which functions both as an early biomarker for DOX-induced cardiotoxicity detection and as a therapeutic target that simultaneously inhibits cancer growth and protects the myocardium. Keywords: Cardio-oncology, 5-oxoproline, Cardiotoxicity, Heart failure, Doxorubicin, Breast cancer Highlights * • 5-oxoproline is a promising prevention of DOX-induced cardiotoxicity. * • 5-oxoproline supplement may serve as an adjuvant treatment for tumors. * • 5-oxoproline is potential for early detection of DOX-induced cardiotoxicity. 1. Introduction Cardiovascular mortality has emerged as the leading cause of noncancer-related death among cancer patients [[47]1]. The emergence of cardio-oncology has increased interdisciplinary collaboration between oncologists and cardiologists, driving rapid advancements in preventing the side effects of antitumor chemotherapies. This field aims to improve patient prognosis through early detection, monitoring and treatment of cardiotoxicity. Doxorubicin (DOX), a classical anthracycline antibiotic, is widely used because of its potent antitumor effects [[48]2]. However, its clinical application is substantially limited by dose-dependent cardiotoxicity, known as DOX-induced cardiotoxicity, which leads to irreversible cardiac damage following cardiac fibrosis development. Early intervention in DOX-induced cardiotoxicity, even in asymptomatic patients, is crucial for preventing cardiac dysfunction and reducing adverse cardiac events [[49]3]. Dexrazoxane is currently the only FDA-approved agent for preventing DOX-induced cardiotoxicity. However, its use is limited by an elevated risk of secondary malignancies [[50]4]. Consequently, exploring early biomarkers of DOX-induced cardiotoxicity and developing preventive strategies to mitigate DOX-induced cardiotoxicity without compromising antitumor efficacy are urgently needed. Protein inhibitors, such as THZ1 and TP-10, as well as gene mutations in PI3Kγ and Mfn2, have shown promising cardioprotective and antineoplastic effects during DOX chemotherapy [[51][5], [52][6], [53][7], [54][8]]. However, these inhibitors are exogenous compounds rather than endogenous substances within the body. In contrast, biologically active metabolites are closely linked to human disease phenotypes, and they play crucial roles in regulating pathophysiological functions, such as anti-inflammatory, antioxidant, and metabolic regulation [[55][9], [56][10], [57][11]]. Various research groups, including ours, have reported that small-molecule metabolites have potential for the prevention and treatment of cardiovascular diseases [[58][12], [59][13], [60][14]]. Nonetheless, evidence of the therapeutic value of metabolites in DOX-induced cardiotoxicity remains scarce. Moreover, no studies have reported the dual effects of metabolite supplementation on both cardiac and tumor tissues during DOX therapy. This approach may represent a potential adjunctive therapy for cancers requiring anthracycline-based chemotherapy. In the present study, we explored metabolic alterations associated with early DOX-induced cardiotoxicity and identified therapeutic targets that simultaneously inhibit cancer progression and protect the myocardium. We found that glutathione synthesis was disrupted in early DOX-induced cardiotoxicity and identified 5-oxoproline as a biomarker for this condition. 5-Oxoproline exhibits antitumor effects while concurrently protecting the heart from DOX-induced cardiotoxicity. Mechanistically, 5-oxoproline exerts cardioprotective effects by enhancing glutathione synthesis via 5-oxoprolinase (OPLAH) and suppresses tumor growth through the downregulation of gamma-glutamyl cyclotransferase (GGCT) expression. 2. Methods The Supplemental Methods and Materials include details on the following methods: DOX-induced cardiotoxicity models and tumor models; culture and treatment of neonatal rat cardiomyocytes (NRCMs) and cancer cells; in vivo imaging analysis; histological analysis; terminal deoxynucleotidyl transferase dUTP nick-end labeling staining; seahorse respirometry; transmission electron microscopy; mitochondrial assays; C11 and DHE staining; detection of cell proliferation; measurement of MDA and GSH/GSSG; RNA sequencing; metabolomic profiling; 5-oxoproline analysis; liquid chromatography‒tandem mass spectrometry (LC‒MS); Western blot analysis; real-time quantitative polymerase chain reaction (RT‒qPCR); and ELISA. 3. Human studies All human samples used in this study were obtained according to protocols approved by the institutional review board of Shanghai Jiao Tong University (KY2023-070-C). Written informed consent was obtained from all participants, with approval from the Ethics Committee of Renji Hospital, School of Medicine, Shanghai Jiao Tong University. The study adhered to the guidelines of the Declaration of Helsinki (2013). Patients were included in this study if they met all the following criteria [[61]1]: female patients aged between 18 and 70 years [[62]2]; newly diagnosed with histologically confirmed breast cancer (stage I-IIIC) and scheduled to receive chemotherapy on the basis of evaluation by breast specialists [[63]3]; absence of cardiac symptoms or discomfort [[64]4]; no documented heart diseases [[65]5]; normal range of cardiac biomarkers, including troponin I (TnI) and brain natriuretic peptide (BNP), and left ventricular ejection fraction (LVEF) ≥ 50 % as determined on echocardiography [[66]6]; ability to complete face‒to-face follow-up visits; and [[67]7] willingness to provide written informed consent. The exclusion criteria were as follows [[68]1]: prior history of radiotherapy [[69]2]; another concurrent tumor [[70]3]; cardiovascular disease identified during screening at enrollment [[71]4]; uncontrolled blood pressure [[72]5]; severe chronic or acute renal failure (glomerular filtration rate <30 ml/min/1.73 m^2) [[73]6]; severe liver disease [[74]7]; history of immunodeficiency or organ transplantation; or [[75]8] any other medical condition assessed by the investigator as inappropriate for participation. One hundred eighteen plasma samples were collected from September 10, 2023, to July 15, 2024. Clinical data, including age, sex, history of hypertension, diabetes mellitus, hyperlipidemia, and current smoking, were collected. All patients received a predefined treatment protocol consisting of doxorubicin (80 mg/m^2) in 4 cycles, administered every 3 weeks. Echocardiography was performed at baseline of DOX therapy and 3 weeks after the final cycle. Plasma samples were separated by centrifugation at 400×g for 10 min at 4 °C and stored at −80 °C until further analysis. 4. Animal studies For the animal experiments, age-, sex-, and genetic background-matched mice were randomly separated into the indicated groups. All animal experiments were conducted in accordance with the National Institutes of Health Guidelines on the Care and Use of Laboratory Animals (NIH Publication, 8th Edition, 2011) and were approved by the Animal Ethics Committee of Renji Hospital (RJ 2023-148A). All mice were housed under specific pathogen-free conditions in temperature- and humidity-controlled rooms with a 12-h light/dark cycle and provided ad libitum access to food and water. Chronic Protocol: For the DOX-induced cardiac dysfunction model, 8-week-old male and female C57BL/6J mice received DOX (5 mg/kg) on days 0, 7, 14, and 21 [[76]15]. For the preventive test, three days before DOX treatment, mice were pretreated with 0.05 mg/kg/day 5-oxoproline or the same volume of vehicle (saline solution) via intraperitoneal injection and then continuously received the same treatment for another 28 days. For the therapeutic test, 0.05 mg/kg/day 5-oxoproline or the same volume of saline was intraperitoneally injected for another 4 weeks after 4 weeks of DOX treatment. Acute Protocol: In the preventive protocol, mice received 5-oxoproline (0.05 mg/kg/day, i.p.) or vehicle for 3 days prior to DOX administration, and treatment continued for an additional 7 days. DOX (15 mg/kg, i.p.) was administered as a single bolus to induce cardiotoxicity. In the rescue protocol, DOX (15 mg/kg, i.p.) was administered first, followed by 5-oxoproline treatment starting on day 7 and continuing for 7 consecutive days. For the syngeneic breast tumor model, 4T1 cells (1 × 10^6) mixed with 100 μL of Matrigel matrix were subcutaneously injected into female BALB/c nude mice aged 8 weeks. For the xenograft assays, 2 × 10^6 MDA-MB-231 cells were transplanted subcutaneously into the flanks of NSG mice aged 6 weeks. When the tumor volume reached 50–100 mm^3, the animals were randomly divided into 4 groups. DOX (5 mg/kg/week) and 5-oxoproline (0.05 mg/kg/day) were administered intraperitoneally as described above. Tumor size was measured twice a week using a caliper and was calculated according to formula (L × W^2)/2, with L and W being the longest and shortest tumor diameters, respectively, in millimeters. Cardiac function was evaluated via echocardiography 1 week after the last DOX administration. Images of the tumor were taken, and the final tumor weight was measured and recorded. To explore the mechanisms underlying DOX-induced cardiac dysfunction, male and female C57BL/6J OPLAH^+/+ or OPLAH^−/− mice aged 8 weeks were treated with DOX (5 mg/kg/week) and 5-oxoproline (0.05 mg/kg/day) as described above. Global OPLAH knockout (OPLAH^−/−) mice on a C57BL/6J background were generated using the clustered regularly interspaced short palindromic repeat/CRISPR-associated nuclease 9 (CRISPR/Cas9) system at Cyagen Biosciences, Inc. (China). The details are described in the Supplemental Material. OPLAH^+/+ littermates of the same age and sex were used as controls. 5. Statistical analysis The exact sample size of each group is listed in [77]Supplementary Table S1 or mentioned in the corresponding figure legends. Continuous variables are expressed as the means ± SEMs for parametric data or medians (interquartile ranges) for nonparametric data. Categorical variables are presented as counts and percentages. The normality of data distributions was assessed using the Shapiro‒Wilk test. For two-group comparisons, Levene's test was performed to assess the equality of variances. Normally distributed data were compared using the Student's t-test (equal variances) or Welch's t-test (unequal variances), whereas nonnormally distributed data were analyzed using the Mann‒Whitney U test. For comparisons involving more than two groups, homogeneity of variance was assessed with the Brown-Forsythe test. Normally distributed data were analyzed using one-way or two-way ANOVA, followed by Tukey's, Šídák's, or Bonferroni's post hoc correction, as appropriate [[78]12]. Multiway ANOVA was used when more than two independent variables were included, with Holm‒Šídák post hoc testing for pairwise comparisons. For nonparametric data, the Kruskal‒Wallis test was applied. For paired comparisons in human anthracycline-treated patients, the Wilcoxon matched-pairs signed-rank test was used. Correlations between continuous variables were assessed using Pearson's correlation analysis. Survival analyses were performed using the Kaplan‒Meier method, and the results were compared by log-rank tests. All statistical analyses were conducted using GraphPad Prism version 8.0 (GraphPad Software Inc., San Diego, CA, USA). A two-sided P value < 0.05 was considered to indicate a statistically significant difference. 6. Results 6.1. Glutathione metabolism is dysregulated in early DOX-induced cardiotoxicity To identify candidate therapeutic targets, we established a standard DOX-induced cardiac dysfunction mouse model and monitored dynamic cardiac functions ([79]Fig. 1A and [80]Figure S1, A-B). Since early detection of cardiac dysfunction is imperative, we used the global longitudinal strain (GLS) as an indicator of early cardiotoxicity in accordance with the European Society of Cardiology recommendation [[81]16]. One week after DOX administration was set as the early phase of myocardial impairment [[82]17], based on significant GLS reduction ([83]Fig. 1B) in the absence of alterations in traditional systolic function parameters, including the LVEF and left ventricular end-systolic diameter ([84]Figure S1, C-D). Fig. 1. [85]Fig. 1 [86]Open in a new tab Glutathione metabolism is dysregulated, and 5-oxoproline is decreased in the early phase of DOX-induced cardiotoxicity. A. Schematic protocol: Wild-type mice were treated with 4 rounds (once per week) of intraperitoneal DOX (5 mg/kg). Hearts were collected 24 h after the second administration of DOX for analysis of the early stage of DOX-induced cardiotoxicity. B. Global longitudinal strain and the global longitudinal strain rate were measured at 1 week and 4 weeks; n = 7. C. Volcano plot revealing the genes with altered expression in the control group and the group treated with DOX for 1 week (DOX-1W); n = 3. D. Volcano plot for Gene Ontology (GO) analysis of differentially expressed genes (DEGs). E. The top 10 Kyoto Encyclopedia Genes and Genomes (KEGG) pathways of the DEGs. F. Heatmap of altered cardiac tissue metabolites in the control group and DOX-1W group; n = 3. G. The top 15 KEGG pathways of the differentially expressed metabolites. H. Multiomic joint pathway analysis of transcriptomes and metabolomes. The highest-ranked pathway was glutathione metabolism. I. Schematic illustration of metabolites and enzymes involved in glutathione metabolism. J. 5-Oxoproline levels in the serum and heart tissues of the control and DOX-1W groups determined by liquid chromatography–mass spectrometry/MS analysis; n = 6. K. Association between 5-oxoproline (plasma and myocardium) and global longitudinal strain. Each symbol represents a single analysis; n = 12. The data are presented as the means ± SEMs. B. One-way ANOVA followed by Bonferroni's multiple comparison test. J. Student's t-test. K. Pearson correlation analysis. To investigate the early myocardial impairment induced by DOX, we performed bulk RNA sequencing and metabolomic analyses on heart samples from wild-type mice treated with either DOX or vehicle for 1 week [[87]17]. The differentially expressed genes (DEGs) identified between the two groups ([88]Fig. 1C) demonstrated that the response to oxidative stress represented the most significantly altered biological process according to Gene Ontology ([89]Fig. 1D). Glutathione metabolism was among the top 10 enriched pathways according to Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis ([90]Fig. 1E). Furthermore, differentially expressed metabolites were confirmed in cardiac tissues between the two groups by metabolomic analyses. KEGG pathway enrichment analysis revealed that several biological pathways were significantly affected by DOX treatment, with glutathione metabolism emerging as the predominant altered pathway ([91]Fig. 1F–G). To further explore the regulatory pathway, we performed a combined analysis of transcriptomes (differentially expressed genes, Benjamini‒Hochberg adjusted P < 0.05 and |log2[foldchange]|≥1) and metabolomics (metabolites, variable importance in projection >2 using orthogonal partial least squares‒discriminant analysis) with the MetaboAnalyst online tool ([92]https://www.metaboanalyst.ca) [[93]18]. As shown in [94]Fig. 1H, the glutathione metabolism pathway ranked as the top pathway according to the joint pathway analysis. These findings suggest that the modulation of glutathione metabolism might play a critical role in the pathogenesis of DOX-induced cardiotoxicity. 6.2. 5-Oxoproline declines in the early phase of DOX therapy The main regulators contributing to this DOX-induced glutathione metabolism pathway alteration were further explored. 5-Oxoproline and its associated catabolic enzyme, OPLAH ([95]Fig. 1I), were identified as the candidate metabolite and gene, respectively, owing to their most pronounced alterations. The levels of 5-oxoproline were evaluated by targeted metabolic measurements. A significant decrease in 5-oxoproline levels was observed in both the plasma and heart tissues of mice with DOX-induced cardiotoxicity as early as 1 week after modeling ([96]Fig. 1J). In addition, plasma and cardiac 5-oxoproline levels were inversely correlated with the GLS ([97]Fig. 1K), indicating a systemic metabolic response to early DOX-induced cardiotoxicity. 6.3. 5-Oxoproline supplementation alleviates DOX-induced cardiac cardiotoxicity and dysfunction in mice To determine whether 5-oxoproline supplementation attenuates DOX-induced cardiotoxicity, a pilot study was performed to confirm the optimal concentration of 5-oxoproline. As a result, supplementation at 0.05 mg/kg/day was shown to restore 5-oxoproline levels in both myocardial tissues and serum to physiological levels in mice ([98]Fig. S2A). 5-Oxoproline supplementation significantly attenuated the DOX-induced decreases in body weight ([99]Fig. 2A), heart weight ([100]Fig. 2B), and cardiac systolic function ([101]Fig. 2C–D). Additionally, it improved cardiac morphology ([102]Fig. S2B) and decreased the levels of myocardial damage biomarkers ([103]Fig. 2E–H). Fig. 2. [104]Fig. 2 [105]Open in a new tab 5-Oxoproline relieved DOX-induced cardiotoxicity in mice. A-B. Body weight and heart weight/body weight evaluated in groups treated with or without DOX and 5-oxoproline; n = 5. C-D. Representative images and quantification of echocardiography data; n = 5. E-H. Plasma levels of LDH, AST, TnT, and CK-MB; n = 5. I. Representative images of hematoxylin and eosin-stained hearts. Scale bars, 1 mm. J-K. Representative images and quantification of WGA staining. Scale bars, 40 μm; n = 5. L-M. Representative images and quantification of interstitial myocardial fibrosis. Scale bars, 50 μm; n = 5. N. Representative images and quantification of TUNEL staining. The arrows indicate apoptotic cardiomyocytes. Scale bars, 20 μm; n = 5. The data are presented as the means ± SEMs. A. Two-way ANOVA followed by the Šídák post hoc multicomparison test. B through N. Two-way ANOVA followed by the Bonferroni multiple comparison test. WGA = wheat germ agglutinin; 5-OXO = 5-oxoproline; LDH = lactate dehydrogenase; AST = aspartate aminotransferase; TnT = troponin T; CK-MB = creatine kinase-myocardial band. Moreover, DOX has been shown to elicit severe cardiac atrophy, fibrosis, and apoptosis [[106]17,[107]19]. Accordingly, we evaluated the effect of 5-oxoproline and found that it significantly attenuated DOX-induced myocardial atrophy, interstitial fibrosis, and cardiomyocyte apoptosis ([108]Fig. 2I–N). Furthermore, 5-oxoproline ameliorated cardiomyocyte apoptosis, as evidenced by a decrease in the proapoptotic factor Bax and cleaved caspase-3 and an increase in the antiapoptotic factor Bcl2 ([109]Fig. S2C). DOX treatment led to notable generation of reactive oxygen species (ROS), which are key inducers of oxidative stress and ferroptosis [[110]20]. The glutathione system plays a crucial role in regulating ferroptosis, as its activation can mitigate oxidative damage and protect against ferroptotic cell death in myocardial injury [[111]21]. Histological and biochemical analyses revealed that DOX treatment increased the levels of MDA and 4-HNE and decreased the levels of GSH/GSSG, indicating excessive lipid peroxidation. Interestingly, these changes were mitigated by 5-oxoproline ([112]Fig. 3A–C), which indicated that 5-oxoproline was involved in the modulation of ferroptosis in DOX-treated cardiomyocytes. In addition, we assessed markers related to ferroptosis, including GPX4 (the end terminator of ferroptosis) [[113]22], ACSL4 (an instigator of ferroptotic signaling) [[114]23], COX2 (an oxidative enzyme) [[115]24], and FTH1 (an iron scavenger) [[116]25]. Decreases in GPX4 and FTH1 and increases in ACSL4 and COX2 were simultaneously observed in cardiomyocytes following DOX treatment, and these changes were attenuated by 5-oxoproline ([117]Fig. 3D). Furthermore, we examined Nrf2, a crucial transcription factor for maintaining redox homeostasis [[118]26]. 5-Oxoproline restored Nrf2 protein levels in hearts subjected to DOX treatment, along with the levels of the downstream proteins HO-1 and SOD2, while reducing the production of protein oxidative byproducts, specifically 3-nitrotyrosine (3-NT) ([119]Fig. 3E). Moreover, DOX treatment led to pronounced mitochondrial damage in cardiomyocytes, characterized by evident mitochondrial swelling, vacuolization, and crista disintegration and lysis, and these features were markedly alleviated by exogenous 5-oxoproline supplementation ([120]Fig. 3F). In addition, necroptosis, a recently discovered form of programmed cell death, was also upregulated by doxorubicin, leading to cardiomyocyte death and cardiac dysfunction [[121]27]. Thus, we also evaluated necroptosis in our study. As shown in [122]Fig. 3G and H, the ratio of phosphorylated RIP1 to total RIP1 was significantly elevated following DOX administration, whereas cotreatment with 5-oxoproline effectively reduced this increase. Similarly, DOX treatment resulted in increased phosphorylation of RIP3 and MLKL, as reflected by increased p-RIP3/total RIP3 and p-MLKL/total MLKL ratios, both of which were attenuated upon 5-oxoproline treatment. Fig. 3. [123]Fig. 3 [124]Open in a new tab 5-Oxoproline inhibits DOX-induced ferroptosis, oxidative stress and necroptosis in vivo. A. Representative images and statistical data of 4-HNE staining. Scale bars, 50 μm; n = 5. B. Quantification of myocardial MDA; n = 5. C. Quantification of the myocardial GSH/GSSG ratio; n = 5. D. Representative images and statistical results of Western blot analysis of ferroptotic indicators (ACSL4, FTH1, GPX4, and COX2); n = 4. E. Representative images and statistical results of Western blot analysis of oxidative stress indicators (3-NT, SOD2, Nrf2, and HO-1); n = 5. F. Representative transmission electron microscopy images of the mitochondrial morphology of cardiac muscle. Scale bars, 1 μm. G-H. Representative images and statistical results of Western blot analysis of necroptosis indicators (p-RIP1/RIP1, p-RIP3/RIP3, and p-MLKL/MLKL); n = 5. The data are presented as the means ± SEMs. The data were analyzed by two-way ANOVA followed by the Bonferroni multiple comparison test. GSH = glutathione; MDA = malondialdehyde; 4-HNE = 4-hydroxynonenal; GSSG = oxidized glutathione; other abbreviations as in [125]Fig. 2. Importantly, in addition to its prophylactic effects, 5-oxoproline also demonstrated therapeutic efficacy. When administered after the onset of DOX-induced cardiac dysfunction, 5-oxoproline significantly improved cardiac function, reduced pathological remodeling and oxidative stress, and mitigated both programmed cell death and mitochondrial structural abnormalities compared with DOX alone ([126]Figs. S3 and S4). In addition, both the preventive and therapeutic effects of 5-oxoproline were independently validated in an acute DOX-induced cardiotoxicity model ([127]Figs. S5 and S6). Collectively, these findings indicate that 5-oxoproline confers both preventive and therapeutic protection against DOX-induced cardiotoxicity in both chronic and acute models, likely through the restoration of myocardial 5-oxoproline levels and the associated glutathione metabolic balance. 6.4. 5-Oxoproline attenuates DOX-induced cardiomyocyte death and mitochondrial dysfunction In vitro To further validate the role of 5-oxoproline, we investigated its effects on DOX-induced cardiotoxicity in vitro in neonatal rat cardiomyocytes (NRCMs). First, the optimal concentration of 5-oxoproline was determined to be 5 μM, as this was the lowest concentration that restored maximal cell viability ([128]Fig. S7A). Consistent with the results of the animal models, TUNEL staining ([129]Fig. S7B) and Western blot analysis ([130]Fig. S7C) demonstrated that 5-oxoproline significantly inhibited DOX-induced cell apoptosis and decreased the levels of apoptotic markers. Subsequently, intracellular ROS and lipid peroxidation were assessed in vitro in NRCMs. The results demonstrated that in NRCMs, DOX treatment markedly increased intracellular ROS ([131]Fig. 4A), lipid peroxidation ([132]Fig. 4B), and MDA levels ([133]Fig. S8A), while significantly decreasing the GSH/GSSG ratio ([134]Fig. S8B). However, all these detrimental effects were reversed by 5-oxoproline. Moreover, mitochondrial ROS levels were markedly increased following DOX treatment, leading to a significant decrease in the mitochondrial membrane potential (MMP), as indicated by elevated MitoSOX fluorescence and an increased JC-1 monomer/aggregate ratio ([135]Fig. 4C–F). These alterations were substantially attenuated by 5-oxoproline supplementation. To further evaluate mitochondrial function, real-time mitochondrial respiration was assessed using extracellular flux analysis. DOX exposure significantly reduced the oxygen consumption rate (OCR), including basal and maximal respiration, spare respiratory capacity, and ATP production ([136]Fig. 4G). Cotreatment with 5-oxoproline effectively ameliorated these impairments, indicating a protective effect on mitochondrial bioenergetics. Additionally, the downregulation of Nrf2, HO-1, SOD2, GPX4, and FTH1, along with the upregulation of 3-NT, p67phox, ACSL4, COX2 and necroptosis-related proteins (p-RIP1/RIP1, p-RIP3/RIP3, p-MLKL/MLKL) induced by DOX, was effectively prevented by 5-oxoproline ([137]Fig. S8C–E). Collectively, these findings demonstrate that 5-oxoproline effectively ameliorated DOX-induced cardiac dysfunction via the suppression of oxidative stress and ferroptosis. Fig. 4. [138]Fig. 4 [139]Open in a new tab 5-Oxoproline suppresses oxidative stress, ferroptosis and mitochondrial damage in vitro. A. Representative images and quantifications of DHE staining in NRCMs; n = 5. Scale bars, 150 μm. B. Analysis of lipid-ROS accumulation using C11 BODIPY 581/591 fluorescence staining; n = 5. Scale bars, 20 μm. C-D. Representative images and quantification of MitoSOX staining in NRCMs; n = 6. Scale bars, 20 μm. E-F. JC-1 staining for assessment of MMP expression in NRCMs. Representative images and quantification of JC-1 monomers/aggregates; n = 6. Scale bars, 20 μm. G. Mitochondrial OCR of NRCMs. Representative images and quantification of basal respiration, maximal respiration, spare respiratory capacity, and ATP production; n = 6. The data are presented as the means ± SEMs. The data were analyzed by two-way ANOVA followed by the Bonferroni multiple comparison test. NRCMs = neonatal rat cardiac myocytes; MMP = mitochondrial membrane potential; OCR = oxygen consumption rate; ROS = reactive oxygen species; DHE = dihydroethidium; other abbreviations as in [140]Fig. 2. 6.5. 5-Oxoproline inhibits tumor growth in addition to exerting cardioprotective effects The potential of 5-oxoproline as a cardioprotective agent in the context of anthracycline therapy was further evaluated in 4T1 tumor-bearing nude mice ([141]Fig. 5A). Consistently, 5-oxoproline supplementation significantly improved the survival rate and mitigated DOX-induced cardiotoxicity, including cardiac function, cardiac atrophy, fibrosis, and apoptosis ([142]Fig. 5B–E; [143]Fig. S9), in 4T1 tumor-bearing nude mice treated with DOX. Unexpectedly, 5-oxoproline delayed tumor growth in vivo ([144]Fig. 5F–H) and suppressed 4T1 cell proliferation both in vivo and in vitro ([145]Fig. 5I–K). Its antitumor effects were further enhanced by cotreatment with DOX. In addition to syngeneic tumor models, we also investigated the dual cardioprotective and antitumor efficacy of 5-oxoproline in xenograft assays. Consistent with the findings in the syngeneic tumor models, 5-oxoproline concurrently significantly inhibited tumor growth in human-derived xenografts while protecting against DOX-induced cardiotoxicity ([146]Figs. S10 and S11). Collectively, these findings reveal that 5-oxoproline is a novel adjuvant that not only protects against DOX-induced cardiotoxicity but also attenuates tumor growth. Fig. 5. [147]Fig. 5 [148]Open in a new tab 5-Oxoproline inhibits 4T1 tumor growth and simultaneously alleviates DOX-induced cardiotoxicity. A. Experimental scheme of 4T1 cell transplantation into the flank of BALB/c nude mice and treatment with either vehicle or 5-oxoproline (0.05 mg/kg/day) intraperitoneally 3 days before saline or DOX (5 mg/kg/week) administration continued for 4 weeks. The tumor volume was measured twice a week for 30 days in total. B. Survival curves in the indicated groups; n = 20. C. Heart weight/body weight; n = 6. D. Representative M-mode echocardiographic images. E. Ejection fraction and fractional shortening after 4 weeks of 5-oxoproline administration; n = 6. F‒G. The volume and weight of tumors in the indicated groups; n = 6. H. Photographs of the excised tumors in the indicated groups. I. Representative images and quantification of Ki67 staining; n = 6. Scale bars, 100 μm. J. CCK-8 activity of 4T1 cells treated with DOX or 5-oxoproline; n = 20. K. EdU assay of 4T1 cells; n = 6. Scale bars, 50 μm. The data are presented as the means ± SEMs. B. Survival data were analyzed by the Kaplan‒Meier method and compared by log-rank tests. F. Two-way ANOVA followed by the Šídák post hoc multicomparison test. C, E, G, I, K. Two-way ANOVA followed by the Bonferroni multiple comparison test. J. One-way ANOVA followed by the Bonferroni multiple comparison test. GGCT = gamma-glutamyl cyclotransferase; other abbreviations as in [149]Fig. 2, [150]Fig. 3. 6.6. 5-Oxoproline alleviates cardiotoxicity via OPLAH-mediated glutathione synthesis OPLAH, the enzyme responsible for converting 5-oxoproline into glutamate acid, is highly expressed in cardiomyocytes. Despite limited research, its role is critically important. Considering that glutamate acid is further converted into glutathione, which possesses robust antioxidant capacity in the heart, we hypothesized that 5-oxoproline might exert its cardioprotective effects through an OPLAH-mediated enzymatic reaction. To validate this hypothesis, OPLAH^+/+ and OPLAH^−/− mice ([151]Fig. 6A; [152]Fig. S12) were treated with DOX and administered either saline or 5-oxoproline daily for 4 weeks. As expected, 5-oxoproline supplementation alleviated DOX-induced cardiotoxicity in OPLAH^+/+ mice, as evidenced by improvements in body weight loss ([153]Fig. 6B), heart weight loss ([154]Fig. 6C), cardiac dysfunction ([155]Fig. 6D–E), cardiac injury ([156]Fig. 6F–I), cardiac atrophy and fibrosis ([157]Fig. 6J–N), cardiac ferroptosis ([158]Fig. 7A–D), cardiomyocyte apoptosis ([159]Fig. 7E; [160]Fig. S13A), mitochondrial dysmorphology ([161]Fig. 7F), cardiomyocyte necroptosis ([162]Fig. 7G–H) and oxidative stress ([163]Fig. S13B). However, these beneficial effects were completely abrogated when OPLAH was knocked out. Fig. 6. [164]Fig. 6 [165]Open in a new tab OPLAH deficiency eliminates the protective effect of 5-oxoproline on DOX-induced cardiotoxicity. A. Western blot images of hearts from OPLAH-WT or OPLAH-KO mice; n = 3. B–C. Body weight and heart weight/body weight; n = 5. D-E. Representative images and statistical data evaluated by echocardiography; n = 5. F–I. Plasma levels of LDH, AST, TnT, and CK-MB; n = 5. J. Representative images of hematoxylin and eosin staining. Scale bars, 1 mm. K. Representative images of WGA staining. Scale bars, 40 μm. L. Myocardial interstitial fibrosis. Scale bars, 50 μm. M. Quantification of total fibrosis; n = 5. N. Quantification of cardiomyocyte atrophy; n = 5. The data are presented as the means ± SEMs. B. Two-way ANOVA followed by the Šídák post hoc multicomparison test. C through N. Two-way ANOVA followed by the Bonferroni multiple comparison test. Abbreviations as in [166]Fig. 2. Fig. 7. [167]Fig. 7 [168]Open in a new tab OPLAH knockout abolishes the benefits of 5-oxoproline in vivo. A. Representative images and statistical data of 4-HNE staining. Scale bars, 50 μm; n = 5. B. Quantification of myocardial MDA; n = 5. C. Representative images and statistical results of Western blot analysis of ferroptotic indicators (ACSL4, FTH1, GPX4, and COX2); n = 4. D. Quantification of the myocardial GSH/GSSG ratio; n = 5. E. TUNEL staining of OPLAH-WT or OPLAH-KO mice treated with 5-oxoproline. The arrows indicate apoptotic cardiomyocytes. Scale bars, 20 μm; n = 5. F. Representative transmission electron microscopy images of the mitochondrial morphology of cardiac muscle. Scale bars, 1 μm. G-H. Representative images and statistical results of Western blot analysis of necroptosis indicators (p-RIP1/RIP1, p-RIP3/RIP3, and p-MLKL/MLKL); n = 5. The data are presented as the means ± SEMs. The data were analyzed by two-way ANOVA followed by the Bonferroni multiple comparison test. Abbreviations as in [169]Fig. 3. Consistent with the in vivo findings, the protective effects of 5-oxoproline against cellular apoptosis were significantly diminished by the downregulation of OPLAH in NRCMs ([170]Fig. S14A–C). Moreover, OPLAH knockdown impaired the inhibitory effects of 5-oxoproline on excessive ROS accumulation ([171]Fig. S14D–E), necroptosis ([172]Fig. S14F), lipid peroxidation ([173]Fig. S15A–D) and mitochondrial dysfunction ([174]Fig. S15E–I) in DOX-treated cardiomyocytes. These results indicate that 5-oxoproline exerts its cardioprotective effects in an OPLAH-dependent manner. 6.7. 5-Oxoproline suppresses tumor growth by inhibiting GGCT expression in tumor cells Since 5-oxoproline alleviated cardiotoxicity via glutathione synthesis, we first measured glutathione levels in 4T1 cells following 5-oxoproline administration. However, no significant changes in glutathione levels were observed after 5-oxoproline supplementation ([175]Fig. 8A). Unlike that in cardiomyocytes, the expression of OPLAH in tumor tissues was nearly 100-fold lower than that in heart tissues after DOX therapy ([176]Fig. 8B). In contrast to OPLAH, GGCT, the upstream enzyme of 5-oxoproline in the glutathione pathway, is upregulated in most tumors, and its depletion has been shown to inhibit tumor proliferation [[177]28]. In our study, higher levels of GGCT were associated with significantly shorter relapse-free survival in breast cancer patients ([178]Fig. 8C). Accordingly, we hypothesized that 5-oxoproline exerts an inhibitory effect on tumors through negative feedback inhibition of GGCT expression. Consistent with this hypothesis, we found that GGCT expression decreased in DOX-treated mice and was further reduced with 5-oxoproline supplementation ([179]Fig. 8D). Similarly, GGCT mRNA and protein levels were downregulated in 4T1 cells following DOX treatment, and 5-oxoproline further suppressed GGCT expression ([180]Fig. 8E–F). Moreover, GGCT knockdown significantly reduced 4T1 cell growth and proliferation, as shown by cell viability and EdU absorption ([181]Fig. 8G–I). These findings suggest that 5-oxoproline suppresses tumor growth by inhibiting GGCT expression in tumor cells. Fig. 8. [182]Fig. 8 [183]Open in a new tab 5-Oxoproline suppresses tumor growth by downregulating GGCT in tumor cells. A. GSH/GSSG ratios in 4T1 cells with or without the administration of DOX and 5-oxoproline; n = 6. B. Relative mRNA levels of OPLAH in heart tissues and tumor tissues; n = 6. C. Data were obtained from an online database of potential cancer biomarkers ([184]http://www.kmplot.com) to assess the prognostic implications of GGCT (Affymetrix ID: 215380_s_at) expression in patients with breast cancer. D. Representative images of GGCT staining; n = 6. Scale bars, 100 μm. E. Representative Western blot images and statistical results of GGCT in 4T1 cells with or without the administration of DOX and 5-oxoproline; n = 6. F. Relative mRNA levels of GGCT in 4T1 cells; n = 6. G. Representative Western blot images and relative mRNA levels of GGCT in 4T1 cells after transfection with NC-siRNA or GGCT-siRNA; n = 6. H. CCK-8 activity of 4T1 cells after transfection with NC-siRNA or GGCT-siRNA; n = 6. I. EdU assay of 4T1 cells after transfection with NC-siRNA or GGCT-siRNA; n = 6. Scale bars, 50 μm. The data are presented as the means ± SEMs. A, D, E, F, H, I. Two-way ANOVA followed by Bonferroni multiple comparison tests. B. Mann‒Whitney U test. G. Student's t-test. Abbreviations as in [185]Fig. 3, [186]Fig. 5. 6.8. Efficacy of 5-oxoproline for the early detection of anthracycline-induced cardiotoxicity To further verify the decreased level of 5-oxoproline in mice, we extended our investigation to human participants with breast cancer ([187]Fig. 9A; [188]Table S1). Using liquid chromatography‒mass spectrometry/MS analysis, we tested 118 blood samples from 59 patients. Plasma levels of circulating 5-oxoproline significantly decreased after the first cycle of anthracycline administration, whereas cTnI levels did not significantly change ([189]Fig. 9B). Among these patients, 12 were diagnosed with early cardiac dysfunction on the basis of GLS assessment via echocardiography. Receiver operating characteristic analysis revealed good performance of the 5-oxoproline prediction model in predicting early anthracycline-induced cardiotoxicity (AUC 0.848, 95 % CI: 0.745–0.950) ([190]Fig. 9C). In addition, a significant correlation was observed between decreased 5-oxoproline levels and an increase in the LVEDD, as well as a decrease in the IVST at anthracycline cycle 4 ([191]Fig. 9D–E). These findings suggest that 5-oxoproline could serve as a biomarker for the early detection of anthracycline-induced cardiotoxicity. Fig. 9. [192]Fig. 9 [193]Open in a new tab Role of 5-oxoproline in human anthracycline therapy. A. Schematic illustration of the human study design. B. Serum levels of 5-oxoproline, BNP, and cTnI at baseline and after 1 cycle of anthracycline administration; n = 59. C. ROC curve of the ability of 5-oxoproline to predict early anthracycline therapy-induced cardiotoxicity. D. LVEDD and IVST changes after 4 cycles of anthracycline administration; n = 15. E. Association between the 5-oxoproline change and left ventricular morphological change; n = 15. The values are presented as the means ± SEMs. B. Mann‒Whitney U test. D. Wilcoxon matched-pairs signed-rank test. E. Spearman's rank correlation. BNP = brain natriuretic peptide; LVEDD = left ventricular end-diastolic diameter; IVST = interventricular septal thickness. 7. Discussion In the present work, we identified 5-oxoproline as a key metabolite associated with DOX-induced cardiotoxicity, demonstrating its dual cardioprotective and antitumor functions. This study presents several novel findings (Central Illustration). First, the glutathione pathway was identified as the most significantly altered metabolic pathway in the cardiac tissues of patients with DOX-induced cardiotoxicity, and the 5-oxoproline/OPLAH axis was the critical node in the pathway. Second, treatment with 5-oxoproline alleviated DOX-induced cardiotoxicity, as evidenced by improved cardiac function, reduced oxidative stress, and attenuated cell death, while simultaneously enhancing the antitumor efficacy of DOX. Third, mechanistic investigations revealed that 5-oxoproline exerted cardioprotective effects by restoring glutathione metabolic homeostasis via its downstream enzyme OPLAH and inhibited tumor proliferation by suppressing its upstream enzyme GGCT. Collectively, these findings provide the first evidence for a novel regulatory role of the GGCT/5-oxoproline/OPLAH metabolic axis in mitigating cardiovascular complications and suppressing tumor growth during cancer therapy. Finally, the level of circulating 5-oxoproline may serve as an early biomarker for the detection of anthracycline-induced cardiotoxicity. In recent years, the incidence of anthracycline-induced cardiotoxicity has become increasingly prominent, largely due to improved cancer survival rates. The development of effective cardioprotective strategies remains an unmet clinical need. Several therapeutic approaches have been explored, including conventional agents such as carvedilol [[194]29] and vericiguat [[195]30], as well as small molecule inhibitors [[196]31] and microRNAs [[197]32]. Several studies have also focused on mechanisms of anthracycline-induced cardiotoxicity involving cellular senescence [[198]33] and immune modulation [[199]34]. However, little attention has been given to the role of endogenous small-molecule metabolites in mitigating anthracycline-induced cardiotoxicity. Compared with supplementation with exogenous chemical agents, supplementation with endogenous metabolites may better align with physiological processes and potentially reduce off-target effects. 5-Oxoproline is an important endogenous intermediate in the γ-glutamyl cycle, which is critical for glutathione synthesis. It is converted to glutamate via 5-oxoprolinase (OPLAH), thereby contributing to the replenishment of intracellular GSH pools [[200]35,[201]36]. Emerging studies have highlighted the regulatory role of the 5-oxoproline/OPLAH axis in maintaining both metabolic and redox homeostasis [[202]37,[203]38]. For example, OPLAH deficiency has been shown to cause oxidative stress, fibrosis, and elevated cardiac filling pressures [[204]39], underscoring the importance of this metabolic pathway in cardiovascular health. In the present study, through both exogenous supplementation with 5-oxoproline and in vivo experiments in OPLAH knockout mice, we demonstrated that 5-oxoproline confers both preventive and therapeutic benefits against DOX-induced cardiotoxicity in an OPLAH-dependent manner. Mechanistically, this protective effect is associated with the restoration of cardiac GSH metabolism. These findings reveal a previously unrecognized function of the 5-oxoproline/OPLAH axis and suggest its potential as a therapeutic strategy for mitigating anthracycline-induced cardiac injury. Glutathione, a critical nonenzymatic antioxidant in mammalian cells, plays a central role in redox regulation, protection against oxidative stress, and maintenance of cellular homeostasis. In addition to its well-established ability to scavenge ROS, GSH also regulates multiple forms of programmed cell death, including ferroptosis and apoptosis [[205][40], [206][41], [207][42], [208][43]]. Given the pivotal role of GSH in DOX-induced cardiotoxicity, GSH-related pathways—especially those involving ferroptosis and apoptosis—have emerged as key therapeutic targets for the prevention and treatment of cardiac injury [[209]44,[210]45]. For example, a small-molecule inhibitor targeting BAX has been shown to block both apoptosis and necrosis, thereby preventing DOX-induced cardiotoxicity [[211]31]. Similarly, the inhibition of ferroptosis, which is characterized by iron-dependent lipid peroxidation, significantly alleviates DOX-induced cardiac dysfunction and cardiomyocyte loss [[212]46,[213]47]. Consistent with these findings, our study revealed that activation of the 5-oxoproline/OPLAH axis restores GSH metabolism and ameliorates DOX-induced oxidative stress and cardiomyocyte death. These results not only reinforce the therapeutic importance of targeting ferroptosis and other regulated cell death pathways but also establish a novel upstream regulatory mechanism of cardiac GSH homeostasis, expanding our understanding of redox-based cardioprotective strategies. Ideally, optimal cardioprotective strategies should not only mitigate the primary mechanisms underlying chemotherapy-induced cardiotoxicity but also preserve—or even potentiate—the efficacy of antitumor treatment. Given the substantial differences between cardiomyocytes and cancer cells in terms of gene expression, protein profiles, and metabolic signatures, it is conceivable that the same therapeutic interventions may exert protective effects on the heart while enhancing the cytotoxicity of chemotherapy toward cancer cells. Indeed, a previous report demonstrated that PI3Kγ inhibition synergizes with the antitumor effects of chemotherapy and protects against DOX-induced cardiac injury [[214]7]. Similarly, our study reveals a striking dual role for 5-oxoproline: 5-oxoproline mitigates DOX-induced cardiomyocyte injury by reducing oxidative stress and cell death while concurrently synergizing with DOX to enhance overall antitumor efficacy. These divergent effects may be attributed to the differential expression of 5-oxoproline-related metabolic enzymes between cardiomyocytes and cancer cells. Specifically, the higher expression of OPLAH (5-oxoprolinase) in cardiomyocytes facilitates the conversion of 5-oxoproline to glutamate, thereby replenishing intracellular glutathione and strengthening antioxidant defenses. In contrast, the anticancer activity of 5-oxoproline may, at least in part, be mediated by its feedback inhibition of γ-glutamyl cyclotransferase (GGCT), an upstream oncogene involved in tumor metabolism and progression [[215]48]. These findings highlight a promising future research direction, wherein high-throughput omic technologies could be employed to elucidate molecular distinctions between cardiomyocytes and cancer cells, thereby guiding the development of therapeutics that offer both cardioprotection and enhanced antitumor efficacy. Early myocardial alterations that precede overt functional decline have been increasingly recognized as critical windows for therapeutic intervention in doxorubicin (DOX)-induced cardiotoxicity [[216]49]. In line with this concept, our study focused on the early phase of DOX-induced myocardial injury, aiming to identify sensitive biomarkers and actionable targets for timely detection and intervention before irreversible cardiac dysfunction develops. Previous studies [[217]50] have highlighted the utility of circulating microRNAs as promising genomic biomarkers for the early diagnosis of DOX-related cardiotoxicity. Extending this line of investigation, our study is the first to identify plasma 5-oxoproline as a potential early biomarker for anthracycline-induced myocardial injury. These findings not only underscore the clinical importance of targeting early metabolic disturbances but also open new avenues for developing minimally invasive diagnostic tools. Nevertheless, further validation in larger and more diverse patient cohorts across multiple cancer types and treatment settings is needed to establish the clinical applicability and predictive value of 5-oxoproline in routine cardio-oncology practice. 8. Study limitations Several limitations of this study should be acknowledged. First, DOX does not fully represent the cardiotoxic effects of all chemotherapeutic agents, and the cardioprotective effect of 5-oxoproline observed in this context may not be universally applicable to all forms of chemotherapy-induced cardiotoxicity. Second, while breast cancer is a major clinical context for DOX usage, we recognize that the findings cannot be directly generalized to other tumor types, which may differ in their microenvironments, metabolic dependencies, and responses to 5-oxoproline. Finally, our findings shed light on 5-oxoproline supplementation as a promising therapeutic avenue for DOX-induced cardiotoxicity. However, more clinical research is warranted to confirm the benefits observed in this study. 9. Conclusions In conclusion, our findings suggest that 5-oxoproline may serve as a promising small-molecule metabolite for preventing both DOX-induced cardiotoxicity and tumor growth. Thus, it may serve as an incremental therapeutic strategy for the treatment of cancer patients requiring anthracycline chemotherapy. Additionally, our findings provide a potential biomarker for the early detection of anthracycline-induced cardiotoxicity. [218]Image 1 [219]Open in a new tab CRediT authorship contribution statement Xinning Guo: Writing – review & editing, Writing – original draft, Formal analysis, Data curation, Conceptualization. Meng Jiang: Writing – review & editing, Resources, Funding acquisition. Zhengyu Tao: Writing – review & editing, Formal analysis, Data curation. Huijuan Dai: Writing – review & editing, Project administration, Methodology. Chen Wu: Formal analysis, Data curation. Yinan Wang: Formal analysis, Data curation. Zi Wang: Data curation. Xiaoning Wang: Data curation. Zhixuan Zhang: Investigation. Kun Qian: Resources, Methodology. Shanshan Zeng: Methodology, Investigation. Yihua Bei: Writing – review & editing, Project administration, Investigation. Jun Pu: Writing – review & editing, Resources, Investigation, Conceptualization. Central Illustration Schematic representation of the molecular mechanisms of 5-oxoproline in the heart and in tumors. In myocardial cells, 5-oxoproline regulates glutathione metabolism via OPLAH and alleviates cardiomyocyte ferroptosis caused by glutathione depletion; in tumor cells, it inhibits GGCT to suppress the growth of tumor cells without eliciting cardiac injury. Sources of funding This study received funding support from the National Natural Science Foundation of China (82421001, U21A20341 and 82470394), the Shanghai Municipality Science and Technology Commission (24DZ2202700 and 20Y11910500), the Shanghai Municipal Health Commission (202440156) and Basic-Clinical Collaborative Innovation Project from Shanghai Key Laboratory of Computational Chemistry and Nanomedicine (2025ZYB-007). Declaration of competing interest The authors declare that they have no competing interests. This manuscript is an original contribution not previously published and not under consideration for publication elsewhere. Acknowledgments