Abstract The restoration of cardiac function post-myocardial infarction (MI) remains a significant clinical challenge. Emerging evidence indicates that Goji berries (“Gouqi” in Chinese) and their extracts exhibit substantial cardioprotective properties. Here, we introduce fibrin gel-loaded Gouqi-derived nanovesicles (GqDNVs-gel) as a delivery system targeted at the infarcted myocardium. The application of GqDNVs-gel resulted in a marked improvement in survival rates over a 14-day period post-MI, enhanced cardiac function, reduced infarct size, myocardial apoptosis, and excessive fibrosis, and facilitated endogenous repair. Through a combination of transcriptomics and proteomics analyses, alongside in vitro and in vivo experiments, we identified that the cardioprotective effect of GqDNVs are mediated through the inhibition of the p38 MAPK-NF-κB p65 signaling pathway. Furthermore, GqDNVs contain abundant bioactive compounds, including proteins, genetic materials, lipids, polysaccharides, and flavonoids. GqDNVs-gel intervention can reshape the post-MI cardiac environment and modulate myocardial lipid metabolism, specifically impacting glycerophospholipid and α-linolenic acid metabolic pathways. The upregulation of the peptide Arg-Thr-Ile-Glu and the downregulation of phosphatidylethanolamine in the hearts of MI mice after GqDNVs-gel intervention may play crucial roles in modulating the associated metabolic pathways. This study is the first to highlight the multifaceted therapeutic effects of GqDNVs-gel, offering a promising strategy for enhancing cardiac function post-MI. Graphical abstract [52]graphic file with name 12951_2025_3615_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-025-03615-4. Keywords: Myocardial infarction, Cardiac patch, Goji berry, Exosome, Plant-derived nanovesicle Introduction According to the latest heart disease and stroke statistics, the incidence and mortality rates of myocardial infarction (MI) among adults in the United States in 2020 were 9,300,000 (3.2%) and 109,199, respectively [[53]1]. Despite substantial advancements in acute MI management, restoring long-term cardiac function remains a major challenge [[54]2]. This difficulty arises from irreversible damage to myocardial cells and the formation of scar tissue during the reparative process. Notably, approximately 13.6% of acute MI patients die within 30 days [[55]1], and a staggering 10-year post-MI heart failure mortality rate of 58.9% persists [[56]3]. Consequently, regenerative medicine, which focuses research efforts on enhancing post-MI prognosis by promoting myocardial regeneration and repair, has become a remarkable research frontier. Mammalian-derived nanovesicles (NVs) for cardiac repair post-MI have been extensively investigated; however, their clinical application is constrained by suboptimal targeting, inherent immunogenicity, and potential infection risk [[57]4]. Meanwhile, plant-derived NVs offer many therapeutic properties and may serve as a viable alternative due to their low immunogenicity, biocompatibility, and easier production and storage [[58]5, [59]6]. Emerging evidence suggests that plant-derived NVs exhibit remarkable cardioprotective effects through their antioxidant and immunomodulatory activities [[60]7, [61]8]. Nowadays, there is a growing interest in goji berries (Lycium barbarum L., “Gou qi” in Chinese), which have been used in traditional Chinese medicine for over 2000 years and are acknowledged for their dual role in medicine and nutrition [[62]9–[63]11]. Several clinical studies have indicated that goji berries and their extracts can reduce cardiovascular disease (CVD) risk factors, including blood lipids, pressure, and glucose [[64]12, [65]13]. Experimental investigations have shown that Lycium barbarum polysaccharides (LBPs) can confer protection against myocardial injury in vivo [[66]14–[67]19] and inhibit doxorubicin and H[2]O[2]-induced cardiotoxicity [[68]20, [69]21]. Recently, nanoparticles derived from LBPs were found to protect retinal ganglion cells from ferroptosis following ischemia–reperfusion injury [[70]22]. Our previous research highlighted the effectiveness of GqDNVs in mitigating muscle atrophy [[71]23]. Nevertheless, the potential application of GqDNVs in cardiac repair remains unexplored. Direct injection into infarcted regions leads to rapid clearance due to the short plasma half-life of NVs [[72]24]. As an adjunctive therapy post-reperfusion, cardiac patches have garnered significant attention for their potential to enhance MI prognosis [[73]24, [74]25]. Fibrin gel, a natural biomaterial approved by the FDA, exhibits excellent biocompatibility and facilitates cell adhesion and proliferation [[75]26–[76]28]. Consequently, we have developed a cardiac patch embedded with GqDNVs, culminating in the creation of the GqDNVs-fibrin gel (GqDNVs-gel). This study aims to elucidate the mechanisms underlying the therapeutic effects of GqDNVs in the treatment of MI in mouse models by seeding them into fibrin gel. We will assess their efficacy in myocardial repair and regeneration, and explore their potential applications in the long-term management of MI as an adjunctive therapy following reperfusion. Materials and methods Isolation and characterization of GqDNVs Briefly, GqDNVs were purified from fresh goji berries (Lycium barbarum L., obtained from Zhongwei, Ningxia, via cold-chain transportation) via a sucrose density gradient centrifugation method following our laboratory’s previous publication [[77]23]. GqDNVs were then suspended in phosphate-buffered saline (PBS) and filtered through a 0.22 μm filter to remove impurities (Fig. [78]1a). Before use, the size and concentration of GqDNVs were examined by nanoparticle tracking analysis (NTA) via a NanoSight NS300 (Malvern Instruments Ltd., UK). The morphology of the GqDNVs was visualized via transmission electron microscopy (TEM, HT-7700, Hitachi, Japan). The contents of GqDNVs were subsequently detected via untargeted and saccharides-targeted metabolomic sequencing via the UPLC–MS/MS and the GC–MS platform, respectively. Fig. 1. [79]Fig. 1 [80]Open in a new tab Preparation and characterization of GqDNVs and GqDNVs-gel. a GqDNVs were isolated and purified using sucrose density gradient ultracentrifugation. b The distribution of particle size and concentration of GqDNVs was analyzed. c A scatter plot showing the intensity distribution of GqDNVs. d Representative transmission electron microscopy image showing the morphology of GqDNVs. e GqDNVs content was analysed by non-targeted metabolome sequencing. f The saccharides content of GqDNVs was assessed through saccharides-targeted metabolome sequencing. g Scanning electron micrograph of the fibrin gel. h GqDNVs-gel preparation process. i Scanning electron microscopy reveals GqDNVs encapsulated within the fibrin gel. White arrows indicate encapsulated GqDNVs. GqDNVs: Gouqi-derived nanovesicles, GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel Generation of GqDNVs-gel GqDNVs-fibrin gel was prepared according to previously published methods for exosome-loaded fibrin [[81]29, [82]30]. A defined amount of GqDNVs was added to the fibrinogen solution and mixed with thrombin to form GqDNVs-gel. GqDNVs were integrated into the fibrinogen solution by mixing a 10 mg/mL fibrinogen solution (dissolved in PBS) with a 1 × 10^10 particles/mL GqDNVs solution (dissolved in PBS). Then, the GqDNVs-rich fibrinogen solution and thrombin solution were mixed to quickly form a stable gel patch. The morphologies of the fibrin gel and GqDNVs-gel were characterized via the Scanning electron microscope (SEM, HITACHI S4800). Cell culture HL-1 cells (SCC065) were obtained from EMD Millipore, Shanghai, China. The cells were cultured in minimum essential medium (MEM) supplemented with 10% fetal bovine serum (Pricella Life Science & Technology Co., Ltd, China) at 37 °C in a 5% CO[2] humidified atmosphere. Hypoxia was induced using the AnaeroPack System (Mitsubishi Gas Chemical Co., Ltd) and glucose-free Earle’s buffer (Gibco). Dehydrocorydaline chloride (DHC) (300 nM, MedChemExpress, China) [[83]31] and SB 203580 (SB) (10 μM, MedChemExpress, China) [[84]32] were used to activate and inhibit the activity of p38 MAPK, respectively. Cardiomyocyte biocompatibility with GqDNVs-gel To evaluate the impact of each component of the GqDNVs-gel on cardiomyocytes, HL-1 cells were seeded in 96-well plates and cultured for 24 h. Subsequently, the medium was replaced with fresh medium containing 5 µL of either fibrin gel, GqDNVs solution, or GqDNVs-gel, ensuring that the GqDNVs concentration was consistent across the GqDNVs solution and GqDNVs-gel. Following a 12-h co-incubated period, the viability of the HL-1 cells was assessed using the Cell Counting Kit-8 (CCK-8, Beyotime, China) assay to determine the cytotoxicity associated with each component of the GqDNVs-gel. GqDNVs uptake assay To evaluate the uptake of GqDNVs by cardiomyocytes, we initially labelled GqDNVs with PKH26 (MedChemExpress, China) and subsequently prepared a GqDNVs-gel. HL-1 cells were co-incubated with PBS, PKH26-prelabelled GqDNVs, or PKH26-prelabelled GqDNVs-gel for 6 h. The cytoskeleton and nucleus of the cells were stained with Phalloidin (MaoKang, China) and 4’,6-diamidino-2-phenylindole (DAPI) (Beyotime, China), respectively. The samples were subsequently analysed using confocal fluorescence microscopy (FV3000, OMTOOLS, China). In vivo and ex vivo imaging of GqDNVs distribution in mouse hearts To assess the feasibility of using GqDNVs-gel for the delivery of GqDNVs, we administered an equivalent dose of DiR (AAT Bioquest, USA)-prelabelled GqDNVs via a myocardial GqDNVs drop, GqDNVs injection, or GqDNVs-gel to the hearts of C57BL/6 mice. At 2-, 12-, and 24-h post-administration, the distribution of GqDNVs in live mice and their hearts was evaluated using the Lago X optical imaging systems (SI Imaging, USA). The fluorescence intensities within the region of interest (ROI) were quantified using Aura imaging software (SI Imaging, USA). Mouse model of MI Eight-week-old male C57BL/6 mice were procured from Vital River Laboratories in Beijing, China. Following international standards, all animal care and experiments were approved by the Institutional Animal Care and Use Committee, Huazhong University of Science and Technology (IACUC Number: 3626). After one week of adaptive feeding, an acute MI model was established in the mice following protocols detailed in prior studies [[85]33, [86]34]. In summary, ischemia was induced through permanent ligation of the left anterior descending coronary artery. Post-MI, the mice received either PBS or GqDNVs-gel (containing 1 × 10^8 particles of GqDNVs), with 35 mice in each group. Cardiac echocardiography A blinded veterinary cardiologist performed transthoracic echocardiography at 4 h and 14 days post-MI using the VisualSonics Vevo 1100 system (FUJIFILM, China). Mice from each experimental group were randomly selected and anesthetized using a 1–3% isoflurane mixture in 95% oxygen. Parasternal left ventricular long-axis images were subsequently recorded. M-mode echocardiography was employed to assess various cardiac parameters, including left ventricular (LV) mass, left ventricular ejection fraction (LVEF), left ventricular short-axis fractional shortening (LVFS), left ventricular end-diastolic volume (LVEdV), left ventricular end-systolic volume (LVEsV), heart rate, QRS duration, and cardiac output. Heart histology Following euthanasia, the hearts were excised and processed for paraffin embedding according to established procedures. For hematoxylin–eosin (HE) staining, hearts were sliced into 4 μm-thick sections and stained with the HE staining kit (G1005, Servicebio). For Masson’s staining, hearts from the apex to the ligation level were sectioned at a thickness of 5 μm and stained with a Masson’s staining kit (G1006, Servicebio). Microscopic images were captured, and morphometric parameters, including infarct size and infarct wall thickness, were quantified using ImageJ software. Immunofluorescence Immunofluorescence was performed on paraffin-embedded heart cryosections and HL-1 cells. Heart cryosections were blocked in 3% BSA at room temperature for 30 min, then incubated with a mixture of primary antibodies against rabbit anti-p38 (phospho T180 + Y182) (1:100 dilution, ab195049, Abcam) and mouse anti-alpha sarcomeric actin (α-SMA) (1:200, 67,735-1-Ig, Proteintech), mouse anti-CD31 (1:200 dilution, GB12063, Servicebio), rabbit anti-proliferation marker protein Ki-67 (Ki67) (1:100 dilution, [87]GB111499, Servicebio), and mouse anti-proliferating cell nuclear antigen (PCNA) (1:100 dilution, GB12010, Servicebio) at 4 °C overnight. After washing, the sections were incubated with appropriate secondary antibodies (goat anti-rabbit IgG (Alexa Fluor 488, GB25303, Servicebio) or goat anti-Mouse IgG (Alexa Fluor 594, 115–585-003, Jackson) at room temperature for 60 min, followed by counterstaining with DAPI (G1012, Servicebio) for 10 min in the darkroom. HL-1 cells inoculated in 12-well plates after the indicated treatments were fixed with 4% paraformaldehyde for 10 min. After washing, HL-1 cells were co-incubated with 0.1% Triton and 5% BSA for 2 h at room temperature, followed by incubation with a mixture of primary antibodies against rabbit anti-p38 (phospho T180 + Y182) (1:100 dilution, ab195049, Abcam), rabbit anti-Phospho-NF-κB p65 (1:100 dilution, AP1294, ABclonal), rabbit anti- Ki-67 (1:100 dilution, [88]GB111499, Servicebio), and mouse anti- PCNA (1:100 dilution, GB12010, Servicebio) at 4 °C overnight. Goat anti-rabbit IgG (Alexa Fluor 594, ab150080, Abcam) was used as the secondary antibody for one hour of incubation at room temperature. Subsequently, F-actin was stained with FITC-labelled phalloidin (1:1000 dilution, MX4404, Maokangbio) for 30 min at room temperature. After washing, the sections were incubated with appropriate secondary antibodies (goat anti-rabbit IgG (Alexa Fluor 488, GB25303, Servicebio) or goat anti-mouse IgG (Alexa Fluor 594, 115-585-003, Jackson)) at room temperature for 60 min, followed by counterstaining with DAPI (G1012, Servicebio) for 10 min in the darkroom. For terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) staining, heart cryosections or HL-1 cells were incubated using the TMR (red) TUNEL Cell Apoptosis Detection Kit (G1502, Servicebio). Following washing, DAPI (G1012, Servicebio) was used for nuclear staining. All washes (3 × 3 min) between steps were performed in PBS. All images were taken with a confocal fluorescence microscope (FV3000, OMTOOLS, China). Quantitative real-time polymerase chain reaction (q-PCR) Total RNA extraction was carried out using the RNA Easy Fast (DP451, TIANGEN) following the manufacturer’s instructions. Reverse transcription was performed using the FastKing gDNA Dispelling RT SuperMix (KR118, TIANGEN). The q-PCR was performed with the FastKing OneStep RT-PCR Kit (KR123, TIANGEN). Relative gene expression was obtained and analysed with a Quant Studio 7 Flex Real-Time PCR system (Applied Biosystems Thermo Fisher Scientific). Western blotting Total protein was extracted from the infarcted region of heart tissue. A 10% SDS–polyacrylamide gel was used to separate the target proteins under a constant voltage of 150 V for electrophoresis. Subsequently, the target proteins were transferred onto a nitrocellulose (NC) membrane, which was then blocked with a universal blocking solution for 15 min. The NC membrane was incubated with the appropriate primary antibodies overnight on a shaker at 4 °C: p38 (phospho T180 + Y182) (ab170099, Abcam), p38 alpha/MAPK14 (ab195049, Abcam), Phospho-ERK1/2 (Thr202/Tyr204) (AF1015, Affinity Biosciences), ERK1/2 (AF0155, Affinity Biosciences), Phospho-JNK1/2/3 (Thr183 + Tyr185) (AF3318, Affinity Biosciences), JNK1/2/3 (Thr183 + Tyr185) (AF6318, Affinity Biosciences), Phospho-NF-κB p65 (AP1294, ABclonal), NF-κB p65 (A19653, ABclonal), Cleaved Caspase-7 (8438S, Cell signaling), Caspase-7 (12827S, Cell signaling), Cleaved Caspase-3 (9664S, Cell signaling), Caspase-3 (9662S, Cell signaling), Bax (ET1603-34, Huabio), Bcl-2 (3498S, Cell signaling), and GAPDH (GB15004, Servicebio). On the second day, the NC membrane was treated with corresponding secondary antibodies for one hour at room temperature: Anti-rabbit IgG, HRP-linked Antibody (7074, Cell Signaling), and Anti-mouse IgG, HRP-linked Antibody (7076, Cell signaling). The targeted protein expression levels were calculated by ImageJ and normalized to GAPDH. Enzyme-linked immunosorbent assay (ELISA) Serum samples were collected after centrifuging the coagulated blood at 3000g for 15 min. The levels of transforming growth factor beta 2 (TGFβ-2, E-EL-M1191, Elabscience Biotechnology Co., Ltd, China), insulin-like growth factor 1 (IGF-1, E-EL-M3006, Elabscience Biotechnology Co., Ltd, China), and vascular endothelial growth factor A (VEGF-A, E-MSEL-M0005, Elabscience Biotechnology Co., Ltd, China) in the serum were measured according to the manufacturer's instructions. Transcriptome RNA sequencing and analysis A total amount of l μg RNA per sample of heart tissue was used as input material for the RNA sample preparations. The sequencing libraries were prepared using the NEBNextRUltraTMRNA Library Prep Kit for Illumina (NEB, USA). After cluster generation by the TruSeq PE Cluster Kit v3-cBot-HS (Illumina), the library preparations were sequenced on an Illumina platform. Gene alignment was calculated using featureCounts. The enrichment analysis is performed based on the hypergeometric test. A heat map of differentially expressed genes was generated using the online tool Morpheus. Genes whose fold change (FC) ≥ 2 or ≤ 0.5, variable importance in projection (VIP) > 1, and P-value ≤ 0.05 were considered significantly different. Differential expression analysis between the two groups was performed by the DESeq2 package. Gene set enrichment analysis (GSEA) was performed using GSEA v3.0 software (Broad Institute). All genes included in the DESeq2 output were mapped to HUGO Gene Nomenclature Committee symbols and ranked according to the following: − log[10] (padj) for upregulated genes and (− 1) ×  − log[10](padj) for downregulated genes. Gene Ontology biological processes were obtained from the Molecular Signatures Database. GSEA was run in classic prerank mode with 1000 permutations to assess the false discovery rate (FDR). A weighted enrichment score was used, and the gene set size was limited to 15 to 500 genes. Gene sets with an FDR < 0.05 were defined as significantly enriched. Proteomic analysis The 4D-data-independent acquisition (DIA) quantitative proteomics analysis utilized the diaPASEF acquisition mode of the timsTOF Pro2 series mass spectrometer (MS) to achieve differential quantitative proteomic analysis. The samples were subjected to a series of processes, including protein extraction, enzyme digestion, liquid chromatography-mass spectrometry (LC–MS) tandem analysis, database retrieval analysis, and bioinformatics analysis. The MS raw data were analyzed using DIA-NN (v1.8.1) with the library-free method. The DIA data were reanalysed using this spectral library to obtain protein quantification. After standardization, protein differences were quantitatively analysed using FC and t-test, and the corresponding P values were calculated. FC ≥ 2 or FC ≤ 0.5, VIP > 1, and P-value ≤ 0.05 were defined as significantly different proteins. Metabolomics analysis Metabolites were separated by an ultra-performance liquid chromatograph (LC-30A, Shimadzu, Japan) and identified by a mass spectrometer (TripleTOF 6600 + , SCIEX, USA). Peaks with a missing rate > 50% were excluded, with blank values imputed by the KNN method, and peak area was corrected using the SVR method. An Agilent 8890 gas chromatograph coupled to a 5977B mass spectrometer with a DB-5MS column (J&W Scientific, USA) was employed for GC–MS analysis of saccharides. Unsupervised principal component analysis (PCA) was performed by the statistics function prcomp. The hierarchical cluster analysis results of samples and metabolites are presented as heatmaps with dendrograms, and Pearson correlation coefficients between samples were calculated. Significantly regulated metabolites between groups were determined by the FC. The identified metabolites were annotated using the KEGG compound database ([89]http://www.kegg.jp/kegg/compound/), and the annotated metabolites were then mapped to the KEGG Pathway database ([90]http://www.kegg.jp/kegg/pathway.html). Statistical analysis The data were processed and analysed using GraphPad Prism v.8.0.2 (GraphPad). For the comparison of two datasets, Student’s t-test was used. In comparative analyses involving multiple groups, one-way ANOVA was employed, followed by Tukey’s multiple comparisons test to assess significant differences among datasets. The Mantel test was used to evaluate correlations between omics and between omics and outcome measures, with a correlation coefficient ≥ 0.4 and a P-value < 0.05 considered a significant correlation. Data are presented as mean ± standard deviation unless otherwise specified, and a P-value < 0.05 was set to determine statistical significance. Results and discussion Preparation and characterization of GqDNVs-gel The separation and extraction methods for GqDNVs followed our laboratory’s previous publication [[91]23], with a detailed description provided in Fig. [92]1a. Briefly, a suspension of GqDNVs was prepared via the sucrose density gradient centrifugation method. The NTA results revealed that the concentration of the prepared GqDNVs solution was approximately 1 × 10^10 particles/mL, with an average particle size of 146.0 ± 0.8 nm (Fig. [93]1b, c). TEM analysis further confirmed that the GqDNVs exhibited a characteristic cup-shaped morphology (Fig. [94]1d). Goji berries are known to contain numerous bioactive compounds, including unique LBPs, phenolic acids, and flavonoids [[95]11]. To elucidate the substances presented in the isolated GqDNVs, metabolomic sequencing analysis was conducted. A total of 564 metabolites were identified within GqDNVs through nontarget metabolomic sequencing, with the five most prevalent substances being free fatty acids (11%), amino acids and their derivatives (10%), phenolic acids (8%), saccharides (7%), and alkaloids (7%) (Fig. [96]1e). LBPs are complex mixtures of highly branched and partially characterized polysaccharides and proteoglycans [[97]35] and are considered the most valuable functional compounds in goji berries [[98]35–[99]37]. About 5–8% of polysaccharides are found in dried goji berries [[100]38], which is similar to the saccharide content detected in our isolated GqDNVs. Thus, we further determined the content of saccharides in GqDNVs by saccharides-targeted metabolomic sequencing. Fifteen distinct saccharides were identified, with the most prevalent being sucrose (1.24 ± 0.16 mg/mL), glucose (0.15 ± 0.04 mg/mL), and D-Fructose (0.14 ± 0.02 mg/mL) (Fig. [101]1f & Table S1). According to existing literature, LBPs predominantly comprise glucose and xylose with smaller quantities of arabinose, rhamnose, mannose, and galactose [[102]39], all of which have also been detected in GqDNVs. SEM displayed a three-dimensional network microstructure of the fibrin gel (Fig. [103]1g). The gelation process is shown in Fig. [104]1h. SEM images revealed that the fibrin gel effectively encapsulated the GqDNVs (Fig. [105]1i), corroborating findings from other studies on exosome-based fibrin gels [[106]28, [107]40]. GqDNVs-gel is feasible to deliver GqDNVs to MI hearts in mice To evaluate GqDNVs uptake, we prelabeled them with PKH26 [[108]41] before gel preparation. Then we treated HL-1 cells with the equal volumes of either PBS, GqDNVs in PBS, or GqDNVs-gel (Fig. [109]2a). We observed the uptake of GqDNVs by HL-1 cells through confocal fluorescence microscopy (Fig. [110]2b), and the quantified uptake rate further confirmed the feasibility of the use of GqDNVs-gel to deliver GqDNVs to mouse cardiomyocytes (Fig. [111]2c). To determine whether the GqDNVs-gel continuously releases GqDNVs in the mouse heart, we delivered the same dose of DIR (1,1’-dioctadecyl-3,3,3’,3’-tetramethylindotricarbocyanine iodide) [[112]42]-prelabelled GqDNVs through a myocardial drop, injection or GqDNVs-gel to the hearts of the mice (Fig. [113]2d). Fluorescence images of live mice and excised hearts were captured at 2, 12, and 24 h (Fig. [114]2e). Our in vivo and ex vivo imaging data showed that the GqDNVs released by GqDNVs-gel were quickly distributed in the heart from 2 h after delivery. Notably, GqDNVs-gel intervention led to high retention of GqDNVs in the heart after 12 h, which was sustained after 24 h compared with the GqDNVs drop or GqDNVs injection (Fig. [115]2f–g). Overall, our results suggest that GqDNVs-gel can promote the heart delivery efficiency of GqDNVs. Fig. 2. [116]Fig. 2 [117]Open in a new tab Uptake of GqDNVs by cardiomyocytes and in vivo imaging in mice. a A schematic diagram illustrates the assay for GqDNVs uptake. b Representative fluorescence images show PKH26-labeled GqDNVs (red) taken up by cardiomyocytes (green) in different groups, including PBS, GqDNVs, and GqDNVs-gel groups. c Quantitative analysis of GqDNVs uptake by cardiomyocytes. d A schematic diagram of in vivo and ex vivo IVIS imaging design. e Representative IVIS images of the mice and hearts at different time points. Quantification of fluorescence signals in vivo (f) and quantification of fluorescence in isolated hearts after GqDNVs treatment (g). All data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. GqDNVs: Gouqi-derived nanovesicles The fibrin gel serves as a stable carrier that proficiently encapsulates and controls the release of GqDNVs. It provides mechanical support and forms a high-concentration exocrine reservoir within the cardiac tissue. This enhances the targeting capability, prolongs the retention of the exocrine system within in the target area, and augments its concentration in cardiac tissue. Additionally, fibrin gel is biodegradable and can be gradually degraded in vivo [[118]28], thus controlling the release rate of GqDNVs. This controlled release mechanism helps achieve sustained therapeutic effects in the heart, mitigating the risk of diminished efficacy due to associated with the rapid release of GqDNVs. Cardiomyocyte biocompatibility and potential therapeutic effects of GqDNVs-gel HL-1 cardiomyocytes are widely used to study cardiac biology, particularly hypoxia-induced cellular responses [[119]43]. Utilizing the CCK-8 assay, it was observed that the viability of HL-1 cells remained largely unaffected following incubation with PBS, fibrin gel, GqDNVs, or GqDNVs-gel (Fig. [120]3a). In vitro assessments demonstrated that each constituent of the GqDNVs-gel was non-toxic and had excellent cell affinity and biocompatibility with cardiomyocytes. Subsequently, we sought to investigate the potential therapeutic effects of GqDNVs and GqDNVs-gel in vitro. As illustrated in Fig. [121]3b, c, 12 h of hypoxic treatment significantly decreased the viability of HL-1 cells; however, treatment with GqDNVs-gel effectively restored cell viability under hypoxic conditions. Compared with the Hypoxia group and the Hypoxia + gel group, GqDNVs-gel treatment significantly mitigated the hypoxia-induced overexpression of Bax mRNA levels, thereby increasing the relative mRNA expression ratio of Bcl-2/Bax (Fig. [122]3d). Immunofluorescence analysis indicated a reduction in TUNEL^+ cells and an increase in Ki67^+ and PCNA^+ cells in the Hypoxia + GqDNVs-gel group than in both the Hypoxia group and the Hypoxia + gel group (Fig. [123]3e–g). The results indicated that the fibrin gel was only a vehicle rather than a component contributing to apoptosis inhibition and proliferation promotion, while the GqDNVs were the main therapeutic portion. This finding is consistent with the previous publications, which found that the function of fibrin gel was prolonging the residence of exosomes on infarcted tissue as well as providing structural support [[124]40, [125]44]. Overall, GqDNVs-gel was biocompatibility, could reduce apoptosis, and promote the proliferation of cardiomyocytes via GqDNVs. Fig. 3. [126]Fig. 3 [127]Open in a new tab Biocompatibility and potential protective effects of GqDNVs-gel on cardiomyocytes. a The effect of each component of the GqDNVs-gel on cardiomyocyte viability. b Influence of hypoxia duration on cell viability. c Cardiomyocyte proliferation in various media under hypoxic conditions. d Bax and Bcl-2 mRNA expression under hypoxia in different media. e A TUNEL assay shows apoptotic nuclei (red) in cardiomyocytes across different groups, including PBS control, Hypoxia control, Hypoxia + gel control, and Hypoxia + GqDNVs-gel groups. f Confocal images show Ki67^+ (red) nuclei in cardiomyocytes (green). g Confocal images showed PCNA^+ (red) nuclei in cardiomyocytes (green). Quantitative analysis was performed via ImageJ software. All the data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel GqDNVs-gel benefits cardiac repair after MI in mice In this study, C57BL/6 mice were subjected to left anterior descending artery ligation to establish an MI model [[128]45]. Subsequently, an equivalent volume of either PBS or GqDNVs-gel was administered to the cardiac tissue of the mice (Fig. [129]4a). Over the 14-day experimental period, the survival rate of mice treated with GqDNVs-gel was significantly higher than that of the MI group, indicating the therapeutic efficacy of our intervention in rescuing mice post-MI (Fig. [130]4b). Echocardiographic evaluations were conducted to assess ventricular systolic function and the structural integrity of LV at 4 h and 14 days post-MI (Fig. [131]4c). At 4 h post-MI, both the MI group and the MI + GqDNVs-gel group presented a marked decrease in LVEF and LVFS, along with a significant increase in LV mass and LVEdV, as compared to the Sham group (Fig. S1). These results confirmed the successful establishment of the MI model before GqDNVs-gel intervention had a chance to repair the heart. At 14 days post-MI, the MI + GqDNVs-gel group demonstrated a significant reduction in heart rate, QRS duration, and LV mass, as well as a notable improvement in cardiac output, relative to those of the MI group (Fig. [132]4d). Furthermore, treatment with GqDNVs-gel significantly enhanced overall LV function, as evidenced by increased LVEF and LVFS, along with decreased LVEdV and LVEsV (Fig. [133]4e). Myocardial infarct size and fibrosis were evaluated using H&E and Masson’s trichrome staining (Fig. [134]4f). Masson’s staining revealed that GqDNVs-gel treatment significantly reduced fibrotic tissue and increased the wall thickness of the infarct zone (Fig. [135]4g, h), suggesting that GqDNVs-gel effectively mitigated cardiac fibrosis and adverse remodeling following MI. Fig. 4. [136]Fig. 4 [137]Open in a new tab Therapeutic efficacy of GqDNVs-gel in MI mice. a Schematic outlines the mouse study design. Sham group: open chest without artery ligation, MI group: MI mice with PBS-treated, and MI + GqDNVs-gel group: MI mice with GqDNVs-gel treatment. b Mouse survival rates. c Representative echocardiographic images were captured at 4 h and 14 days post-MI. d–e Quantitative analysis of echocardiography data; the parameters included heart rate, QRS duration, LV mass, cardiac output, LVEF, LVFS, LVEdV, and LVEsV. f Representative images from HE staining and Masson’s trichrome staining. Higher magnification images are shown in dashed outline boxes. g Infarct size and (h) wall thickness quantified using ImageJ. All data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. GqDNVs: Gouqi-derived nanovesicles, GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel, LV: left ventricle, LVEF: left ventricle ejection fraction, LVES: left ventricle fractional shortening, LVEdV: left ventricle end-diastolic volume, LVEsV: left ventricle end-systolic volume, MI: myocardial infarction Combined analysis of transcriptomics and proteomics of the mouse heart To elucidate the alterations in gene and protein expression associated with the therapeutic efficacy of GqDNVs-gel in MI mice, we conducted transcriptomic and proteomic sequencing analyses on the MI sites within the hearts of mice from both the MI group and the GqDNVs-gel intervention group (Fig. [138]5 & Fig. S2). The PCA results revealed distinct separation trends in gene and protein expression profiles between the MI and MI + GqDNVs-gel groups (Fig. S2a, b). Differential expression of genes (Fig. S2c) and proteins (Fig. S2d) between these two groups was assessed using volcano plots. KEGG annotation analysis was subsequently used to identify the highly enriched pathways. The differentially expressed genes and proteins were significantly involved in the Mitogen-Activated Protein Kinase (MAPK) signaling pathway and various cardiomyopathy-related processes (Fig. [139]5a). By integrating mRNA data from the transcriptome with protein data from the proteome, we identified a total of 44 differential biomarkers common to both omics analyses (Fig. [140]5b). To provide a more intuitive representation of the expression differences in the levels of genes and proteins between the MI and MI + GqDNVs-gel groups, a radar plot for differentially expressed biomarkers is visualized (Fig. S2e). Among these, 28 biomarkers exhibited consistent downregulation at both the transcriptional and protein levels, while 10 were consistently upregulated (Fig. [141]5c). Mantel test analysis showed that changes in these biomarkers affected MI outcomes such as mortality rate, cardiac function, and myocardial infarction area in MI mice (Fig. [142]5d). Notably, Mapk14, also known as p38 MAPK [[143]46], showed a significant association with all three outcomes mentioned above. A subsequent KEGG pathway enrichment analysis was conducted on these outcome-related key biomarkers (Fig. [144]5e). The pathways mostly enriched with these key biomarkers were linked to diabetic cardiomyopathy (Mapk14, Tgfb2, Col1a1, and Col3a1), the relaxin signaling pathway (Mapk14, Col1a1, and Col3a1), platelet activation (Mapk14, Col1a1, and Col3a1), and apoptosis (Casp7 and Bax). Furthermore, the relative expression levels of Il1r1, Mapk14, Casp7, Bax, Tgfb2, Fn1, Col3a1, and Col1a1 were downregulated in the GqDNVs-gel intervention group compared with the MI group (Fig. S2f). The relaxin signaling pathway has been found to play a regulatory role in cardiac repair by facilitating extracellular matrix remodelling and promoting angiogenesis [[145]47–[146]49]. In addition, relaxin signaling has been shown to activate the p38 MAPK pathway, thus playing an important role in cell proliferation and survival [[147]50]. These results showed that GqDNVs-gel intervention primarily functions to inhibit the p38 MAPK signaling pathway, reduce myocardial apoptosis and fibrosis, promote endogenous repair; and ultimately improve myocardial injury. Fig. 5. [148]Fig. 5 [149]Open in a new tab Transcriptomic and proteomic analysis of mouse hearts in MI and MI + GqDNVs-gel groups. a A bar chart of the KEGG enrichment pathways for genes and proteins differentially expressed between the two groups. b A Venn diagram displays the overlap of differentially expressed genes and proteins. c A nine-quadrant diagram shows the distribution of these differentially expressed genes and proteins. d A correlation network plot of differentially expressed biomarkers and MI clinical outcomes. e Chord plot of GO pathways enriched with differentially expressed biomarkers. Differentially expressed genes or proteins were identified with |Fold Change|> 2, P < 0.05 and VIP > 1. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel = Gouqi-derived nanovesicles-fibrin gel, MI = myocardial infarction GqDNVs-gel attenuates myocardial apoptosis, limits preexisting cardiac fibrosis, and promotes endogenous repair To verify the therapeutic functions of the GqDNVs-gel intervention as predicted by omics analyses, we conducted an examination of apoptosis, fibrosis, and endogenous repair in the infarcted area. We found that the mRNA expression levels of the Bax, Caspase-7, Caspase-3, and protein expression levels of the Bax, cleaved caspase-7, and cleaved-caspase-3 were significantly lower in the GqDNVs-gel intervention group than in the MI group (Fig. [150]6a–c). PCR analysis further revealed that the gene expression levels of α-SMA, Col3a1, Col1a1, and fibronectin 1 (FN1) were lower in the GqDNVs-gel treatment group than in the MI group (Fig. [151]6d), which is consistent with the results of our transcriptomic analysis. TUNEL staining and quantitative analysis confirmed a reduction in apoptotic nuclei in the GqDNVs-gel-treated MI hearts than in the MI hearts (Fig. [152]6e, f), further demonstrating that GqDNVs-gel treatment inhibited cell apoptosis after MI. We stained heart sections for α-SMA (fibroblasts), CD31 (vascular endothelial cells), or Ki67/PCNA (indicative of proliferating cells). Fewer fibroblasts (Fig. [153]6g, h), more vascular endothelial cells (Fig. [154]6i, j), and more proliferated cells (Fig. [155]6k–m) were detected in the infarcted area in the hearts of the GqDNVs-gel treated group than in those of the MI group. Next, we tested the release of growth factor in the mouse serum. We found that the TGFβ-2 levels were reduced, whereas the levels of IGF-1 and VEGF-A were increased in the GqDNVs-gel intervention group compared with those in the MI group (Fig. [156]6n). Fig. 6. [157]Fig. 6 [158]Open in a new tab Effects of GqDNVs-gel on post-MI apoptosis, fibrosis, and angiogenesis in mice. a The mRNA expression levels of Bax, Bcl-2, Caspase-7, and Caspase-3. b, c Protein expression levels of Bax, Bcl-2, Cleaved Caspase-7, Caspase-7, Cleaved Caspase-3 and Caspase-3. d The mRNA expression levels of α-SMA, Col3a1, Col1a1, and FN1. e Immunostaining of TUNEL in the infarcted area. Higher magnification images are shown in dashed outline boxes. Red, TUNEL; blue, DAPI. f Quantification of TUNEL-positive area of each group. g Immunostaining for α-SMA in the infarcted area. Higher magnification pictures are shown in dashed outline boxes. Red, α-SMA; blue, DAPI. h Quantification of α-SMA fluorescence intensity in each group. i Immunostaining of CD31 in the infarcted area. Higher magnification images are shown in dashed outline boxes. Red, CD31; blue, DAPI. h Quantification of CD31 fluorescence intensity of each group. g Immunostaining of Ki67 and PCNA in the infarcted area. Higher magnification pictures are shown in dashed outline boxes. Red, Ki67; green, PCNA, blue, DAPI. Quantification of Ki67-positive (l) and PCNA-positive (m) areas in each group. n The serum levels of growth factors in different groups. All the data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. GqDNVs-gel = Gouqi-derived nanovesicles-fibrin gel, MI = myocardial infarction Apoptosis after MI is an important contributor to cardiac dysfunction [[159]51, [160]52]. GqDNVs contain various components with antioxidant properties, which might help alleviate oxidative stress, diminish free radical damage to myocardial cells, and inhibit the activation of endogenous and exogenous pro-apoptotic signaling pathways in myocardial cells [[161]53]. Myocardial fibrosis is usually caused by inflammation and myocardial cell necrosis, which not only affects the contractile function of the myocardium but also leads to changes in the structure of the heart [[162]54]. TGF-β is a key regulatory factor in fibroblast activation, mediating myofibroblast transformation and stimulating extracellular matrix protein synthesis [[163]55]. TGF-β effectively induces the expression of α-SMA in cardiac fibroblasts, which is a hallmark of myofibroblast transformation [[164]56]. In addition, TGF-β significantly and continuously stimulates the synthesis and secretion of extracellular matrix proteins such as collagen I, collagen III, and fibronectin [[165]57]. Our research found that GqDNVs reduce the occurrence of cardiac fibrosis by inhibiting the secretion of TGF-β2, suppressing excessive fibroblast activation and collagen deposition. LBPs have also been reported to inhibit CCl[4]-induced fibrosis and α-SMA expression in the rat liver by suppressing the expression of the TLRs/NF-kB axis [[166]58]. In addition, we found that GqDNVs can increase the secretion of growth factors such as VEGF and IGF-1, stimulate local angiogenesis and promote cardiac remodelling. This could be because GqDNVs can transmit information between cells through their tiny structures and regulate the endogenous repair ability of the damaged myocardium. These growth factors can promote the generation of new blood vessels, improve the blood supply to myocardial ischemic areas, and support the repair and regeneration of myocardial tissue [[167]59, [168]60]. GqDNVs-gel inhibits p38-NF-κB p65 pathway in MI hearts Next, we sought to identify the key pathway perturbed by GqDNVs-gel intervention. GqDNVs-gel treatment significantly alleviated the MI-induced overexpression of p38, JNK, and p65 mRNA levels (Fig. [169]7a) and significantly inhibited the MI-induced increase in p38 and p65 protein phosphorylation levels (Fig. [170]7b, c). Immunofluorescence analysis revealed a striking accumulation of phospho-p38 in perivascular and interstitial cells in cardiac sections from MI mice. In contrast, phospho-p38 was less observed in GqDNVs-gel-treated mice hearts (Fig. [171]7d–e). In vitro, we found that GqDNVs-gel treatment successfully reversed the hypoxia-induced nuclear accumulation of phospho-p38 and phospho-p65 in mouse cardiomyocytes, although some cytoplasmic phospho-p38 remained (Fig. [172]7f–i). Fig. 7. [173]Fig. 7 [174]Open in a new tab GqDNVs-gel reverses MI-induced activation of p38/NF-κB p65 pathway. a The mRNA expression levels of p38, JNK, ERK, and p65. b, c The phosphorylation levels of p38, JNK, ERK and p65 proteins were detected by Western blotting. d Immunostaining of p-p38 in the infarcted region. Green, p-p38; blue, DAPI. e Quantification of p-p38 fluorescence intensity in mouse hearts from each group. f Immunostaining of p-p38 in cardiomyocytes cultured in different mediums. Higher magnification images are shown in dashed outline boxes. Red, p-p38; blue, DAPI. g Quantification of p-p38 fluorescence intensity in each group in vitro. h Immunostaining of p-p65 in cardiomyocytes in different mediums. Higher magnification pictures are shown in dashed outline boxes. Red, p-p65; blue, DAPI. i Quantification of p-p65 fluorescence intensity in each group in vitro. All the data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel, MI: myocardial infarction We next examined the impact of GqDNVs-gel on p38-dependent apoptosis activation (Fig. [175]8). Immunofluorescence detection of phospho-p38 in cultured HL-1 cells showed that the use of DHC, a specific p38-activator, significantly activated the acute phosphorylation of p38 and promoted the nuclear accumulation of phospho-p38. However, treatment with GqDNVs-gel led to a reduction in the total and nuclear accumulation of phospho-p38 (Fig. [176]8a). When a specific inhibitor of p38 (SB 203580) or GqDNVs-gel was used, the acute phosphorylation of p38 was significantly blunted (Fig. [177]8a). In a p38-activated or p38-inhibited cell model, the activation of p38 inhibited the number of stimulated apoptotic nuclei and reduced proliferation, whereas the inhibition of p38 or treatment with GqDNVs-gel led to fewer apoptotic nuclei and an increase in proliferating cardiomyocytes (Fig. [178]8b–d). The quantitative analysis results further confirmed these findings (Fig. [179]8e). Our results demonstrated that GqDNVs-gel may alleviate heart damage after MI by inhibiting the activation of the p38 MAPK and NF-κB p65 pathways. Fig. 8. [180]Fig. 8 [181]Open in a new tab Phospho-p38 is essential for GqDNVs-gel-induced cardio-protection. a Immunostaining of p-p38 in cardiomyocytes in different mediums. Higher magnification images are shown in dashed outline boxes. Red, p-p38; blue, DAPI. b Immunostaining of TUNEL in the infarcted area. Red, TUNEL; blue, DAPI. c Confocal images showing Ki67^+ (Red) nuclei in cardiomyocytes (green). d Confocal images showing PCNA^+ (Red) nuclei in cardiomyocytes (green). e Quantification of p-p38 fluorescence intensity, TUNEL-positive, Ki67-positive, and PCNA-positive cells in each group in vitro. All the data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. DHC: dehydrocorydaline chloride, a specific p38-activator; GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel; MI: myocardial infarction; SB: SB 203580, a specific p38-inhibitor Effect of GqDNVs-gel intervention on the metabolite profile of mouse hearts via saccharides-targeted and untargeted metabolome analysis A total of 22 types of saccharides were identified in the hearts of the MI group, 21 of which were also present in the MI + GqDNVs-gel group (Fig. S3a). In addition, 12 types of saccharides identified in GqDNVs were detected in both groups of mice (Fig. S3b). The saccharides composition in mouse hearts between the two groups is displayed in Fig. S3c and Table S2, and no significant difference in saccharides levels was found between the MI and MI + GqDNVs-gel groups. Heat maps were also created to display the relative distribution of saccharides in GqDNVs and mouse hearts of the MI + GqDNVs-gel group (Fig. S3d). Besides, Mantel test analysis found no significant associations between saccharides in GqDNVs and the clinical outcomes or biomarkers of genes and proteins in mouse hearts (Fig. S3e). There were 2470 metabolites detected in the cardiac tissues of the mice across two experimental groups, and all the metabolites detected in the MI groups were also present in the MI + GqDNVs-gel group (Fig. [182]9a). A total of 50 metabolites in the GqDNVs were also detected in the cardiac tissues of the both groups (Fig. S4a). PCA analyses revealed a separation trend in metabolites between the MI and MI + GqDNVs-gel groups (Fig. S4b). Furthermore, the volcano plot showed that 19 metabolites were significantly higher after GqDNVs-gel intervention than the MI group, whereas 14 metabolites were significantly lower after GqDNVs-gel intervention (Fig. [183]9b). Among these, octadecadienamide, which was significantly elevated in the heart after GqDNVs-gel intervention, was also present in the GqDNVs (Fig. [184]9c). Octadecadienamide is usually produced in the fatty acid metabolism of some plants and microorganisms and may have biological functions such as antioxidation and antibacterial effects [[185]61]. This finding indicated that GqDNVs are absorbed by the mouse heart, thereby affecting their metabolite composition. The differentially abundant metabolites between the MI and MI + GqDNVs-gel groups were primarily associated with specific metabolic pathways and Glycerophospholipid metabolism (Fig. [186]9d). The top 20 substances with the highest differential multiples are displayed in Fig. [187]9e. To further determine the key metabolites affected by GqDNVs-gel intervention, a Mantel test was performed between the differentially abundant metabolites and clinical outcomes or key biomarkers of genes and proteins (Fig. S4c). The significant associations were further visualized via a network plot (Fig. [188]9f). Eventually, six metabolites that were significantly related to outcomes or key biomarkers, and were among the top 20 highly differentially expressed substances were screened out. Among them, Arg-Thr-Ile-Glu, N-(octadecanoyl)-sphing-4-enine-1-phosphocholine, Liquiritin, and Plastoquinone A were upregulated after GqDNVs-gel intervention, whereas PE-NMe (18:1 (11Z) /16:0) and (7R, 8Z)-bacteriochlorophyll b were downregulated after GqDNVs-gel intervention. Additionally, four upregulated key metabolites were positively associated with improved clinical outcomes and negatively associated with the expression levels of key biomarkers of genes and proteins, whereas two downregulated key metabolites were just the opposite (Fig. [189]9g–i). Arg-Thr-Ile-Glu is a small peptide chain, which is associated with energy metabolism and muscle recovery, was found to be upregulated following the GqDNVs-gel intervention. Conversely, PE-NMe (18:1 (11Z) /16: 0), a specific N-methylated phosphatidylethanolamine (PE), was downregulated post-intervention. The abnormal expression or modification of methylated PE may be associated with imbalances in membrane lipid metabolism in cardiovascular diseases [[190]62]. Fig. 9. [191]Fig. 9 [192]Open in a new tab Metabolomic profiling of whole mouse hearts in MI and MI + GqDNVs-gel. a A venn diagram of metabolites in mouse hearts from the MI and MI + GqDNVs-gel groups. b A volcano plot of metabolites between the hearts of the two groups of mice. Red and green represent up- and down-regulation, respectively. c A venn diagram of metabolites present in GqDNVs and those differentially abundant between the MI and MI + GqDNVs-gel groups. d KEGG enrichment analysis for differential metabolites between the MI and MI + GqDNVs-gel groups. e A correlation network plot of differential metabolites and their associations with clinical outcomes, gene and protein biomarkers. Only metabolites with significant correlations, as determined by Mantel’s analysis are displayed (P-value < 0.05, r ≥ 0.4). Red boxes indicate significant correlations with outcomes, gene and protein biomarkers. Rose red boxes indicate significant correlations with two of those indicators. Blue boxes indicate a significant correlation with one of these indicators. f A dynamic distribution map of differentially abundant metabolites. g A heatmap of the correlations between selected metabolites and clinical outcomes of mice. h A heatmap of the correlation between selected metabolites and the expression of gene biomarkers. i A heatmap of the correlations between selected metabolites and the expression of protein biomarkers. Differentially expressed metabolites were identified with |Fold Change|> 2, P-value < 0.05, and VIP > 1. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel: Gouqi-derived nanovesicles-fibrin gel, MI: myocardial infarction These findings align with previous research conducted in our laboratory [[193]23], demonstrating that the effects of GqDNVs was closely related to their influence on metabolic pathways, particularly lipid metabolisms. Glycolipid metabolism plays a crucial role in cardiac remodelling by providing energy and maintaining cellular function [[194]63]. However, myocardial ischemia can disrupt energy metabolism and lead to dysregulation of glucose and lipid metabolism, ultimately resulting in cell death and impaired cardiac function [[195]64]. Our results suggest that while sugar components affect the energy supply during cardiac repair process after MI, lipid metabolic pathways, particularly those involving glycerophospholipids metabolism, linoleic acid metabolism, and α-linolenic acid metabolism play more prominent roles in this process. Conclusions To our knowledge, this is the first study investigating the cardioprotective effects of GqDNVs, offering a novel therapeutic approach for cardiovascular diseases. We found that GqDNVs-gel increased the delivery efficiency of GqDNVs, effectively improved survival rates over a 14-day post-MI period, enhanced cardiac function, reduced infarct size, mitigated myocardial apoptosis and excessive fibrosis, and promoted endogenous cardiac repair. Moreover, we uncovered that the cardioprotective effects are mediated through the inhibition of the p38 MAPK-NF-κB p65 signaling pathway. The intervention of GqDNVs can influence the structure of cardiac substances and impact myocardial lipid metabolism, with a particular emphasis on glycerophospholipids metabolism and α-linolenic acid metabolism pathways in mouse hearts. These findings represent a novel and promising strategy for treating cardiovascular diseases, highlighting the potential of GqDNVs in advancing cardiac therapeutics. Supplementary Information [196]12951_2025_3615_MOESM1_ESM.docx^ (2MB, docx) Additional file 1: Figure S1. Quantitative analysis of cardiac function in mice assessed by echocardiography at 4 h post MI. (a) Left ventricle mass of mice. (b) Cardiac output. (c) Left ventricle ejection fraction. (d) Left ventricle fractional shortening. (e) Left ventricular end-diastolic volume. (f) left ventricular end- systolic volume. All the data are presented as mean ± standard deviation. Comparisons were performed using one-way ANOVA followed by Tukey’s multiple comparison analysis. The notation ‘ns’ denotes non-significance, while *, **, and *** correspond to P-values of < 0.05, < 0.01, and < 0.001, respectively. Figure S2 Profiling of genes and proteins in hearts from MI and GqDNVs-gel intervention groups. (a) Principal component analysis (PCA) of genes for all samples. (b) PCA of proteins for all samples. (c) A volcano plot for genes between the hearts of two groups of mice. Red and green represent up- and down-regulation, respectively. (d) A volcano plot of protein expression in the hearts of two groups of mice. Red and green represent up- and down-regulation, respectively. (e) A radar plot showing the expression of biomarkers with differential expression of both genes and proteins between the MI and MI + GqDNVs-gel groups. (f) Violin plots of key biomarkers in the MI and GqDNVs-gel intervention groups.. Differentially expressed genes or proteins were identified with |Fold Change|> 2, P-value < 0.05, and VIP > 1.. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel = Gouqi-derived nanovesicles-fibrin gel, MI = myocardial infarction. Figure S3 Saccharides-targeted metabolome profiling of whole mouse hearts in MI and GqDNVs-gel intervention groups. (a) A venn diagram of saccharides composition in hearts of two groups of mice. (b) A bar chart showing the levels of saccharides in the hearts of two groups of mice. (c) A venn diagram of saccharides composition in GqDNVs and in hearts of two groups mice. (d) A heatmap of the correlation between saccharides in GqDNVs and the hearts of mice in the GqDNVs-gel intervention group. (e) A correlation network plot of saccharides profiling in GqDNVs and clinical outcomes/gene biomarkers/protein biomarkers of MI mice. Differentially expressed saccharides were identified with |Fold Change|> 2, P-value < 0.05, and VIP > 1.. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel = Gouqi-derived nanovesicles-fibrin gel, MI = myocardial infarction. Figure S4 Metabolome profiling of whole mouse hearts in MI and GqDNVs-gel intervention groups. (a) PCA of metabolites for all samples. (b) A venn diagram of metabolites in GqDNVs and in hearts of two groups mice. (c) A correlation network plot of metabolite profiles and clinical outcomes/gene biomarkers/protein biomarkers of mouse hearts in two groups.. Differentially expressed metabolites were identified with |Fold Change|> 2, P-value < 0.05, and VIP > 1.. GqDNVs = Gouqi-derived nanovesicles, GqDNVs-gel = Gouqi-derived nanovesicles-fibrin gel, MI = myocardial infarction. [197]Additional file 2.^ (29.2KB, docx) Acknowledgements