Abstract Microplastics (MPs) are widespread environmental pollutants that can enter the human body through the food chain, potentially leading to lung damage. However, the underlying mechanisms responsible for this damage remain unclear. Ducks, a commonly consumed poultry species in China, are particularly susceptible to MPs exposure due to their farming environment. In this study, Shaoxing ducklings were administered two distinct concentrations of polystyrene microplastics (PS-MPs) (1 mg/L and 100 mg/L) via oral route, alongside a control group, over a period of four weeks to establish an in vivo model for evaluating the effects of microplastic exposure in ducks. Simultaneously, rat type II alveolar epithelial (RLE-6TN) cells were exposed to different concentrations of PS-MPs (0, 10, 100, and 500 µg/mL) for 48 h, thereby constructing an in vitro exposure model. Our results showed that PS-MPs caused pathological damage, inflammatory cell infiltration, and activation of the LPS/TLR4 inflammatory pathway in the lung. Further analysis revealed that PS-MPs disrupted the tricarboxylic acid (TCA) cycle and inhibited oxidative phosphorylation. Mechanistic investigation demonstrated that PS-MPs induced mitochondrial dysfunction and consequent excessive mitophagy. This study investigates the mechanisms by which PS-MPs contribute to mitochondrial dysfunction and mitophagy, potentially exacerbating lung inflammation, offering valuable insights for mitigating the toxic effects of PS-MPs on human and animal health. Graphical Abstract [38]graphic file with name 12951_2025_3503_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03503-x. Keywords: Duck, Polystyrene microplastics, Mitochondrion, TCA cycle, Mitophagy Introduction The use of plastic products significantly improves the quality of human life. However, the convenience of “white garbage” has led to widespread plastic waste, raising global concerns about serious environmental issues [[39]1, [40]2]. Microplastics (MPs), defined as particles with diameters smaller than 5 mm, are a common component of plastic materials [[41]3]. MPs are particularly concerned about their extensive distribution, strong adsorption properties, and resistance to degradation, making them a growing threat to both health and environment [[42]4, [43]5]. Ingestion of MPs has been increasingly reported across various animal species, where these particles accumulate in multiple organs such as the liver, heart, nervous system, and intestines, causing hepatotoxicity, cardiotoxicity, neurotoxicity, and gastrointestinal toxicity [[44]6–[45]10]. Among these species, ducks are especially vulnerable due to their feeding habits and the contamination of aquatic environments with MPs. Subsequently, ducks contaminated with MPs could enter the human diet, resulting in the bioaccumulation of MPs in the human body. By ingesting MPs from polluted water and food sources, these particles accumulate in various organs, especially the lungs, where they can cause severe damage. Despite the growing awareness of microplastic pollution, research into the molecular mechanisms underlying the toxic effects of MPs accumulation in the lungs remains limited. Mitochondria play a key role in cellular energy metabolism, redox balance, and programmed cell death [[46]11, [47]12]. Moreover, it is also the key regulator of inflammatory responses, driven by both its structural components and the diverse metabolic products it generates [[48]13]. Under the condition of dysfunction, mitochondria display fragmentation and membrane depolarization, leading to the production of large amounts of reactive oxygen species (ROS) and activating the mitochondrial cell death pathway [[49]14]. To counteract this damage, cells initiate mitophagy, a specialized autophagy that selectively degrades damaged, dysfunctional or surplus mitochondria, thereby safeguarding cellular homeostasis [[50]15]. Excessive accumulation of ROS in the mitochondria can trigger abnormal mitophagy, which ultimately results in cell death and contributes to the development of diseases [[51]16]. Remarkably, the accumulation of MPs in the body can cause oxidative damage to the heart, liver, and skeletal muscle, resulting in significant toxic effects [[52]9, [53]17, [54]18]. However, whether mitochondrial dysfunction triggers mitophagy in MPs-induced lung injury remains unclear. We hypothesize that MPs-induced lung damage occurs via a pathogenic cascade in which mitochondrial dysfunction triggers ROS overproduction, dysregulating mitophagy and perpetuating a self-amplifying cycle of oxidative stress and chronic inflammation. In the mitochondrion, the TCA cycle serves as a pivotal metabolic pathway that plays a critical role in regulating mitochondrial function and managing oxidative stress in eukaryotic cells [[55]19]. The metabolites in the TCA cycle are widely recognized for their roles in energy metabolism, hypoxic response, and immunity regulation [[56]19]. Numerous observations suggest that TCA cycle disruption is closely linked to mitochondrial dysfunction, promoting chain-dependent ROS production and ultimately leading to cell death [[57]20–[58]22]. However, it remains unclear whether TCA cycle imbalance occurs under MPs exposure and contributes to MPs-induced lung cell death. Consequently, there is a strong interest in exploring whether MPs exposure disrupts TCA cycle balance, then accelerating mitochondrial dysfunction in the lung. Ducks, as a species of waterfowl, inhabit aquatic environments such as rivers and ponds, accessing water for drinking and sourcing food primarily from zooplankton and fish. Consequently, ducks exhibit a higher risk of exposure to and accumulation of MPs in comparison to other poultry species. To better understand the potential mechanism behind the toxic effect of PS-MPs, a duck model for PS-MPs (5 μm) was used to illustrate their effect on lung injury. We conducted metabolomics, and succinylomics to explore the hazards of PS-MPs exposure in the lung. Additionally, transmission electron microscopy (TEM), flow cytometry, oxygen consumption rate (OCR) analysis, immunofluorescence, qPCR, and western blot were utilized to determine the molecular regulatory mechanism involved in PS-MPs-exposed lung injury both in vivo and in vitro. These results provide new theories for comprehensively understanding the mechanisms of toxicity caused by MPs in birds and mammals. Results PS-MPs caused lung injury in ducks To investigate the toxicity of PS-MPs exposure to the lung, we detected the distribution of PS-MPs in lung tissues by scanning electron microscopy (SEM). We found that PS-MPs were widely distributed throughout the lung tissues with increasing doses of PS-MPs treatment (Fig. [59]1A–C). Compared to the control (CK) group, PS-MPs were significantly detected in lung tissues of exposed ducks, confirming the pulmonary deposition of microplastics. HE staining revealed dose-dependent structural lung injury, with exposed groups exhibiting obvious vascular congestion (characterized by erythrocyte infiltration into pulmonary capillaries) and dense inflammatory cell infiltration (Fig. [60]1D–F). These findings establish a direct correlation between PS-MPs accumulation and escalating pulmonary damage, underscoring the pathological consequences of microplastic exposure in ducks. Fig. 1. [61]Fig. 1 [62]Open in a new tab PS-MPs distributed in the lung and triggered lung injury in ducks. A–C SEM examination (scale bar = 50 μm). Yellow arrows point to PS-MPs. D–F HE staining (scale bar = 400 μm and 100 μm, respectively) PS-MPs exacerbated lung inflammation through the LPS/TLR4 pathway We explored the molecular pathways modification underlying inflammation induced by PS-MPs. PS-MPs markedly elevated LPS levels in the lung tissues of ducks compared to the CK group (p < 0.05, Fig. [63]2A), with almost two fold enhancement under high level PS-MPs exposure. Notably, higher TLR4 and MyD88 levels were observed in PS-MPs group, indicating that LPS overproduction in the lungs activated the TLR4 pathway and increased the phosphorylation of NF-κB and IκBα (p < 0.05, Fig. [64]2B). The lung tissues of ducks exhibited a notable elevation in the mRNA expression levels of MyD88, NF-κB, and IκBα in response to low and high doses of PS-MPs, in a dose-dependent manner (p < 0.05, Fig. [65]2C). Additionally, PS-MPs markedly elevated the release of IL-1β, IL-6, and TNF-α (p < 0.05, Fig. [66]2D–F) and promoted the protein expression level of pro-IL-1β and TNF-α (p < 0.05, Fig. [67]2G). Immunofluorescence (IF) analysis of lung sections revealed elevated TNF-α expression levels in PS-MPs-exposed ducks (Fig. [68]2H). Similarly, both mRNA and protein levels indicated that PS-MPs induced the expression of inflammatory mediators iNOS and COX2 in the lungs (p < 0.05, Fig. S1). These results showed that PS-MPs exacerbated lung damage by activating LPS/TLR4 pathway. Fig. 2. [69]Fig. 2 [70]Open in a new tab PS-MPs caused LPS overproduction and inflammatory response in the lung of ducks. A LPS levels (n = 10). B The relative expression levels of TLR4-related proteins (TLR4, MyD88, p-NF-κB, NF-κB, p-IκBα and IκBα) (n = 3). C The relative mRNA expression levels of TLR4-related genes (MyD88, NF-κB and IκBα) (n = 6). D-F The levels of IL-1β, IL-6 and TNF-α in the lung (n = 10). G The relative protein expression levels of pro-IL-1β and TNF-α (n = 3). H IF staining for TNF-α in the lung (scale bar = 50 μm). Different letters (a-c) indicate statistically significant differences (p < 0.05) The metabolite profile of lung tissues indicated that PS-MPs potentially induced mitochondrial dysfunction Given the crucial role of LPS/TLR4 activation in lung injury with or without PS-MPs, non-targeted metabolic profiles of the lungs were conducted. In the positive mode, 820 metabolites were identified, while 422 metabolites were identified in the negative mode across all samples. To better understand the metabolic differences in the lung tissues based on PS-MPs exposure, principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were performed. PCA results showed that the PC1 explained 20.4% of the variations in positive mode and 17.1% in negative mode (Fig. [71]3A). OPLS-DA score plot analysis further confirmed the clear separations between lung tissues from the CK and HMPs groups in both positive mode and negative mode (Fig. [72]3B). Meanwhile, the explained variation (R2Y) and prediction capability (Q2) values in both modes were > 0.9 and > 0.7, respectively, indicating the reliability of the model’s results (Fig. S2A). Fig. 3. [73]Fig. 3 [74]Open in a new tab PS-MPs altered the composition of metabolites of the lung in ducks. A The PCA plot in positive and negative mode. B The OPLS-DA score plot in positive and negative mode. C Volcano plot of the differentially expressed metabolites in positive and negative modes. D KEGG pathway in positive and negative mode. E The common pathways contribute to the difference of energy metabolism in PS-MPs-exposed ducks. Red dot indicated upregulated metabolites, green dot indicated downregulated metabolites To identify differential metabolites in the lungs after PS-MPs exposure, we set a variable importance in projection (VIP) score > 1 and p < 0.05 as thresholds. Compared to the CK group, 30 upregulated and 292 downregulated metabolites were screened in the positive mode, while 12 upregulated and 136 downregulated metabolites were observed in the negative mode (Fig. [75]3C, Fig. S2B, C). Functional enrichment analysis revealed that the affected metabolic pathways were primarily involved in amino acids biosynthesis, alanine, aspartate and glutamate metabolism, pyruvate metabolism and arginine biosynthesis (Fig. [76]3D). Notably, L-lactate, S-lactoylglutathione, phosphoenol pyruvate, succinate exhibited a notable decrease in response to high doses of PS-MPs, while L-glutamate represented an elevation in high PS-MPs group (Fig. [77]3E, Fig. S2D). These metabolites highlighted a key metabolic pathway network impacting pyruvate metabolism (downregulation in L-lactate and S-lactoylglutathione), glycolysis (downregulation in phosphoenol pyruvate), TCA cycle and oxidative phosphorylation (downregulation in succinate), arginine biosynthesis (upregulation in L-glutamate) following PS-MPs exposure (Fig. [78]3E, Fig. S2D). These findings suggested that PS-MPs exposure could disrupt mitochondrial metabolism. PS-MPs disrupted tricarboxylic acid cycle (TCA) cycle metabolism To determine whether PS-MPs exposure directly affected TCA cycle metabolism, we conducted an absolute quantitative metabolomic analysis in lung tissues (Fig. [79]4A). Interestingly, we found that PS-MPs markedly reduced the abundance of key TCA cycle intermediates, including pyruvate, α-ketoglutarate, succinate, fumarate, malate, and oxaloacetate, indicating significant impairment of the TCA cycle (Fig. [80]4B, [81]C). This disruption aligned with concurrent reductions in energy-related metabolites (ATP, cyclic AMP, GTP, GDP) and redox cofactors (NADP^+, NAD^+), collectively pointing to profound dysfunction in oxidative phosphorylation (Fig. [82]4B, [83]C). These results indicated that PS-MPs exposure disrupted the balance of the TCA cycle and oxidative phosphorylation metabolism. Fig. 4. [84]Fig. 4 [85]Open in a new tab The effect of PS-MPs exposure on the TCA cycle and oxidative phosphorylation metabolism in ducks. A Schematic diagram of TCA cycle. B Heatmap of differential metabolites. C The levels of metabolites in the TCA cycle and oxidative phosphorylation. *p < 0.05 vs CK group Succinylomics analysis revealed that TCA cycle-related proteins were involved in response to PS-MPs exposure To further explore whether PS-MPs could disrupt the TCA cycle and contribute to lung damage, we analyzed protein succinylation of lung tissues treated with or without PS-MPs. We identified 1489 succinylation sites across 521 proteins, with 1376 sites and 458 proteins being quantifiable (p < 0.05, Fig. [86]5A, Table S1). Among these, 67 succinylated proteins were localized to mitochondria (Fig. [87]5B, Table S2). Comparison between HMPs and CK group revealed that 13 succinylation sites across 13 proteins were significantly upregulated, while 51 succinylation sites across 44 proteins were significantly downregulated (Fig. [88]5C, Table S3). Notably, 12 out of the 57 differentially succinylated proteins were localized to mitochondria (Fig. [89]5D, Table S4). Fig. 5. [90]Fig. 5 [91]Open in a new tab Succinylation of the TCA cycle-related protein was involved in the PS-MPs-exposed lung injury. A The numbers of succinylated sites and the corresponding proteins detected in PS-MPs-exposed lung tissues. B The distribution of succinylated proteins. C The numbers of differential succinylated sites and the corresponding differential proteins. D The distribution of differential succinylated proteins. E KEGG, GO-BP, and GO-MF analysis of differential succinylated proteins. F The differential succinylated sites and the corresponding proteins in the TCA cycle. Red dot indicated significantly upregulated succinylated sites. Green dot indicated significantly downregulated succinylated sites. G Succinylated profiling identified differential succinylated sites of TCA cycle-related proteins (IDH1 and MDH2). H Pattern diagram of the succinylated sites of IDH1 and MDH2 Pathway enrichment analysis, including Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology-Biological Process (GO-BP), and GO-Molecular Function (MF), showed significant enrichments in the process such as oxaloacetate metabolism, fumarate metabolism, NAD^+ activity, and ADP binding (Fig. [92]5E, Tables S5, S6). Among the differentially succinylated proteins, we observed that the succinylation levels of IDH1 at lysine 245 (K245) and MDH2 at lysine 204 (K204) were significantly decreased, whereas the succinylation of MDH2 at lysine 182 (K182) was significantly increased following PS-MPs treatment (Fig. [93]5F–H). This evidence highlighted the potential of PS-MPs to impair the mitochondrial function and contributed to lung damage through alterations in protein succinylation within the TCA cycle. PS-MPs triggered mitochondrial dysfunction and oxidative stress Based on these results, we inferred that PS-MPs exposure could impair lung mitochondrial function. To explore this, we assayed the ATP levels and mtDNA copies in the lungs under PS-MPs exposure. Compared with CK ducks, the lungs of PS-MPs-treated ducks showed a significant decrease in ATP production with a dose-dependent manner (p < 0.05, Fig. [94]6A) and mtDNA levels were also decreased in PS-MPs-treated lungs compared to CK lung (p < 0.05, Fig. [95]6B). Then, we observed the mitochondrial morphology by TEM. The lungs of the PS-MPs ducks displayed significant mitochondrial abnormalities including swelling and broken or absent cristae (Fig. [96]6C). We also analyzed the expression of key mitochondrial regulators at both the protein and mRNA levels. PS-MPs ducks markedly inhibited the SIRT1, SIRT3, and TFAM levels (p < 0.05, Fig. [97]6D, [98]E). Furthermore, we investigated the expression of markers associated with mitochondrial dynamics. The OPA1, MFN1, and MFN2 levels were significantly downregulated (p < 0.05, Fig. [99]6F, [100]G). To assess the extent of oxidative stress, we detected the ROS level and antioxidant enzyme activity. PS-MPs markedly elevated the ROS and MDA levels, while the activities of SOD and CAT were substantially reduced compared to the CK group (p < 0.05, Fig. [101]6H–K). These results indicated that excessive PS-MPs exacerbated mitochondrial damage and oxidative stress response in the duck lungs. Fig. 6. [102]Fig. 6 [103]Open in a new tab PS-MPs caused the mitochondrial function damage. A ATP levels (n = 10). B mtDNA copy number levels (n = 6). C TEM examination (scale bar = 500 nm and 200 nm, respectively). D The relative protein expression levels of SIRT1, SIRT3, and TFAM (n = 3). E The relative mRNA expression levels of SIRT1, SIRT3, and TFAM (n = 6). F The relative protein expression levels of OPA1, MFN1, and MFN2 (n = 3). G The relative mRNA expression levels of OPA1, MFN1, and MFN2 (n = 6). H ROS levels (n = 10). I MDA levels (n = 10). J, K The activities of SOD and CAT (n = 10). Different letters (a-c) indicate statistically significant differences (p < 0.05) PS-MPs enhanced mitochondrial damage and led to ROS overproduction in RLE-6TN cells To further explore the toxicity of PS-MPs on mitochondrial damage in vitro, RLE-6TN cells were utilized as our model. First, the excessive PS-MPs on the mitochondrial abundance were assessed. Interestingly, PS-MPs treatment (100 and 500 μg/mL PS-MPs) significantly reduced mtDNA copy numbers in RLE-6TN cells (p < 0.05, Fig. [104]7A). In addition, PS-MPs exposure decreased mitochondrial respiratory capacity, indicating lower basal and maximal respiration levels, ATP production across different PS-MPs concentrations (p < 0.05, Fig. [105]7B). A reduction in mitochondrial membrane potential (MMP) was also observed in cells exposed to high doses of PS-MPs, consistent with an increase in ROS production (p < 0.05, Fig. [106]7C, [107]D). We further examined PS-MPs on the mitochondrial function in RLE-6TN cells. As shown in Fig. [108]7E and [109]F, high dose of PS-MPs significantly reduced the protein expression of the mitochondrial regulator (SIRT1, SIRT3, and TFAM) as well as mitochondrial fusion markers (OPA1, MFN1, and MFN2) (p < 0.05). Overall, our findings suggested that PS-MPs induced mitochondrial dysfunction and triggered ROS overproduction in vitro, exacerbating oxidative stress in RLE-6TN cells. Fig. 7. [110]Fig. 7 [111]Open in a new tab PS-MPs impaired the mitochondrial function and activated ROS production in RLE-6TN cells. A mtDNA copy number levels (n = 3). B Measurement of mitochondrial oxygen consumption ratio (OCR). C Measurement of mitochondrial membrane potential (MMP). D ROS levels (n = 3). E The relative protein expression levels of SIRT1, SIRT3, and TFAM (n = 3). F The relative protein expression levels of OPA1, MFN1 and MFN2 (n = 3). Different letters (a-d) indicate statistically significant differences (p < 0.05) PS-MPs induced mitophagy both in vivo and in vitro Mitophagy is responsible for removing damaged mitochondria. Thereby, we wondered whether mitophagy was activated by PS-MPs. Indeed, we observed that both protein and mRNA levels of LC3, ATG7, and BECN1 in lung tissues were significantly increased, while p62 was obviously decreased with increasing doses of PS-MPs exposure (p < 0.05, Fig. [112]8A, [113]B). These findings were further supported by IF staining of LC3 and p62 (Fig. [114]8C, [115]D). Additionally, the expression levels of Parkin showed a concentration-dependent increase (p < 0.05, Fig. [116]8E, [117]F). Fig. 8. Fig. 8 [118]Open in a new tab PS-MPs mediated mitophagy. A The relative protein expression levels of LC3, ATG7, BECN1, and p62 (n = 3). B The relative mRNA expression levels of LC3, ATG7, BECN1 and p62 (n = 6). C, D IF staining for LC3 and p62 in the lung (scale bar = 50 μm). E The relative protein expression levels of Parkin (n = 3). F The relative mRNA expression levels of Parkin (n = 6). Different letters (a-c) indicate statistically significant differences (p < 0.05) We then examined the effect of PS-MPs on mitophagy in vitro. As shown in Fig. [119]9A, we found the high dose of PS-MPs treatment contributed to an obvious increase in the protein levels of LC3, ATG7, and BECN1, alongside a reduction in p62 level, consistent with the IF staining results (p < 0.05, Fig. [120]9B, [121]C). Meanwhile, high dose of PS-MPs treatment significantly elevated Parkin protein level (p < 0.05, Fig. [122]9D). TUNEL assay was further determined that PS-MPs-exposed RLE-6TN cells directly led to cell death, which was markedly decreased by Mdivi-1 (p < 0.05, Fig.S3A). In addition, RLE-6TN cells exposed to PS-MPs presented much higher level of cleaved caspase 3 compared to untreated RLE-6TN cells, which was reversed by Mdivi-1 (p < 0.05, Fig. S3B). Collectively, our data suggested that increased mitophagy occurred under PS-MPs-induced mitochondrial dysfunction. Fig. 9. [123]Fig. 9 [124]Open in a new tab PS-MPs aggravated mitophagy in RLE-6TN cells. A The relative protein expression levels of LC3, ATG7, BECN1, and p62 (n = 3). B, C IF staining for LC3 and p62 in RLE-6TN cells (scale bar = 50 μm). D The relative protein expression levels of Parkin (n = 3). Different letters (a-d) indicate statistically significant differences (p < 0.05) Discussion Microplastic residues in water and farmland have emerged as potential health risks and were also identified and accumulated within the lung tissues in both human and animals [[125]23–[126]25]. As waterfowl, ducks inhabit rivers and ponds, drink from these water sources. This lifestyle makes ducks more prone to encounter and ingest microplastics compared to other poultry species. In our study, we found that PS-MPs exposure to duck could accumulate in the lungs, leading to distinct pathological changes, including blood permeation into pulmonary capillaries and infiltration of inflammatory cells (Fig. [127]1D–F). Additionally, we observed an increase in LPS levels (Fig. [128]2A). LPS triggers inflammatory responses by binding to TLR4, activating the adaptor MyD88, and initiating NF-κB signaling pathways, which in turn, boost the release of critical pro-inflammatory mediators [[129]26, [130]27]. Consistent with these findings, we observed the upregulation of TLR4, MyD88, phosphorylated NF-κB, and phosphorylated IκBα in the lung tissues, resulting in elevating TNF-α, IL-6, and IL-1β (Fig. [131]2B–H). Our findings confirmed that PS-MPs exacerbate lung damage by activating the LPS/TLR4 pathway. Notably, previous studies have shown that MPs disrupt intestinal barrier integrity via the NF-κB/NLRP3/IL-1β/MLCK pathway and compromise reproductive function via the miR-199a-5p/HIF-1α and CNR1/CRBN/YY1/CYP2E1 signaling axes [[132]28, [133]29]. Together, these findings underscore the systemic toxicity of PS-MPs across multiple organ systems, with our study specifically elucidating the lung damage. Given the evolutionary conservation of inflammatory and stress-response pathways, these results raise critical concerns about the potential human health risks posed by microplastic pollution, particularly in populations with prolonged environmental or dietary exposure. Given the crucial role in regulating inflammatory responses utilizing multiple signals, that can drive chronic inflammation-associated diseases, such as periodontitis and osteoarthritis, in response to cell stress and loss of homeostasis [[134]30], we wondered whether mitochondria could act as potential inducers contributing to lung inflammation. Strikingly, exposure to PS-MPs led to significant impairment in critical mitochondrial pathways, notably the TCA cycle and OXPHOS system, as evidenced by pathway activity profiles (Fig. [135]3E). Absolute quantitative metabolomics analysis further corroborated these findings, demonstrating pronounced reductions in key metabolites associated with both the TCA cycle and OXPHOS system (Fig. [136]4B, [137]C). These results collectively confirm a systemic disruption in mitochondrial energy metabolism, compromising cellular energy production. Moreover, succinylomics analysis revealed that key enzymes of the TCA cycle were markedly modified in the PS-MPs-exposed group (Fig. [138]5F–H). Previous studies also showed that environmental hazards can disrupt the TCA cycle, leading to proteotoxic stress [[139]31]. Besides, disruption of the TCA cycle triggers mitochondrial damage by activating retrograde signaling pathways, which communicate metabolic dysfunction and induce profound metabolic reprogramming [[140]32, [141]33]. These findings strongly suggest that mitochondrial function is severely compromised under PS-MPs exposure. Mitochondria is widely recognized as the center of energy metabolism, playing an important role in ATP production and maintaining normal cellular functions [[142]34]. We assessed ATP levels in both tissual and cellular samples, and as expected, ATP production was significantly reduced (Figs. [143]6A, [144]7B), directly indicating mitochondrial dysfunction in response to PS-MPs exposure. This is consistent with observed mitochondrial morphological changes, such as swelling and disrupting or absent cristae in the PS-MPs group (Fig. [145]6C). Next, we explored whether the self-protection mechanisms including mitochondrial biogenesis and fusion were activated to preserve the mitochondrial function. However, mitochondrial biogenesis was significantly impaired, as evidenced by the notable reduction in TFAM protein levels. Additionally, we observed significant downregulation of OPA1, MFN1, and MFN2 proteins, essential for mitochondrial fusion. SIRT3, which promotes mitochondrial fusion by deacetylating OPA1 [[146]35], was also downregulated, further suggesting that mitochondrial fusion was inhibited (Fig. [147]6D, [148]F). Mitochondrial fusion is critical for maintaining the mitochondrial function, as it helps dilute damaged mitochondrial DNA, redistribute metabolic intermediates, and improve mitochondrial respiration [[149]36]. However, in this case, these self-repair pathways were disrupted, indicating that PS-MPs caused profound mitochondrial dysfunction. This prompted investigation into whether oxidative stress threshold has been exceeded. Our hypothesis was supported by significant increases in ROS production and MDA levels (Fig. [150]6H, [151]I). ROS, a critical mediator of stress-related signaling pathways, directly reflects cellular oxidative stress. Similarly, elevated MDA, a hallmark of lipid peroxidation, correlates with ROS overproduction. The marked rise in both biomarkers signifies severe oxidative damage, aligning with prior studies where PS-MPs induced ROS accumulation in ovarian granulosa cells and colon tissues [[152]37, [153]38]. Notably, key antioxidant enzymes, such as superoxide dismutase (SOD) and catalase (CAT), were significantly suppressed under PS-MP exposure, diverging from earlier findings [[154]39]. These enzymes are essential for maintaining redox balance by scavenging ROS, and their depletion likely exacerbates oxidative injury [[155]40–[156]42]. Combine with the increased ROS and MDA levels, along with decreased antioxidant activity (e.g., SOD and CAT) (Fig. [157]6H–K). This evidence showed that mitochondrial dysfunction promotes the accumulation of ROS, amplifies oxidative stress, and impairs antioxidant defenses. Critically, pre-treatment with the antioxidant N-acetylcysteine (NAC) or oxidative stress inhibitors AM251 substantially attenuated ROS generation and inflammatory cascades, alleviating PS-MP-induced toxicity [[158]38, [159]43]. These findings collectively identify oxidative stress as a central mediator of PS-MP toxicity, linking mitochondrial impairment to systemic cellular damage. In addition to impair mitochondrial biogenesis and reduce mitochondrial fusion, we observed a significant reduction in mtDNA copies (Figs. [160]6B, [161]7A), a direct marker of mitochondrial abundance. Therefore, it makes sense to anticipate that damaged mitochondria will be removed by the process of mitophagy, which is the activation of a quality control mechanism. To keep the mitochondria intact, mitophagy is necessary because it maintains the proper ratio of healthy to damage mitochondria, hence regulating the quality of mitochondria [[162]44]. A prior study demonstrated that mitophagy is intricately associated with the generation of ROS [[163]45]. Furthermore, the ROS-mediated mitophagy induced by PS-MPs contributes to kidney damage [[164]46]. In our study, we systematically investigated whether mitophagy occurred in the lungs following PS-MPs exposure. We found that ATG7 and BECN1 were significantly upregulated. BECN1 is a key component of autophagy, involved in the formation, extension, and maturation of autophagosomes [[165]47, [166]48]. Additionally, the LC3 II/LC3 I ratio, a well-established marker of mitophagy, was significantly increased under PS-MPs treatment, along with a notable downregulation of p62. It was reported that cell-specific p62 ablation led to excessive IL-1β production, which was associated with hypersensitivity to endotoxin-induced shock [[167]49]. Activated mitophagy could reduce p62 levels as well [[168]50]. Meanwhile, IF staining consistently confirmed the upregulation of LC3 and the downregulation of p62. Furthermore, we observed a significant upregulation of Parkin, a protein closely involved in mitophagy [[169]51]. At the cellular level, these findings were further validated in RLE-6TN cells. Collectively, our results demonstrated strong activation of mitophagy in response to PS-MPs exposure-induced mitochondrial dysfunction. While mitophagy is necessary to preserve the quality of the mitochondria, abnormal or excessive mitophagy can cause an excess of ROS in the mitochondria, which can lead to cell death [[170]52]. Notably, pharmacological inhibition of autophagy partially relieves PS-MP-induced mitochondrial and lysosomal damage [[171]37], suggesting that PS-MPs drives pathological mitophagy, disrupting mitochondrial homeostasis and exacerbating lung inflammation in ducks. In addition, this evidence showed that MPs penetrated the lungs to reduce the viability of lung cells by inducing oxidative stress and causing cell death [[172]53]. However, to address current biological limitations, future studies should integrate primary duck lung cells with RLE-6TN cell models to further elucidate the mechanisms underlying PS-MP-induced mitochondrial dysfunction. Furthermore, the dose-dependent effects were evident: low-dose PS-MPs exposure caused less prominent histopathological changes, oxidative stress, and mitophagy activation compared to high-dose groups. Intriguingly, despite high-dose PS-MPs exposure, ducks exhibited no obvious clinical symptoms and maintained normal survival rates, raising concerns about potential human consumption of PS-MPs-contaminated poultry products. Previous studies suggested that MPs caused serve damage and inflammation in poultry products, such as heart, liver, muscle and intestine [[173]10, [174]54, [175]55]. While such meat may appear safe, PS-MPs bioaccumulation in tissues could pose long-term risks to human health through dietary exposure. Notably, NAC, an effective antioxidant, markedly reduced polystyrene nanoplastics (PS-NPs)-triggered oxidative damage in lung epithelial cells by restoring mitochondrial function and inhibiting cell death [[176]56, [177]57], as well as Mdivi-1 [[178]58]. Although antioxidants (e.g., NAC) and autophagy inhibitors (e.g., Mdivi-1) show therapeutic potential in mitigating plastics toxicity, these interventions were not explored in the current study. Nevertheless, our mechanistic insights into the mitochondrial dysfunction-inflammatory axis provide a foundation for developing targeted strategies to counteract microplastic-related health risks in humans. Taken together, our findings verified that PS-MPs disrupted mitochondrial homeostasis, leading to excessive mitophagy and LPS/TLR4 pathway activation, which potentially contributed to lung injury. Our study provides new insights into the mechanism behind the damage in the lung, highlighting the critical role of mitochondrial dysfunction under PS-MPs exposure. Future research should aim to explore the effects of PS-MPs on other organ systems and investigate the specific mechanisms and potential therapeutic strategies by which PS-MPs interacts with cellular and tissue responses. Materials and methods Preparation of animals and experimental design PS-MPs (Unibead, 5 μm in size) were obtained from Tianjin BaseLine Scientific Co., Ltd. (Tianjin, China). The characterization of PS-MPs was measured using electron microscopy (Zeiss, Germany) and SEM (Fig. S4). One-day-old Shaoxing ducklings (n = 60), procured from Hanchao Poultry Company in Hangzhou, China, were randomly assigned to three experimental groups, each comprising 20 individuals: (1) The control group (CK) received standard drinking ultrapure water without PS-MPs; (2) the low-dose group (LMPs) was administered drinking ultrapure water containing a concentration of 1 mg/L of PS-MPs; and while (3) the high-dose group (HMPs) received drinking ultrapure water with a concentration of 100 mg/L of PS-MPs. The exposure doses of PS-MPs were selected based on previous toxicological studies of PS-MPs in mice and poultry [[179]9, [180]18]. In these previous studies, 1 mg/L, 10 mg/L and 100 mg/L were used to feed the chicken [[181]9]. Considering the previous research reported the 1 mg/L PS-MPs is an environment-related concentration [[182]59, [183]60]. Therefore, we chose moderate doses of 1 mg/L and 100 mg/L for this study. After one week of the adaptation period, the ducklings were maintained on these treatments for four weeks. During the experimental period, ducks were raised in cage, and kept at room temperature with flowing air. The ducks were fed ad libitum with the same formula diet and subjected to a standard light regimen of 17 h light (17L:7D). All experimental ducks were healthy and were not administered any antibiotic treatments during the experiment. At five weeks old, the animals were euthanized (approved number: 2024ZAASLA019), lung tissues were collected and stored in 4% formaldehyde or at −80°C for further analysis. Cell culture RLE-6TN cells were obtained from Wanwu Biotechnology Co., Ltd (Anhui, China). Cells were cultured in Ham’s F-12K medium supplemented with 10% FBS (Gibco), 1% ITS, and 5 ng/mL EGF at 37 °C with 5% CO[2]. Upon achieving 50% confluency, the cells were treated with various concentrations of PS-MPs (0, 10, 100, 500 μg/mL) for 48 h [[184]18]. In addition, Mdivi-1 (1 μM, Selleck, USA) was administered on 100 μg/mL PS-MPs-treated RLE-6TN cells for 24 h. Oxidative stress detection Lung malondialdehyde (MDA), catalase (CAT), and superoxide dismutase (SOD) were measured (n = 10). Briefly, a total of 0.1 g of lung tissue was homogenized in normal saline and centrifuged to collect the supernatant. The levels of MDA and the activities of CAT and SOD were then detected following the manufacturer’s protocols (Solarbio, Beijing, China). Inflammatory cytokine and LPS assays A total of 0.1 g of lung tissue was homogenized and centrifuged to collect the supernatant. The IL-1β, IL-6, and TNF-α levels (n = 10) in the tissue were then measured according to the manufacturer’s instructions (mlbio, Shanghai, China). The LPS levels (n = 10) were quantified from Beijing Sinouk Institute of Biological Technology (Beijing, China). Reactive oxygen species (ROS) level detection A total of 0.05 g of lung tissue was homogenized and centrifuged to obtain the supernatant. Then, the supernatant was mixed with 50 μM 2,7-dichlorofluorescein diacetate (DCFH-DA) solution and incubated at 37 °C for 30 min. The fluorescence intensity was measured (n = 10) using a microplate reader (Tecan, Switzerland). Cells were incubated with 5 μM CellROX (Thermo, USA) for 30 min, washed twice with PBS, and the intracellular ROS levels (n = 3) were measured by flow cytometry (Beckman, USA). ATP content measurement Lung tissue was lysed and centrifuged to collect the supernatant. The ATP content in the supernatant was measured (n = 10) according to the manufacturer’s instructions (Thermo, MA, USA). Hematoxylin and eosin (H&E) staining Lung tissues were fixed in 4% polyformaldehyde overnight and embedded in paraffin wax. Then the tissues were sectioned into 5-μm slices and stained with hematoxylin and eosin (H&E). After decolorization in alcohol, the stained sections were examined under an upright light microscope (Olympus, Japan). PS-MPs distribution detection Lung tissues were fixed in 2.5% glutaraldehyde at 4 ℃ overnight. Then, the samples were fixed in a 1% osmium tetroxide (pH 7.4), dehydrated with ethanol solutions, and plated onto gold in a vacuum coating device. The PS-MPs in the lung were observed with SEM (SU8010; Hitachi, Japan). Mitochondrial morphology observation Lung tissues (≈1 mm^3) were fixed in 2.5% glutaraldehyde at 4 ℃ overnight. The tissues were then post-fixed with 1% osmium tetroxide, dehydrated, and embedded in epoxy resin, followed by staining with 2% uranyl acetate. Images of the sections were captured by TEM (HT7650; Hitachi, Japan). Detection of the gene expression level by qPCR qPCR was performed on a LightCycle 96 Real-Time System (Roche, Switzerland) using the TB Green^® Premix Ex Taq (TAKARA, Tokyo, Japan). Reaction conditions included one cycle of 95 °C for 30 s, 45 cycles of 95 °C for 5 s and 60 °C for 20 s. GAPDH was employed as the housekeeping gene, and relative expression levels were calculated using the 2^−ΔΔCt method [[185]61]. The gene primer sequences are detailed in Table S7 and Fig. S5. Mitochondrial DNA (mtDNA) copies detection Relative levels of mtDNA copy number were determined as described by Eaton et al. [[186]62]. Briefly, total DNA was extracted from all tissues or cells using the QIAamp DNA Mini Kit (Qiagen, Germany). A total of 2 ng DNA was used for qPCR analysis. For duck lung tissues (n = 6), primers targeting the mitochondrial COX1 (mtCOX1) gene were designed to assess mtDNA copy number, with the 18 s rRNA gene serving for normalization. For RLE-6TN cells (n = 3), primers for the mitochondrial ND5 (mtND5) gene were used to measure the mtDNA copy number, with the Rpl13 gene used for normalization. The primer sequences for mtDNA detection are provided in Table S8 and Fig. S6. Protein expression level detection Tissues and cell lines were lysed using RIPA buffer (adding PMSF and phosphatase inhibitor). Protein concentrations were detected using the BCA protein assay kit (Beyotime, China). The methods followed in this study were based on our previous work [[187]54]. The band of protein is detected by exposing an X-ray film to the membrane. The information on primary antibodies is shown in Table S9. Immunofluorescence (IF) staining For the in vivo assay, lung tissues were fixed in 4% polyformaldehyde, embedded in paraffin, and sectioned into 5-μm slices. For the in vitro assay, cells were fixed with 4% polyformaldehyde and permeabilized with 0.5% Triton X-100. Both the tissue sections and cells were blocked with 5% bovine serum and incubated with primary antibodies against LC3, p62, and TNF-α overnight. Afterward, samples were incubated with FITC-conjugated IgG (H + L) for 1 h. DAPI was used for nuclear staining. Fluorescence images were acquired using a microscope (Zeiss, Germany). Details of the primary antibodies are provided in Table S10. Oxygen consumption rate (OCR) assays OCR was measured using Seahorse XFe96 Extracellular Flux Analyzer (Agilent, USA) [[188]63]. Briefly, a total of 1 × 10^4 cells treated with different concentration of PS-MPs (n = 3) were seeded into the XF96 cell culture microplate and incubated overnight. Then, the culture medium was replaced with assay buffer, prepared from XF base medium supplemented with 10 mM D-glucose, 2 mM sodium pyruvate, and 4 mM L-glutamine. The OCR was measured following the sequential addition of oligomycin (2 μM), FCCP (0.5 μM), and rotenone/antimycin A (1 μM each). Data acquisition and analysis were performed using Seahorse XF96 Wave software. TUNEL staining Apoptotic cells were detected using an In Situ Cell Death Detection Kit according to its manufacturer’s instructions (Roche, Germany). Briefly, the cells were seeded in 12-well plate, fixed with 4% polyformaldehyde and permeabilized with 0.5% Triton X-100. Then, TUNEL reaction mixture was added to incubated cells for 1 h at 37 °C in a light-resistant container. DAPI was used for nuclear staining. Fluorescence images were acquired using a microscope (Zeiss, Germany). Non-targeted metabolomic analysis Lung tissues from CK (n = 10) and HMPs (n = 10) ducks were prepared for non-targeted metabolomics analysis as described by Li et al. [[189]64]. Briefly, samples were extracted with methanol and centrifuged to obtain the supernatant. The supernatants were then freeze-dried under vacuum and re-dissolved in 80% methanol solution containing 2-chlorobenzalanine. Metabolic profiling was performed using liquid chromatography-mass spectrometry (LC–MS) on an ACQUITY UPLC^® HSS T3 column (Thermo, USA). Absolute quantification of TCA cycle and oxidative phosphorylation intermediates based on MRM method Lung tissues from CK (n = 10) and HMPs (n = 10) ducks were prepared for targeted metabolomics analysis according to Zhang et al. [[190]65]. Briefly, the samples were processed for metabolite extraction, followed by separation of metabolites using ultra-high performance liquid chromatography (UHPLC; Agilent). Mass spectrometry analysis was performed using an AB SCIEX 5500 QTRAP mass spectrometer (USA). Quantitative succinylomics detection Lung tissues from CK (n = 3) and HMPs (n = 3) ducks were collected for succinylomic analysis according to Liu et al. [[191]66]. Briefly, the lung tissues were lysed with lysis buffer. Proteins were precipitated with trichloroacetic acid, washed with pre-cooled acetone, and redissolved in triethylammonium bicarbonate (TEAB). The proteins were then digested, reduced, alkylated, and enriched using anti-succinyllysine antibody-conjugated agarose beads (PTM-402, PTM BIO) for LC–MS/MS analysis (timsTOF HT, Bruker, Germany). Tandem mass spectra were searched against the Anas platyrhynchos Uniprot database used to search tandem mass spectra. Upregulated and downregulated proteins/sites were identified with fold changes > 1.5 or < 0.67, respectively. Functional annotations and pathway enrichment of differentially succinylated proteins were analyzed using GO and KEGG analysis. Subcellular localization was predicted using Wolfpsort software. Statistical analysis The results of this study are reported as mean ± standard deviation (SD), and graphical representations are generated using GraphPad Prism 8.0 software (CA, USA). The comparisons in different concentration of PS-MPs in vitro and in vivo were fulfilled using one-way ANOVA with Tukey tests and statistical comparisons of muti-omics between CK and HMPs groups were performed using a t-test analysis. The p value cutoff was set as 0.05. Supplementary Information [192]Additional file 1.^ (2.1MB, docx) [193]Additional file 2.^ (3.1MB, xlsx) [194]Additional file 3.^ (371.5KB, xlsx) [195]Additional file 4.^ (97.4KB, xlsx) [196]Additional file 5.^ (24KB, xlsx) [197]Additional file 6.^ (15.1KB, xlsx) [198]Additional file 7.^ (18.9KB, xlsx) [199]Additional file 8.^ (18.6KB, docx) [200]Additional file 9.^ (17.2KB, docx) [201]Additional file 10.^ (20.2KB, docx) [202]Additional file 11.^ (16.4KB, docx) Author contributions T.T.G., and M.H.K. carried a majority of experiments and analyzed data. T.T.G., and M.H.K. wrote the main manuscript. T.T.G., and M.H.K. prepared figures. M.C.D. and L.C. provided support for animal experiment. T.T.G. and Y.T. analyzed the bioinformatic data. T.T.G. and W.W.X. performed the statistical analysis. T.T.G., M.H.K., T.Z. and L.Z.L. conceived the study, designed the study, evaluated data, and revised the manuscript. All authors read and approved the final manuscript. Funding This work was supported by the Key R&D Program of Zhejiang Province (2024C02004), Zhejiang Provincial Natural Science Foundation of China (LQ23C170001), National Natural Science Foundation of China (32372862), and China Agriculture Research System of MOF and MARA (CARS-42–6). Availability of data and materials No datasets were generated or analysed during the current study. Declarations Ethics approval and consent to participate All procedures were approved by the Zhejiang Academy of Agricultural Sciences Ethics Committee (Approved number: 2024ZAASLA019). Consent for publication All authors have agreed to publish this manuscript. Competing interests The authors declare no competing interests. Footnotes Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Tiantian Gu and Minghua Kong contributed equally to this work. Contributor Information Tiantian Gu, Email: gtt19931029@126.com. Lizhi Lu, Email: lulizhibox@163.com. References