Abstract Bronchopulmonary dysplasia (BPD) is a prevalent chronic respiratory condition in preterm infants with an increasing incidence, severely affecting their survival rate and quality of life. Exploring the underlying mechanisms of BPD helps to develop novel effective therapeutic strategies. In this study, integrated metabolomic analyses of tracheal aspirates (TAs) from BPD infants and non-BPD infants, along with lung tissues from hyperoxia-induced experimental BPD neonatal rats and control rats, demonstrated that BPD was associated with a significant reduction in 3-hydroxyanthranilic acid (3-HAA), which was confirmed to be partly caused by tryptophan-metabolizing enzyme disorders. In vivo and in vitro models were subsequently established to assess the efficacy and underlying mechanisms of 3-HAA in relation to BPD. Compared with the BPD group, 3-HAA nebulization improved lung development and suppressed inflammation in rats. Limited proteolysis-small molecule mapping (LiP-SMap) proteomic analysis revealed the involvement of the ferroptosis pathway in the underlying mechanism by which 3-HAA alleviated hyperoxia-induced BPD injury. Ferroptosis was identified by detecting Fe^2+ levels, malondialdehyde (MDA), 4-HNE, total aldehydes, mitochondrial morphology, ferroptosis-associated protein and mRNA expression, and this dysregulation was indeed ameliorated by 3-HAA nebulization in vivo. Furthermore, a combination of LiP-SMap, molecular docking, SPR and Co-IP analyses confirmed that 3-HAA can bind directly to FTH1 and disrupt the nuclear receptor coactivator 4 (NCOA4)-FTH1 interaction. In conclusion, our study is the first to reveal that BPD is linked to the reduction of 3-HAA, and 3-HAA could inhibit the ferroptosis pathway by targeting FTH1, thereby alleviating hyperoxia-induced injury in rats and alveolar type II epithelial cells, highlighting the potential of targeting 3-HAA and ferroptosis for clinical applications in BPD. Keywords: Bronchopulmonary dysplasia, Neonatal rat, 3-Hydroxyanthranilic acid, Ferroptosis, Ferritin heavy chain 1, Alveolar type II epithelial cells Graphical abstract [37]Image 1 [38]Open in a new tab Highlights * • BPD is associated with a significant reduction in the tryptophan metabolite 3-hydroxyanthranilic acid (3-HAA). * • 3-HAA protects against hyperoxia-induced injury by inhibiting ferroptosis in lung tissue and AECII. * • 3-HAA inhibits the ferroptosis pathway by targeting FTH1 and disrupting the NCOA4-FTH1 interaction. * • This study first reveals the dysregulation of tryptophan metabolism in BPD and the mechanism of 3-HAA in alleviating BPD. 1. Introduction Bronchopulmonary dysplasia (BPD) is a prevalent chronic respiratory disease in preterm infants [[39]1], and its incidence has increased with the increasing survival rates of extremely premature newborns. BPD is characterized mainly by abnormal alveolar development, alveolar simplification, impaired pulmonary microvascular development, oxidative stress, and pulmonary inflammation [[40]2], which severely affect the survival rate and quality of life of preterm infants [[41]3,[42]4]. The pathogenesis of BPD remains incompletely understood, although it is thought to be the result of an imbalance between lung injury and repair processes in preterm infants with immature lung development [[43]5]. Despite advancements in perinatal care and the implementation of various therapeutic interventions in neonatal intensive care units (NICUs) to address BPD, no individual therapy has been shown to significantly decrease the incidence or severity of the condition. Considering the widespread occurrence of BPD and the limitations of existing treatments [[44]6,[45]7], it is essential to explore the mechanisms of BPD and to create novel, effective therapies that can counteract airway inflammation and alveolar abnormalities in affected infants. Cumulative research has indicated that BPD is associated with complex metabolic dysregulation [[46][8], [47][9], [48][10], [49][11]], including disturbances in essential amino acid metabolism. Changes in metabolites are believed to accurately reflect molecular characteristics that are closely linked to BPD, integrating the internal biological state and its external environment [[50]12]. Tryptophan, which has gained much attention in recent years, is metabolized through the 5-hydroxytryptamine (5-HT), kynurenine, and indole pathways, and its metabolites possess a variety of biological activities, such as anti-inflammatory, anti-tumor, and antioxidant effects [[51]13,[52]14]. Most papers address the contributions of tryptophan and its metabolites to pathologies of the central nervous system, the digestive system, and the immune system. However, their contribution to the respiratory system has almost been ignored and understudied [[53]15]. Some evidence indicates the potential role of indole-3-acetic acid, a tryptophan metabolite, in modulating inflammation, emphysema and decreased lung function in chronic obstructive pulmonary disease (COPD) [[54]16], which shares some pathological features with BPD. However, the complex functions and mechanisms of tryptophan metabolites in neonatal BPD remain to be explored. Given that tryptophan metabolism is affected under pathological conditions, tryptophan and its metabolites are very attractive biomarkers for diagnosis, prognosis, and treatment. In addition, the biological effects of tryptophan metabolites and their changes in disease suggest that they may serve as potential therapeutic targets. In this study, we identified a reduction in the tryptophan metabolite 3-hydroxyanthranilic acid (3-HAA) in both the tracheal aspirates (TAs) of infants with BPD and the lung tissues of hyperoxia-induced experimental BPD rats. Mechanistically, our data revealed that 3-HAA inhibited the ferroptosis pathway by targeting ferritin heavy chain 1 (FTH1) and disrupting the NCOA4-FTH1 interaction, thereby alleviating hyperoxia-induced damage in rats and alveolar type II epithelial cells (AECII). Collectively, our findings offer new insights into the management and treatment of BPD, potentially leading to targeted therapies that address the metabolic dysfunctions associated with BPD. 2. Materials and methods 2.1. TA samples and sample size Infants with a gestational age (GA) < 32 weeks who underwent invasive mechanical ventilation (IMV) on the first day of life at the NICU of the Children's Hospital of Chongqing Medical University (CHCMU) between June 2023 and October 2023 were included in this study, and parents provided informed consent. TA samples were collected during routine tracheal aspiration in infants receiving mechanical ventilation as previously described [[55]17] and frozen at −80 °C until tryptophan metabolites were analyzed. Sample size estimation was performed using G∗Power 3.1 software [[56]18] with the following parameters: two-tailed α = 0.05, Cohen's effect size d = 1.5 (based on previous research [[57]19,[58]20]), and power (1 - β) of 0.8 [[59]21]. The calculation indicated that a sample size of at least 9 infants per group was required to detect significant differences between the groups. Relevant clinical data were collected from electronic medical records. The infants were followed up until 36 weeks postmenstrual age and categorized into the BPD group and the non-BPD control group. BPD was diagnosed according to the National Institutes of Health workshop definition in 2018 [[60]22]. After confirming that there were no differences between the two groups in terms of postmenstrual age or weight at the time of TA sample collection (postmenstrual age: 30.9 ± 1.2 vs 31.2 ± 0.8 weeks, p = 0.49; weight: 1404.0 ± 290.3 vs 1473.0 ± 258.4 g, p = 0.58), 10 infants with BPD and 10 non-BPD controls were finally enrolled and TA samples were subsequently analyzed. The clinical characteristics of the infants are summarized in [61]Table S1. The Institutional Review Board of CHCMU approved this study (2023–258), which was registered on [62]chictr.org (ChiCTR2400087152). 2.2. Animals and interventions The Animal Ethics Committee of CHCMU approved the animal experiment protocol (No. CHCMU-IACUC20240412007), which complied with the [63]ARRIVE guidelines. The hyperoxia-induced experimental BPD neonatal rat model [[64]23] is recognized as an appropriate animal model for investigating the pathogenesis of BPD and exploring new therapeutic approaches. Neonatal rats are born during the saccular stage of lung development, which is closely similar to that of preterm infants, and exhibit pathological features of BPD under hyperoxic conditions, and have genetic heterogeneity observed in human populations. Pregnant Sprague‒Dawley (SD) rats were obtained from Chongqing Medical University (Chongqing, China). They were housed individually and provided unrestricted access to food and water under conditions of 20–25 °C with a 12:12-h light‒dark cycle, awaiting the birth of the neonates. Newborn SD rats in the BPD group, together with their nursing dams, were placed in a hyperoxia chamber within 12 h of birth that maintained an oxygen concentration of 85 % for 14 days as previously described [[65]8]. The rats in the control group were exposed to air containing 21 % oxygen. The rats in the BPD + 3-HAA group were nebulized every other day (P2, P4, P6, P8, P10, P12, and P14) for 30–50 min each time during the 14-day hyperoxia modeling period. During the nebulization process, newborn rats were placed in a specialized chamber connected to a nebulizer (403S, yuwell), which delivered an aerosolized form of 3-HAA. To minimize maternal and nutritional variations affecting lung development, the number of newborn rats per chamber was equalized, and nursing dams were rotated across all experimental groups. 2.3. Preparation of 3-hydroxyanthranilic acid-nebulized solutions After identifying the differential tryptophan metabolite 3-HAA, we prepared a solution of 3-HAA for nebulized intervention in neonatal rats. 3-HAA (CAS No. 548-93-6, AbMole) was dissolved in dimethyl sulfoxide (DMSO) and brought to a final concentration of 0.5 mg/mL in phosphate-buffered saline (PBS) immediately before use, with a DMSO concentration of 1.7 %. To explore the appropriate dose and effectiveness of 3-HAA nebulization, newborn rats were randomly divided into eight groups: control, control + DMSO, BPD, BPD + DMSO, BPD+3-HAA (25 mg/kg), BPD+3-HAA (50 mg/kg), BPD+3-HAA (75 mg/kg), and BPD+3-HAA (100 mg/kg). Finally, 75 mg/kg was chosen for further experiments. 2.4. Lung sample collection The rats were anesthetized via inhalation of isoflurane on the 15th day after birth, after which trachea and lung tissues were isolated. The lung tissues were lavaged through the right ventricle with precooled PBS (4 °C), and then the right lung tissues were excised and stored at −80 °C for subsequent biochemical analyses. The left lobe was inflated and fixed in situ via the trachea with 4 % paraformaldehyde (PFA) at a pressure of 20 cmH[2]O [[66]24,[67]25] and then incubated in 4 % PFA at 4 °C overnight, enabling subsequent hematoxylin‒eosin (HE) staining, immunohistochemical (IHC) staining and immunofluorescence (IF) staining. 2.5. Metabolomic analysis A targeted metabolomics approach was employed to analyze the levels of tryptophan metabolites in infant TA samples and rat lung tissues. Metabolites were extracted and analyzed via ultra-high-performance liquid chromatography coupled with tandem mass spectrometry (UHPLC-MS/MS) metabolomics as previously described [[68]26,[69]27]. 2.6. Identification of proteins interacting with 3-HAA Identification of proteins interacting with 3-HAA was conducted using the limited proteolysis-small molecule mapping (LiP-SMap) chemical proteomics approach, following established protocols [[70][28], [71][29], [72][30]]. First, lung tissue lysates in the BPD group were aliquoted into 6 equivalent volumes, each containing 100 μg of protein. To determine the proteins that interact with 3-HAA, 0.33 nmol/(μg total protein) 3-HAA was introduced to 3 of the 6 independent replicates and DMSO was added to the other 3 replicates, which were then incubated at 25 °C for 10 min. Proteinase K was added to all samples at a 1:100 enzyme/substrate ratio and incubated at 25 °C for 5 min. The protein fragments generated from the proteinase K-limited proteolysis step were further digested with trypsin at a 1:50 trypsin/substrate ratio to produce peptides for mass spectrometry analysis using a nano-UPLC (nanoElute2, Bruker) coupled to a timsTOF Pro2 instrument (Bruker). 2.7. Cell models Primary human AECII are difficult to passage and expand in vitro, and we used immortalized human AECII (ZQY095, ZQXZ-bio, China) induced by the SV40 gene fragment to study human AECII biology [[73]31]. The cells were grown in H-DMEM (Gibco, USA) supplemented with 10 % FBS, 100 IU/mL penicillin and 100 μg/mL streptomycin (C0222, Beyotime) and maintained in an incubator containing 5 % CO[2] at a suitable temperature (37 °C). AECII were divided into the control group (21 % O2) and hyperoxia group (85 % O2) to confirm the level of 3-HAA. Then in vitro experiments were performed to further demonstrate the mechanisms underlying the modulation of ferroptosis in AECII responded by the 3-HAA. AECII were randomly divided into the control (C) group, hyperoxia (H) group, hyperoxia+3-HAA (HA) group, hyperoxia+3-HAA + erastin (ferroptosis agonist, MCE, HY-15763) (HAE) group, and hyperoxia+3-HAA + deferoxamine (DFO) (ferroptosis inhibitor, MCE, HY-B1625) (HAD) group. The concentrations of 3-HAA, erastin and DFO were determined on the basis of cell viability with a cell counting kit-8 (CCK-8) assay. 2.8. Cell viability assay To explore the appropriate concentrations of 3-HAA, erastin and DFO, they were dissolved in DMSO respectively and brought to different concentrations in serum-free medium immediately before use. AECII were plated in 96-well plates and divided into control and hyperoxia groups. Subsequently, 3-HAA (20, 40, 60, 80, 100, 120, 140, and 160 μM), DFO (10, 25, 50, 75, and 100 μM) and erastin (1, 2, 5, 10, and 20 μM) were administered to both groups, respectively, for 24 h. Cell viability was assessed using the CCK-8 (K1018, ApexBio) assay as described previously [[74]32]. The working concentration of the CCK-8 solution was prepared, and the solution was added to each well. A microplate reader (BioTek Synergy H1) was used to analyze the absorbance (450 nm) after culturing for 2 h at 37 °C. 2.9. Histological analysis, IHC staining, and IF staining of lung tissue The 4 % PFA-fixed lung tissues were dehydrated with gradient ethanol and xylene before being embedded in paraffin. Paraffin-embedded tissues were sliced into 4 μm sections for morphological analysis using HE staining. The radial alveolar count (RAC) was used to assess the development of alveoli [[75]2], and the mean linear intercept (MLI) was used to assess the volume‒to‒surface ratio of alveolar airspaces [[76]33]. Histological images were examined via an optical microscope (Nikon, Japan), with three random fields of view selected per sample. Paraffin-embedded lung tissue sections were deparaffinized, hydrated, microwaved in Tris-EDTA buffer for antigen retrieval, incubated with 3 % hydrogen peroxide for 30 min, and then blocked with 3 % BSA for 1 h at room temperature. For IHC staining, the sections were incubated with primary antibodies of interest at 4 °C overnight. Then, goat anti-rabbit IgG or goat anti-mouse IgG was added, and the samples were incubated for 30 min at room temperature. Subsequently, the DAB reaction was performed. Finally, the sections were counterstained, mounted, and examined under an optical microscope (Nikon, Japan). For IF staining, the sections were incubated with specific primary antibodies at 4 °C overnight and then incubated with an HRP-conjugated secondary antibody for 30 min at room temperature and with tyramide signal amplification (TSA) fluorescent dyes (AFIHC024, Aifang) [[77]34] for 10 min, followed by DAPI staining for 10 min. Images were captured using a fluorescence scanner (KF-FL-020, KFBIO), with three random fields of view selected per sample. The antibodies used are presented in [78]Table S2. 2.10. IF staining and live-cell imaging of AECII AECII were fixed in 4 % PFA for 20 min and subsequently washed three times with PBS. The cell slides were blocked with 10 % goat serum for 10 min following incubation with 0.1 % Triton X-100 for 20 min if necessary and then incubated overnight at 4 °C with the primary antibodies of interest. The appropriate HRP-conjugated secondary antibody was used for 50 min at room temperature, and TSA fluorescent dyes (AFIHC024, Aifang) were added for 5 min, followed by DAPI staining for 6 min. Images were captured using a fluorescence scanner (KF-FL-020, KFBIO), with three randomly selected fields of view per sample. The antibodies used are presented in [79]Table S2. The reactive oxygen species (ROS) level in AECII was measured by the fluorescent probe DCFH-DA (S0033S, Beyotime) [[80]35,[81]36], and lipid peroxidation was assayed by Liperfluo [[82]37,[83]38] (L248, Dojindo). Briefly, cells in different groups were stained with 10 μM DCFH-DA at 37 °C for 20 min or 1 μM Liperfluo at 37 °C for 30 min. After being washed with serum-free medium three times, images were acquired and analyzed using NIS-Elements software at an excitation wavelength of 488 nm (Nikon, Japan). 2.11. Amplex Red hydrogen peroxide (H[2]O[2]) assay The extracellular H[2]O[2] levels were measured using the Amplex Red Hydrogen Peroxide Assay Kit (A22188, Invitrogen), following the manufacturer's instructions [[84]39]. Cell culture supernatant of different experimental groups was collected and incubated with 100 μM Amplex Red reagent containing 0.2 U/mL horseradish peroxidase for 30 min at room temperature. Fluorescence was detected using a microplate reader with excitation at 540 nm and emission at 590 nm, and concentrations were determined based on a standard curve. 2.12. Measurement of redox- and ferroptosis-associated markers The content of total aldehydes in the lung tissues and AECII cells was detected using an Amplite™ Fluorometric Aldehyde Quantitation Kit [[85]36]. The levels of malondialdehyde (MDA) and Fe^2+ in lung tissues and AECII samples were detected by using commercially available assay kits. Reduced glutathione (GSH) levels and superoxide dismutase (SOD) activity in AECII were detected by the corresponding commercial reagents. These markers were detected following the manufacturers’ instructions, and the commercial kits used are presented in [86]Table S3. 2.13. Enzyme-linked immunosorbent assay (ELISA) Lung tissues were homogenized in PBS, and supernatants were obtained by centrifugation at 12,000 rpm and 4 °C for 10 min for analysis of transforming growth factor-beta 1 (TGF-β1), tumor necrosis factor-alpha (TNF-α), vascular endothelial growth factor (VEGF), and interleukin 6 (IL-6) levels. The cell culture supernatant was centrifuged at 1000×g and 4 °C for 20 min for TNF-α and IL-6 analysis. These factors were detected by ELISA kits according to the manufacturers' instructions. The total protein levels in the lung tissues were quantified by a BCA assay (P0010, Beyotime). The absorbance at 562 nm was measured with a microplate reader (BioTek Synergy H1). The ELISA kits used are presented in Table S3. 2.14. Western blot (WB) The lung tissues and AECII samples were lysed with RIPA lysis buffer and then centrifuged at 4 °C and 12000 rpm for 10 min, after which the supernatant was collected. Protein was quantified using a BCA assay kit (P0010, Beyotime) and then diluted with loading buffer, and boiled for 10 min. After electrophoresis on SDS-polyacrylamide gels at 80 V for 30 min and 120 V for 60 min, the proteins were transferred to polyvinylidene difluoride (PVDF) membranes with a pore size of 0.45 μm at 400 mA for 30 min by NcmBlot Rapid Transfer Buffer. The PVDF membrane was quickly blocked for 15 min at room temperature and then incubated with primary antibodies overnight at 4 °C and the appropriate secondary antibody goat anti-rabbit IgG (511203, Zenbio) at room temperature for 1 h. The images of protein bands were captured by ChemiDoc™ Touch Imaging System (BIO-RAD) using an enhanced chemiluminescence (ECL) solution kit (17047, Zenbio), and analyzed using Image lab and ImageJ software. The antibodies used for WB are presented in [87]Table S2, with β-actin serving as a reference. 2.15. Real-time quantitative PCR (RT‒qPCR) Total RNA was isolated and purified from rat lung tissue and AECII samples following the protocol provided by Accurate Biotechnology (AG21023). The RNA yield and quality were evaluated by a NanoDrop spectrophotometer (Thermo Fisher Scientific) with OD260/OD280 ratios between 1.8 and 2.2. Evo M-MLV RT Premix for qPCR (AG11734, Accurate Biotechnology) and the SYBR Green Premix Pro Taq HS qPCR Kit (AG11733, Accurate Biotechnology) were used for reverse transcription and amplification, respectively. Gene expression analysis was performed on a BioRAD Real-Time PCR System, with β-actin serving as a reference, and relative mRNA expression levels were calculated via the 2 ^(−ΔΔCt) method. The primers used for RT‒qPCR are shown in [88]Table S4. 2.16. Transmission electron microscopy (TEM) Fresh lung tissues and collected cells were fixed in 2.5 % glutaraldehyde, followed by 2 h of fixation with 1 % osmium tetroxide, and subsequently dehydrated using a graded ethanol series. The samples were embedded in acetone overnight and sectioned into ultrathin slices (60–80 nm). Finally, the slices were stained with lead citrate and 2 % uranyl acetate. The sections were observed using an HT-7800 TEM (HITACHI, Tokyo, Japan) or a Thermo Scientific™ Talos™ L120C. 2.17. Molecular docking The interaction between 3-HAA and FTH1 was calculated using a computational docking method [[89]40,[90]41] (Autodock vina 1.1.2). The molecular structure of the 3-HAA ligand was sourced from PubChem (CID: 86). The FTH1 structure used for docking was obtained from a Protein Data Bank (PDB) (PDB ID: [91]4OYN). After subjecting the ligand 3-HAA and the receptor FTH1 to hydrogenation and structure optimization, 3-HAA was docked into the potential binding pocket of FTH1. 2.18. Surface plasmon resonance (SPR) The SPR technique, which is a robust method for analyzing real-time biomolecular interactions without the need for labeling, was utilized to identify the interaction affinity between the recombinant human FTH1 protein and compound 3-HAA. The FTH1 protein was directly coupled to the CM5 chip (BR-1005-30, China) for determination. Following incubation, a gradient of 3-HAA was introduced into the FTH1 protein-CM5 system. Similarly, to explore whether human FTH1 interacts with human NCOA4, the recombinant human NCOA4 protein was directly coupled on the CM5 chip, and a gradient of FTH1 protein was introduced into the NCOA4 protein-CM5 system. To determine whether 3-HAA affects the binding of FTH1 to NCOA4, FTH1 protein with and without 3-HAA was introduced into the NCOA4 protein-CM5 system. Binding interactions were analyzed using BIAcore T200 evaluation software (v.2.0, GE Healthcare, USA). The procedure followed the methodology described in previous studies [[92]42,[93]43]. 2.19. Co-immunoprecipitation (Co-IP) assay A Co-IP assay was used to investigate the interaction between NCOA4 and FTH1 in AECII following treatment with 3-HAA. The cells were lysed with IP buffer containing protease inhibitor cocktails, and the total protein concentration was assessed with a BCA assay kit. The lysate supernatant was incubated with 10 μL anti-NCOA4 (ab314553, Abcam) antibody or regular IgG overnight at 4 °C, and repeatedly washed protein A/G agarose beads were added to capture the immunocomplexes. Then, the immunocomplexes were washed 3 times with IP lysis buffer, and the samples were boiled in SDS‒PAGE loading buffer and subjected to WB analysis [[94]44]. 2.20. Statistical analysis Statistical analyses were conducted with SPSS software 26.0 and GraphPad Prism 9.0. All experiments were repeated at least three times biologically, data were presented as mean ± standard deviation (SD). Statistical significance between two groups was determined by Student's t-test, whereas comparisons across three or more groups were performed by one-way analysis of variance (ANOVA) with Tukey's post hoc test for multiple comparisons. Cohen's d was employed as a measure of effect size to quantify the degree of intergroup differences in metabolites, calculated by dividing the mean difference between groups by the pooled standard deviation. Spearman's rank correlation coefficient was performed to quantify the associations between metabolites and clinical characteristics. The Kyoto Encyclopedia of Genes and Genomes (KEGG) database was utilized for pathway evaluation. A p value < 0.05 was considered statistically significant throughout the study (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, ∗∗∗∗p < 0.0001 in the figures). 3. Results 3.1. BPD was associated with decreased 3-HAA In our study, the scheme for identifying differential tryptophan metabolites is shown in [95]Fig. 1A. UHPLC-MS/MS metabolomics analysis was performed to explore the tryptophan metabolites in the TAs of BPD infants and non-BPD infants. The orthogonal projections to latent structures discriminant analysis (OPLS-DA) model revealed the distinction between the two groups (n = 10) ([96]Fig. 1B). Twenty-one metabolites were identified in the TAs, among which 3-HAA (P = 0.033, log[2] fold change (FC) = -2.355, Cohen's d = 1.084) and ILA (P = 0.003, log[2] FC = −0.996, Cohen's d = 1.567) were decreased in the BPD group compared with the control group ([97]Fig. 1C). Correlation analysis among tryptophan metabolites and clinical characteristics revealed that there was no correlation between 3-HAA levels and the duration of antibiotic use ([98]Fig. S1). Fig. 1. [99]Fig. 1 [100]Open in a new tab BPD was associated with decreased 3-HAA. (A): Scheme for the identification of differential tryptophan metabolites in BPD. (B): OPLS-DA statistical model for tryptophan metabolites in TA samples. Each point represents a single sample, colored by group. (C): Volcano plot of detected metabolites in TAs. Red indicates an upregulation in BPD, blue shows a downregulation in BPD, and the circle sizes refer to variable importance in projection (VIP). (D): Representative HE-stained lung tissue sections from control and BPD group rats, scale bar = 25 μm. (E): OPLS-DA statistical model for tryptophan metabolites in lung tissues. Each point represents a single sample, colored by group. (F): Volcano plot of detected metabolites of rat lung tissues. Red indicates an upregulation in BPD, blue shows a downregulation in BPD, and the circle sizes refer to the VIP. Subsequently, to investigate whether tryptophan metabolic dysregulation also exists in an experimental setting, we utilized a well-established SD rat model of hyperoxia-induced BPD. Compared with the control group, the BPD group presented significant alveolar simplification, indicating that there was impaired lung development ([101]Fig. 1D). Therefore, this preclinical rat model is suitable for further exploration of tryptophan metabolites and their role in BPD. As expected, the OPLS-DA model revealed the distinction between the BPD group and the control group (n = 8) ([102]Fig. 1E). Twenty-seven metabolites were identified, and more metabolites were altered in lung tissue, among which 3-HAA (p = 0.0001, log[2] FC = −1.328, Cohen's d = 2.611), NAS (p = 0.015, log[2] FC = −3.851, Cohen's d = 1.603), and 5-HTOL (p = 0.017, log[2] FC = −1.302, Cohen's d = 1.440) decreased in the BPD group compared with the control group ([103]Fig. 1F). Taken together, only 3-HAA was reduced in both the TA samples and the lung tissues from the BPD group and was subsequently selected for further investigation. 3.2. Dysregulation of related metabolic enzyme led to decreased 3-HAA IHC staining for 3-HAA ([104]Fig. 2A) was then performed on rat lung tissues, while IF staining for 3-HAA ([105]Fig. 2G) was conducted on AECII, confirming the reduction in 3-HAA in both in vivo and in vitro hyperoxia models. 3-HAA is an important intermediate metabolite in the Kynurenine (KYN) metabolic pathway. KYN is converted into 3-Hydroxykynurenine by kynurenine 3-monooxygenase (KMO), and then catalyzed by kynureninase (KYNU) to generate 3-HAA. In the subsequent metabolic process, 3-HAA is further catalyzed by downstream enzymes such as 3-hydroxyanthranilate 3,4-dioxygenase (HAAO) to generate Quinolinic acid, etc. ([106]Fig. S2). To investigate the underlying reasons for the differences in 3-HAA levels between groups, WB analysis ([107]Fig. 2B and C) and IHC staining ([108]Fig. 2D–F) of the 3-HAA metabolism-related enzymes KMO, KYNU, and HAAO were performed on rat lung tissues. The results demonstrated that the protein expressions of KMO and KYNU were decreased in the BPD group, which led to a decrease in the production of 3-HAA, whereas the elevated protein expression of HAAO led to an increase in the downstream metabolism of 3-HAA. Additionally, WB analysis ([109]Fig. 2H and I) and IF staining ([110]Fig. 2J-L) of AECII revealed a similar pattern: KYNU and KMO decreased and HAAO increased in the hyperoxia group. The RT‒qPCR results were also consistent in both in vivo and in vitro hyperoxia models ([111]Fig. S3A), providing a reasonable explanation for the observed differences in both the in vivo and in vitro models. Overall, BPD is associated with decreased 3-HAA, which was confirmed to be partly caused by dysregulation of related metabolic enzyme. Fig. 2. [112]Fig. 2 [113]Open in a new tab Dysregulation of related metabolic enzyme led to decreased 3-HAA. (A): IHC staining and the quantitative results in lung tissues of 3-HAA in the control and BPD groups, scale bar = 25 μm. (B): THE expression of KMO, KYNU, and HAAO validated by WB in lung tissues. (C): The quantitative results of KMO, KYNU and HAAO protein expression in lung tissues. The levels of β-actin were used as internal standard control. (D). IHC staining and the quantitative results in lung tissues of KMO in the control and BPD groups, scale bar = 25 μm. (E). IHC staining and the quantitative results in lung tissues of KYNU in the control and BPD groups, scale bar = 25 μm. (F). IIHC staining and the quantitative results in lung tissues of HAAO in the control and BPD groups, scale bar = 25 μm(G): IF staining and the quantitative results in AECII of 3-HAA (red) and DAPI (blue) in the control and hyperoxia groups, scale bar = 60 μm. (H): THE expression of KMO, KYNU, and HAAO validated by WB in AECII lysate. (I): The quantitative results of KMO, KYNU and HAAO protein expression in AECII. The levels of β-actin were used as internal standard control. (J): IF staining and the quantitative results in AECII of KMO in the control and hyperoxia groups, scale bar = 60 μm. (K): IF staining and the quantitative results in AECII of KYNU in the control and hyperoxia groups, scale bar = 60 μm. (J): IF staining and the quantitative results in AECII of HAAO in the control and hyperoxia groups, scale bar = 60 μm. For animal-related experiments, n = 6 per group. n = 3–4 per group. The data are presented as the means ± SD (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001). 3.3. 3-HAA alleviated hyperoxia-induced lung injury in BPD rats We next investigated whether supplementation with 3-HAA could alleviate hyperoxia-induced BPD. Neonatal rats in the BPD + 3-HAA group were nebulized with 3-HAA every other day (P2, P4, P6, P8, P10, P12, and P14) for 30–50 min each time during the 14-day hyperoxia modeling period ([114]Fig. 3A). To explore the appropriate dose and effectiveness of 3-HAA nebulization, newborn rats were randomly divided into eight groups: control, control + DMSO, BPD, BPD + DMSO, BPD+3-HAA (25 mg/kg), BPD+3-HAA (50 mg/kg), BPD+3-HAA (75 mg/kg), and BPD+3-HAA (100 mg/kg). Our results demonstrated that 3-HAA nebulization improved histopathological alterations, as the RAC increased and MLI decreased compared with those of the BPD group ([115]Fig. S4A). In addition, compared with BPD, 3-HAA increased the expression of SPC ([116]Figs. S4B and S4E) and AQP5 ([117]Fig. S4C and S4E), which are specific markers of AECII and AECI, respectively. 3-HAA also decreased the expression of MPO ([118]Figs. S4D and S4E), which can increase oxidative stress and tissue injury. A dose of 75 mg/kg exhibited the best effect and was selected for further experiments. Fig. 3. [119]Fig. 3 [120]Open in a new tab 3-HAA alleviated hyperoxia-induced lung injury in BPD. (A): Schematic representation and timeline of the intervention for the three groups of rats. (B): HE-stained lung tissues and the quantitative results of MLI, scale bar = 25 μm(C): IHC staining of 3-HAA performed on lung tissues and the quantitative results, scale bar = 25 μm. (D): IF staining showed the percentage of SPC + cells relative to all DAPI + cells in the control, BPD and BPD+3-HAA groups per high-power field (HPF), scale bar = 100 μm. (E): THE expression of SPC, MPO, and COX2 validated by WB in lung tissues and the quantitative results. The levels of β-actin were used as internal standard control. For animal-related experiments, n = 6 per group. The data are presented as the means ± SD (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001). In further experiments, we confirmed that 3-HAA nebulization decreased the MLI ([121]Fig. 3B) and effectively increased the 3-HAA level ([122]Fig. 3C). In the present study, 3-HAA partially protected against the reduction in SPC^+ cells after hyperoxia ([123]Fig. 3D). In addition, the percentages of CD68^+ inflammatory macrophages, CD11c^+ dendritic cells and MPO^+ neutrophils in BPD rat lungs were significantly increased [[124]45] when compared with the control group. However, 3-HAA protected myeloid cells from influx to the lung ([125]Figs. S5A–C). The ELISA results revealed that the levels of VEGF increased, whereas the levels of inflammatory cytokines (including TNF-α, IL-6, and TGF-β) in the lung tissues were significantly lower in the BPD + 3-HAA group compared to BPD group ([126]Fig. S3B), which was consistent with the observed mRNA expression patterns ([127]Fig. S3C). At the protein level ([128]Fig. 3E) and mRNA level ([129]Fig. S3D), 3-HAA reduced the expression of MPO and COX2, suggesting an attenuated inflammatory response [[130]46], and increased the expression of SPC, implying an amelioration of AECII damage. Collectively, these findings confirmed that 3-HAA protected against lung damage in a hyperoxia-induced BPD rat model by improving alveolar complexity and suppressing inflammation. 3.4. 3-HAA alleviated ferroptosis in lung tissue caused by hyperoxia-induced BPD To explore the underlying mechanism by which 3-HAA alleviates hyperoxia-induced BPD in neonatal rats, chemical proteomics by the LiP-SMap approach was performed ([131]Fig. 4A). After data preprocessing, 20,519 detected peptides were retained. Differentially expressed peptides were screened according to a p < 0.05 and FC ≤ 0.1 or FC ≥ 10. Further analysis revealed enrichment of a specific form of cell death, known as ferroptosis, which is an iron dependent, lipid peroxidation-driven form of programmed cell death, as indicated by KEGG pathway analysis and Gene Ontology biological process (GO-BP) terms ([132]Fig. 4B and C). Fe^2+ participates in the Fenton reaction, exacerbating lipid peroxidation and cellular damage. Mitochondrial membranes, which are rich in phospholipids, are particularly susceptible to oxidative damage, leading to mitochondrial dysfunction and impaired energy metabolism. Aldehydes such as MDA and 4-HNE are byproducts of lipid peroxidation and are produced when free radicals cause oxidative damage to lipids in cell membranes. We next assessed ferroptosis by evaluating ferroptosis-associated protein and mRNA expression, aldehydes and Fe^2+ concentrations, and mitochondrial morphology. Our results revealed that the contents of total aldehydes, ferrous iron, MDA and 4-HNE were increased in lung tissue because of hyperoxia-induced BPD and were significantly reduced in the 3-HAA + BPD group ([133]Fig. 4D). Furthermore, after 3-HAA treatment, long-chain acyl-CoA synthetase-4 (ACSL4), transferrin (TF), FTH1, NCOA4, and LC3B-II were decreased, whereas GPX4 was increased compared with those in the BPD group ([134]Fig. 4E). The RT‒qPCR results were consistent ([135]Fig. S3E). Mitochondrial alterations in AECII from rat lung tissues were observed using TEM, revealing typical ferroptosis-associated changes, such as mitochondrial pyknosis and loss of cristae, and 3-HAA treatment attenuated these mitochondrial abnormalities ([136]Fig. 4F). Collectively, these findings suggested that 3-HAA inhibited ferroptosis in the hyperoxia-induced BPD rat model, thereby attenuating lung injury. Fig. 4. [137]Fig. 4 [138]Open in a new tab 3-HAA alleviated BPD by inhibiting ferroptosis (A): Flow chart depicting the LiP-SMap assay. (B): Bubble plot shows the KEGG pathway enrichment analysis of the LiP-SMap assay. (C): Bubble plot shows Gene Ontology biological process (GO-BP) terms enriched in the LiP-SMap assay. (D): The content of total aldehydes, MDA, Fe^2+ and 4-HNE in lung tissues. (E): Western blotting of 4-HNE, GPX4, ACSL4, TF, FTH1, NCoA4, LC3B-II and their quantification in lung tissues. The levels of β-actin were used as internal standard control (n = 6). (F): Representative TEM images of mitochondria in AECII of rat lung tissue (x8000). The data are presented as the means ± SD (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001). 3.5. 3-HAA inhibited the hyperoxia-induced ferroptosis pathway in AECII A large amount of evidence suggests that persistent damage to AECII is closely related to the occurrence and development of BPD [[139]47]. The aforementioned in vivo experiments revealed mitochondrial ferroptosis-like changes in AECII from rat lung tissues, so the regulatory mechanism by which 3-HAA affects ferroptosis in AECII in an in vitro model was further confirmed by administering the ferroptosis agonist erastin and the corresponding inhibitor DFO. The optimal concentrations of 3-HAA (140 μmol/L), DFO (75 μmol/L), and erastin (2 μmol/L) were selected by CCK-8 for further experiments ([140]Figs. S6A–F). The results of IF staining of AEC-II ([141]Figs. S6G–H) indicated reduced SPC expression under hyperoxic conditions. Conversely, the increased expression of AQP5 indicates the gradual differentiation of AECII into AECI [[142]48]. While 3-HAA and DFO treatment attenuated the cell transformation induced by hyperoxia, the ferroptosis agonist erastin reversed the 3-HAA response. In the hyperoxia-induced cellular model of AECII, the ROS level measured by the fluorescent probe DCFH-DA ([143]Fig. 5A) and the extent of lipid peroxidation measured by the fluorescence emitted by Liperfluo ([144]Fig. 5B) were significantly elevated compared with those in the control group, and supplementation with 3-HAA and DFO reduced excessive ROS and lipid peroxidation generation, which could be reversed by erastin. A similar pattern was observed for the contents of redox and ferroptosis-associated indicators, such as Fe^2+, total aldehydes, MDA, and H[2]O[2], which increased in the H and HAE groups and decreased following 3-HAA and DFO treatment. In contrast, GSH and SOD levels were decreased in the H and HAE groups but increased in the HA and HAD groups ([145]Fig. 5C–E). The levels of TNF-α and IL-6 in the cell culture supernatant were increased in the hyperoxia group, whereas the levels of inflammatory markers were decreased after 3-HAA and DFO treatment, indicating that 3-HAA and DFO mitigated hyperoxia-induced inflammation. Conversely, erastin counteracted the anti-inflammatory effects of 3-HAA and increased TNF-α and IL-6 levels ([146]Fig. S3F). Compared with those in the H group, the protein levels of FTH1, NCOA4, COX2 and LC3B-II in AECII also decreased, whereas GPX4 expression increased after 3-HAA and DFO treatment ([147]Fig. 5F), which was consistent with the mRNA expression patterns ([148]Fig. S3G). However, the antiferroptotic effect of 3-HAA was reversed by erastin. Mitochondrial shrinkage and increased mitochondrial membrane density [[149]49] were observed in AECII cells due to hyperoxia exposure. Treatment with 3-HAA and DFO attenuated mitochondrial alterations given their crucial role in preventing ferroptosis. In contrast, erastin activated ferroptosis and blocked the protective effects of 3-HAA ([150]Fig. 5G). Collectively, these findings suggested that 3-HAA alleviated hyperoxia-induced ferroptosis in AECII. Fig. 5. [151]Fig. 5 [152]Open in a new tab 3-HAA inhibited the hyperoxia-induced ferroptosis pathway in AECII. (A): Live cell fluorescence imaging of DCFH-DA (green) in AECII and the quantitative results, scale bar = 10 μm. (B): Live cell fluorescence imaging of LiperFluo (green) in AECII and the quantitative results, scale bar = 10 μm. (C): The content of total aldehydes, Fe^2+, MDA, GSH in AECII. (D): The content of SOD in AECII. (E): Extracellular H[2]O[2] levels in cell culture supernatant. (F): THE expression of GPX4, FTH1, LC3BII, NCOA4, and COX2 in AECII validated by WB and the quantitative results. The levels of β-actin were used as internal standard control. (G):TEM images of mitochondria in AECII, scale bar = 1 μm. For the cellular experiments, n = 3 per group. The data are presented as the means ± SD (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001). 3.6. 3-HAA inhibited the ferroptosis pathway by targeting FTH1 Different from some other studies showing a decrease in FTH1 during ferroptosis [[153]50], our results indicated that upregulation of FTH1 was observed during hyperoxia-induced ferroptosis in vivo and in vitro, which guided us to focus on this protein. We reasoned that elevated levels of FTH1 may serve as a protective mechanism against hyperoxia-induced oxidative stress by binding excess iron. However, simultaneously elevated NCOA4 acts as a cargo receptor for ferritin, specifically recognizing and binding to FTH1 and facilitating the autophagic degradation of ferritin [[154]51]. 3-HAA improved this phenomenon. IF staining of AECII revealed that 3-HAA and DFO inhibited ferritinophagy, as indicated by the reduced expression of LC3B, NCOA4, and FTH1, which indicates impaired autophagic degradation of ferritin ([155]Figs. S6I–J). Indeed, by using the LiP-SMap method mentioned above, we identified FTH1 as a potential binder of 3-HAA, as shown in [156]Fig. 6A (with p = 0.04 and log[2] FC = −5.6). SPR was subsequently performed to provide insights into the binding kinetics and molecular affinities between 3-HAA and FTH1. The data indicated that 3-HAA exhibited a strong, dose-dependent affinity for FTH1 (KD = 5.87E-06) ([157]Fig. 6B). Subsequently, we conducted molecular docking to clarify the interaction mode between FTH1 and 3-HAA ([158]Fig. 6C), demonstrating that 3-HAA bonds to SER31 and SER59 through hydrogen bonding (2.0 Å and 2.6 Å). The docking score was −5.1 kcal/mol. Given that the NCOA4-FTH1 interaction is essential for ferritinophagy, we performed SPR to investigate whether 3-HAA alleviated ferritinophagy by disrupting the NCOA4-FTH1 interaction. As shown by SPR, recombinant FTH1 bound the recombinant NCoA4 protein with high affinity ([159]Fig. 6D). However, SPR revealed that 3-HAA was able to prevent FTH1-NCOA4 binding ([160]Fig. 6E). We validated this finding using a Co-IP assay [[161]52]. As shown in [162]Fig. 6F, 3-HAA treatment substantially weakened the interaction between NCOA4 and FTH1 compared to the AECII in the hyperoxia group, as evidenced by decreased FTH1 in the immunoprecipitates pulled down by anti-NCOA4. Collectively, these phenomena indicated that the exogenous administration of 3-HAA successfully alleviated ferroptosis by directly targeting FTH1 and disrupting the NCOA4-FTH1 interaction, thereby inhibiting hyperoxia-induced AECII injury in vitro and in vivo. Fig. 6. [163]Fig. 6 [164]Open in a new tab 3-HAA inhibited the ferroptosis pathway by targeting FTH1 (A): Volcano plot of differentially expressed peptides by LiP-SMap approach. Red indicates an upregulation in the BPD+3-HAA group, and blue shows a downregulation in the BPD+3-HAA group. (B): The binding between human recombinant FTH1 and varying concentrations of 3-HAA was assessed by SPR. (C): The illustrations of molecular docking represent the binding modes of 3-HAA with the human FTH1, depicted as 3D binding and 2D binding views. (D): The binding between human recombinant NCoA4 and varying concentrations of FTH1 was assessed by SPR. (E): Recombinant human NCoA4 was coupled on the chips. The binding between FTH1 and NCoA4 in the presence or not of 3-HAA was assessed by SPR. (F): Co-IP assay of NCOA4 and FTH1 interaction in AECII and corresponding quantitative results. 4. Discussion BPD is the most common chronic lung disease in preterm infants who require respiratory support. The absence of effective prevention and treatment strategies results in higher mortality rates and poorer prognoses in BPD infants than in non-BPD premature infants, imposing heavy economic burden on families and society [[165]53,[166]54]. Understanding the underlying pathogenesis of BPD is crucial for developing targeted interventions [[167]55]. Recent studies have highlighted the role of metabolic dysfunctions in contributing to lung injury caused by hyperoxia [[168]56]. In this study, we aimed to investigate the potential role of tryptophan metabolites in preventing hyperoxia-induced BPD and to explore the potential mechanism involved. Therefore, we analyzed tryptophan metabolic profiles and identified a reduction in 3-HAA in experimental and clinical BPD. Treatment with 3-HAA suppressed inflammation and improved lung development in BPD rats. Our findings indicated that both in vivo and in vitro, 3-HAA modulated the dysregulated expression of ferroptosis-related molecules and mitigated the signs of mitochondrial ferroptosis in hyperoxia-induced models. In addition, molecular docking, SPR and Co-IP assays were performed to confirm that 3-HAA inhibited the ferroptosis pathway by targeting FTH1, thereby alleviating hyperoxia-induced damage in rats and AECII. From a developmental perspective, premature infants who are prone to BPD have distinct metabolic signatures compared with other patient groups. For example, Piersigili [[169]57] reported that the bronchoalveolar lavage fluid (BALF) of BPD infants had relatively high levels of serine, taurine, and citrulline on the first day after birth. Another study showed sphingolipid metabolites in TAs were significantly elevated in BPD infants during the first few days after birth [[170]58], which led to mitochondrial dysfunction [[171]59,[172]60] and abnormal ROS production. These biomarkers of BALF and TAs accurately reflect what is happening in the lungs. Owing to the complexity of BPD pathogenesis, it is necessary to develop sufficient biomarkers to identify premature infants who are susceptible to BPD. Emerging evidence [[173]61] has revealed that tryptophan metabolites are essential for modulating pathophysiological functions such as inflammation, metabolism, and oxidative stress. Interestingly, studies [[174]62,[175]63] suggest that metabolites from tryptophan may significantly influence lung health and modulate immune responses in the respiratory tract, which are critical in conditions like BPD. Our results showed that BPD was associated with dysregulation of tryptophan metabolism and a significant reduction in its metabolite 3-HAA. As discussed earlier, 3-HAA has demonstrated multiple biological activities in different experimental disease models, including anti-inflammatory effects via the inhibition of PI3K/Akt/mTOR activation induced by LPS [[176]64], the ability to modulate immune responses by increasing the binding of NCoA7 and aryl hydrocarbon receptors [[177]65], and the ability to regulate apoptosis in cancer cells by binding to YY1 [[178]66]. Furthermore, 3-HAA significantly facilitates tumor cell escape from ferroptosis [[179]67]. Our study revealed its ability to inhibit hyperoxia-induced ferroptosis by binding to FTH1 and disrupting the NCOA4-FTH1 interaction. In our opinion, 3-HAA can be used as a potential biomarker and potential therapeutic strategy for preventing BPD progression, providing new perspectives on BPD management and treatment. In recent years, ferroptosis, a form of regulated cell death associated with iron accumulation and lipid peroxidation, has gained increasing attention [[180]68,[181]69] because it is closely associated with several diseases. There is increasing evidence that ferroptosis plays an important role in many respiratory disorders [[182]70]. Iron metabolism is crucial for the regulation of ferroptosis. Ferritinophagy [[183]71,[184]72], a specialized autophagic process, breaks down ferritin to release free iron within the cell. This iron can engage in the Fenton reaction, exacerbating lipid peroxidation and cellular damage, key features of ferroptosis. For example, a recent report has shown that cigarette smoke induces labile iron accumulation through ferritinophagy, leading to ferroptosis in lung epithelial cells [[185]73]. The evidence indicates that gene dysregulation associated with ferroptosis and iron accumulation in lung tissues may contribute to BPD [[186][74], [187][75], [188][76]]. Disruption of iron homeostasis and ferritinophagy could lead to increased vulnerability of lung tissues to oxidative stress, exacerbating this condition. Not surprisingly, our study revealed that 3-HAA inhibited hyperoxia-induced ferroptosis by reducing the protein and mRNA expression of ACSL4, TF, FTH1, NCOA4, and LC3B-II while increasing GPX4 expression. The ferroptosis inhibitor DFO attenuated cell damage and ferroptosis, but the ferroptosis agonist erastin reversed the 3-HAA-induced anti-damage response. FTH1 is a key component of the ferritin complex and plays a crucial role in regulating iron storage and the process of ferroptosis, converting toxic ferrous iron (Fe^2+) into its less reactive ferric form (Fe^3+) [[189]77,[190]78]. Elevated levels of FTH1 may serve as a protective mechanism against iron-induced oxidative stress by binding excess iron [[191]79]. However, simultaneous increases in NCOA4 and LC3BII promote ferritinophagy in hyperoxia-induced models. NCOA4 acts as a cargo receptor for ferritin, specifically recognizing and binding to FTH1, facilitating the autophagic degradation of ferritin [[192]51]. When ferritin is tagged for degradation, LC3BII helps encapsulate the autophagic cargo, promoting the fusion of autophagosomes with lysosomes for degradation and subsequent free iron release, leading to the Fenton reaction and cellular damage. Indeed, a combination of LiP-SMap, molecular docking, SPR and Co-IP analyses confirmed that 3-HAA can bind directly to FTH1 and disrupt the NCOA4-FTH1 interaction. This study discovered for the first time that 3-HAA effectively inhibited the ferroptosis pathway by targeting FTH1, thereby alleviating hyperoxia-induced injury in vitro and in vivo. AECII cells have the ability to self-renew and differentiate into AECI cells, which is crucial for alveolar structure recovery and maintenance following lung injury [[193]80]. In the context of BPD, the disruption of alveolar maturation may stem from impaired proliferation and transition of AECIIs. Reports have shown varying results regarding the changes in AECII in experimental BPD lungs. Our immunofluorescence analysis of SPC in lung tissue revealed that the proportion of SPC^+ cells was reduced in the BPD group. Hyperoxia-induced in vivo and in vitro models demonstrated that ferroptosis and a reduction in 3-HAA in AECII were closely related to the progression of BPD. These findings could be crucial for the development of targeted therapeutic strategies against BPD in which ferroptosis of AECII plays a critical role. Though 3-HAA protected myeloid cells such as CD68^+ inflammatory macrophages, CD11c^+ dendritic cells and MPO^+ neutrophils from influx to the lung, whether 3-HAA can similarly inhibit ferroptosis in these cell types in the lung has not been explored, and further studies are needed to clarify these mechanisms. There are some limitations in our current study. Firstly, the clinical part is a single-center, small sample size exploratory study aimed at screening potential biomarkers. It has been challenging to find suitable infants for controls, and the collection of an effective amount of TA samples has been difficult, resulting in limited ability to balance the groups [[194]81]. There are differences in gestational age and duration of antibiotic use between the BPD and non-BPD infants. However, no significant differences were observed between the groups in terms of birth weight, postmenstrual age and weight at the time of TA sample collection, which helps control for some potential confounding factors. Moreover, we found no correlation between 3-HAA levels and the duration of antibiotic use in the correlation analysis among tryptophan metabolites and clinical characteristics. Then we have validated the differences and effects of 3-HAA using experimental models, partially explored the role of 3-HAA in the pathogenesis of BPD, reflecting true biological effects. These multidimensional strategies significantly compensated for the limitations in the sample size of the clinical preliminary exploratory study. Future studies will involve prospective cohorts to further investigate 3-HAA as a potential biomarker for the clinical diagnosis of BPD. Increasing the sample size and conducting subgroup analysis based on gestational age helps mitigate the impact of baseline differences. Secondly, we established an in vitro hyperoxia-induced model using immortalized human AECII. However, it is important to note that these cells may not fully replicate the characteristics of primary human AECII. Despite these limitations, this study provides new insights into the mechanisms of BPD development and interventions. 5. Conclusions Our study is the first to reveal that BPD is associated with significantly reduced 3-HAA levels, indicating dysregulation of the tryptophan metabolic pathway. Supplementation with 3-HAA inhibits the ferroptosis pathway by targeting FTH1, thereby alleviating hyperoxia-induced injury in rats and AECII. This work highlights the potential of targeting 3-HAA and ferroptosis for clinical applications and provides new insights into the management and treatment of BPD. CRediT authorship contribution statement Qiqi Ruan: Writing – original draft, Visualization, Resources, Methodology, Funding acquisition, Formal analysis, Conceptualization. Yingqiu Peng: Writing – review & editing, Methodology, Formal analysis. Xuanyu Yi: Writing – review & editing, Methodology, Formal analysis. Jingli Yang: Writing – review & editing, Resources. Qing Ai: Resources, Data curation. Xiaochen Liu: Writing – review & editing, Methodology. Yu He: Writing – review & editing, Resources, Project administration, Funding acquisition, Conceptualization. Yuan Shi: Writing – review & editing, Resources, Project administration, Funding acquisition, Conceptualization. Authors’ consent for publication All authors of the manuscript titled “The tryptophan metabolite 3-hydroxyanthranilic acid alleviates hyperoxia-induced bronchopulmonary dysplasia via inhibiting ferroptosis” agree to submit it for publication in Redox Biology. We hereby grant editors the right to edit the manuscript and publish it. We confirm that this manuscript is an original work, has not been published before, and is not under consideration for publication elsewhere. We further confirm that all authors have contributed significantly to the manuscript. We have reviewed the final version of the manuscript and approved it for publication. Ethics approval The Institutional Review Board of CHCMU approved the clinical research section (2023-258), which was registered on [195]chictr.org (ChiCTR2400087152). The Animal Ethics Committee of CHCMU approved the animal experiment protocol (No. CHCMU-IACUC20240412007) which complied with the [196]ARRIVE guidelines. Funding This work was supported by the National Key Research and Development Program of China (No. 2022YFC2704803), the National Natural Science Foundation of China (No. 82001602), Clinical Research Project for the Summit Program of Children's Hospital of Chongqing Medical University (CHCMU-2024-XKDF-1002), Key Projects for Technological Innovation and Application Development of Chongqing (CSTC2021jscx-gksb-N0015), Special Funding for Postdoctoral Research Projects of Chongqing (No. 2022CQBSHTB3085), China Postdoctoral Science Foundation (No. 2023MD744152), Natural Science Foundation of Chongqing (No. CSTB2024NSCQ-MSX0491), and Chongqing Graduate Research Innovation Project (No. CYB23211). Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Acknowledgments