Abstract Extracellular vesicles (EVs), especially those derived from stem cells, have emerged as a novel treatment for promoting wound healing in regenerative medicine. However, the clinical application of mammalian cells-derived EVs is hindered by their high cost and low yields. Inspired by the ability of EVs to mediate interkingdom communication, we explored the therapeutic potential of EVs released by the probiotic strain Lactobacillus rhamnosus GG (LGG) in skin wound healing and elucidated the underlying mechanism involved. Using full-thickness skin wound-healing mouse models, we found that LGG-EVs accelerated wound healing procedures, including increased re-epithelialization and promoted angiogenesis. Using in vitro experiments, we further demonstrated that LGG-EVs boosted the proliferation and migration capacities of both epithelial and endothelial cells, as well as promoted endothelial tube formation. miRNA profiling analysis revealed that miR-21-5p was highly enriched in LGG-EVs and LGG-EV treatment significantly increased miR-21-5p level in recipient cells. Mechanically, LGG-EVs induced regulatory effects via miR-21-5p mediated metabolic signaling rewiring. Our results suggest that EVs derived from LGG could serve as a promising candidate for accelerating wound healing and possibly for treating chronic and impaired healing conditions. Graphical abstract [44]graphic file with name 12951_2024_2893_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-02893-8. Keywords: Wound healing, Extracellular vesicles, Lactobacillus rhamnosus GG, miRNA sequence, Bacteria Introduction Skin wound managements pose significant clinical challenges, due to frequent trauma, tearing, cuts, or burns [[45]1]. In certain pathological conditions, such as diabetes, obesity, and aging, the wound healing processes are disrupted, often resulting in pathological conditions, such as chronic or non-healing wounds, and even tumor formation, thereby threatening the life quality for millions of people worldwide [[46]2]. Despite numerous therapeutic strategies being reported, inconsistent clinical outcomes and undesirable side effects restrict their long-term efficacy [[47]1], underscoring the need to develop optimal treatment approaches. Wound healing is a complicated process that aims to regain the structural and functional integrity of damaged tissues. The healing procedure is orchestrated by multiple cells, signaling pathways, and bioactive molecules [[48]3, [49]4]. Typically, it involves a series of sequential and overlapping phases, including inflammation, proliferation (granulation tissue formation and re-epithelization), and resolution. Immediately following injury, blood vessels constrict to minimize blood loss and immune cells are recruited into the injured sites, removing microbes and orchestrating the multiple healing processes. Concurrently, activated fibroblasts migrate to the wounds, forming the granulation tissue, which contains abundant extra-cellular matrix (ECM) and blood vessels. The filled wound space provides a scaffold for tissue repair. In this case, endothelial cells develop new blood vessels that provide nutrients and oxygen to lesion sites, alleviating uncontrolled inflammation. Keratinocytes proliferate and migrate across the wound surface to finish re-epithelization to restore the barrier function. Finally, these episodes stop and the tissue is remodeled in resolution stage. In diabetic wounds, however, hyperglycemia and long-term oxygen deprivation lead to continuous pro-inflammatory state, disturbed angiogenic response and compromised re-epithelialization, which subsequently contribute to wound healing failure. Thus, a feasible approach for chronic wound healing would simultaneously induce angiogenesis and modulate epidermal cell functions. Extracellular vehicles (EVs) are membrane-enclosed particles secreted by almost all cells, ranging in size from 30 to 150 nm in diameter and containing encapsulated bioactive components, such as microRNAs (miRNAs), proteins, and mRNAs [[50]5]. Recent studies have highlighted their remarkable biomedical potential in various wound healing processes, including re-epithelization, immunoregulation, and ECM remodeling [[51]6]. EVs derived from mammalian cells, as exemplified by mesenchymal stem cells (MSCs) [[52]7, [53]8], macrophages [[54]9], and endothelial progenitor cells [[55]10], have been documented to accelerate wound closure by presenting pleiotropic effects simultaneously. Despite the potential advantages of EVs as therapeutics in regenerative medicine, their clinical application still faces many limitations, including difficulty in large quantities, high cost of cell culture, and ethical issues. Interestingly, bacterial EVs (BEVs) have emerged as an alternative with the potential to overcome these limitations. BEVs function as mediators of interkingdom communication enabling the shuttle of bioactive components and regulating various biological processes, making them promising candidates for disease treatment [[56]11]. Several BEVs have been reported for disease therapy. For example, Akkermansia muciniphila-derived EVs improved intestinal barrier function [[57]12]. EVs from Lactobacillus druckerii displayed great promise in anti-fibrosis [[58]13]. Lactobacillus reuteri extracts haven been shown to elicit therapeutic effects on injury repair via PI3K-AKT pathway [[59]14]. On the other hand, oral antibiotic treatment delays wound repair [[60]15], and supplementation of gut microbiome in drinking water accelerates wound healing process [[61]16], indicating that gut bacteria or their products may participate in skin wound healing. However, little is known about the potential role of EVs from probiotics in wound healing. Lactobacillus rhamnosus GG (LGG) is a gut probiotic that benefits health in many aspects, including enhancing intestinal barrier function [[62]17], regulating gut microbiota structure [[63]18], and promoting fin regeneration of zebrafish [[64]19]. Particularly, LGG lysate can increase the re-epithelialization of keratinocytes [[65]20], which endows them with the possibility of pro-healing action. However, the therapeutic effects and possible mechanisms of LGG-EVs on cutaneous wound healing remain to be determined. Here, we isolated EVs from LGG and investigated their effects on wound healing both in vivo and in vitro. Our results demonstrated that the local injection of LGG-EVs promoted mouse wound closure, accompanied by accelerated re-epithelialization and increased angiogenesis. Moreover, LGG-EVs were found to enhance the proliferation and migration of keratinocytes and endothelial cells, as well as the proangiogenic ability of endothelial cells. Using miRNA sequencing, we identified that LGG-EVs contained a high amount of miR-21-5p and mechanistic investigation revealed that this miRNA is crucial for LGG-EVs-mediated activation of endothelial cells and keratinocytes that was related to AKT and HIF1α signaling pathways. Collectively, our study comprehensively illustrates the therapeutic benefits and mechanisms of action for LGG-EVs in wound healing and open innovative avenues for probiotic bacteria EVs in regenerative medicine. Materials and methods LGG culture LGG strain was obtained from ATCC 53103 and cultivated in MRS medium (Solarbio Science & Technology) [[66]21]. Briefly, after twelve hours of culture at 37 °C and 220 rpm (primary culture), LGG was cultivated at 37 °C and 220 rpm for 8 h (secondary culture). Subsequently, the secondary fermentation broth (1 mL) was supplemented with MRS (50 mL) and cultivated at 37 °C and 220 rpm for 24 h. The obtained fermentation supernatant was used for EV isolation. Isolation and identification of LGG-EVs The isolation of EVs from LGG was conducted as previously reported [[67]21]. Firstly, the fermentation supernatant was centrifugated at 10,000 g for 15 min to wipe off bacteria and other debris, followed by filtration with filters (0.22 μm) to remove large particles. After ultra-centrifugation of the solution at 150,000 g for 2 h, the pellet was sequentially resuspended with PBS, concentrated through an ultrafiltration membrane (50–100 kDa), and subjected to a second step ultracentrifugation at 150,000 g for 2 h. The pure EV pellets were resuspended with PBS and filtered with 0.22 μm sterile filters. During EV preparation, all processes were carried out at 4 °C. The putative concentration of LGG-EVs was measured with a BCA assay kit (Beyotime, P0010). The particle size distribution of LGG-EVs was determined by Nanoparticle tracking analysis (NTA; ZetaView). The morphology of LGG-EVs was detected by Transmission electron microscopy (TEM; Hitachi). The zeta potential of LGG-EVs was examined by dynamic light scattering (Zetasizer Nano ZS90, Malvern). Mouse skin wound models and LGG-EV treatment Eight-week-old male C57BL/6J mice (Vital River Laboratory Animal Technology) were used in present study. After anesthetization and hair removal, four full-thickness incisions (6 mm) were made on both sides of midline of mouse back [[68]22]. Twenty mice were randomly separated into two groups Control vehicle (PBS treatment) group and the LGG-EVs group (n = 5 mice per group at each time point). The mice were subcutaneously injected with either 100 µL PBS (Control) or LGG-EVs (50 µg LGG-EVs in 100 µL PBS) around the wounds once a day, respectively. On day 0, 4, and 7 post-wounding, a digital photograph of the wounds was taken. Wound areas were measured by referring to a circle slide with 6 mm in diameter. The wounds and slides were photographed, and the wound area was calculated using ImageJ. On day 4 and 7, mice were sacrificed. The wounds were picked for H&E, immunohistochemical and qRT-PCR analysis. All experimental procedures were approved by the Institutional Animal Care and Use Committee of Shanghai University. Cell culture The human skin keratinocytes (HaCaT cells) were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China), and the human umbilical vein endothelial cell line (HUVEC) was purchased from OriCell (Guangzhou, China). HaCaT was cultured in DMEM containing 10%FBS and 1% penicillin-streptomycin. HUVEC was cultured in endothelial cell medium (ECM #1001; ScienCell) as previously described [[69]23]. HUVEC of passage 3 or 4 was used for experiments in our tests and HaCaT of passage less than 7. The culture medium was changed every other day and the cells were passaged when they are confluent. Histological and immunofluorescent analysis After fixed with 4% paraformaldehyde (PFA), the wound tissues were dehydrated and embedded in paraffin. Then, 5 μm-thickness sections were created. For evaluation of the degree of granulation tissue formation, the sections were subjected to Masson’s trichrome staining. For detection of epidermal regeneration, the sections were subjected to H&E staining as standard protocols. For immunofluorescent analysis, frozen sections (7 μm) were exposed to primary antibodies against CD31 (BD, 557355, 1:1000), F4/80 (Abcam, ab6640,1:500) or Ki67 (Servicebio, [70]GB111499, 1:400), followed by the secondary antibody treatment (Alexa 594-conjugated antibody, Thermo Fisher, A-11007/ A-11037, 1:500; Alexa 488-conjugated secondary antibody, Thermo Fisher, A-11029/ A-11006, 1:500). DAPI staining was conducted. ImageJ software was used to quantify the staining. At least three sections were used for the quantification. Quantitative real-time polymerase chain reaction(qRT-PCR) Toal RNA extraction was conducted using TRIzol [[71]24]. For mRNA detection, RNA reverse-transcription was conducted using a reverse transcription kit (Vazyme, R323-01) and qRT-PCR was conducted using SYBR Green (Vazyme, Q711-02). For miRNA detection, RNA was reverse-transcribed and quantified using miRNA SYBR qPCR Master Mix (Vazyme, MQ101-02). Relative mRNA or miRNA expression was normalized with Gapdh (mRNA) or U6 (miRNA), respectively. The relative expressions were calculated using the 2^−∆∆Ct method. Primer sequences used were as follows: Hif1α_F, ACCTTCATCGGAAACTCCAAAG, Hif1α_R, CTGTTAGGCTGGGAAAAGTTAGG; Vegfα_F, TAGAGTACATCTTCAAGCCG, Vegfα_R, TCTTTCTTTGGTCTGCATTC; Gapdh_F, CATGTTTGTGATGGGTGTGA, Gapdh_R, AATGCCAAAGTTGTCATGGA; miR-21-5p_F: GCAGTAGCTTATCAGACTGATG, miR-21-5p_R, GGTCCAGTTTTTTTTTTTTTTCAAC; U6_F, TGGAACGCTTCACGAATTTGCG, U6_R, GGAACGATACAGAGAAGATTAGC. EV internalization assay The internalization of LGG-EVs was investigated in HUVEC and HaCaT cells. Briefly, Cy5 with red fluorescence was used to label LGG-EVs as previous protocol instructed [[72]25]. Then, the labelled EVs were administrated to cells. After 6 h of incubation, cells were treated with DAPI, and visualized with a confocal microscope. Cell proliferation assay Ki67 staining was used to investigate the cell proliferation. Cells (5 × 10^4 cells/well for HUVEC and 8 × 10^4 cells/well for HaCaT) were seeded on the glass coverslips located in 12-well plates. After transfection with or without miR-21-5p inhibitor (miRi), cells were treated with a medium containing LGG-EVs (50 ng/µL) in 1% FBS for 24 h. Subsequently, the cells were sequentially fixed in 4% PFA, permeabilized with Immunostaining Permeabilization Buffer (Invitrogen, 00-8333-56) and incubated with 100% goat serum (BOSTER, AR0009) to block reactive sites. Subsequently, cells were treated with primary antibody against Ki67 (Servicebio, [73]GB111499, 1:400) at 4°C. After 12 h of incubation, Alexa 488-conjugated antibody (ThermoFisher, A-11008, 1:500) was added to cells at a temperature for 1 h. DAPI staining was conducted. The images were observed by fluorescence microscope (KEYENCE, BZ-X810). ImageJ software was used to quantify the staining. Three independent replicates were performed to examine the Ki67-positive cells. Scratch wound-healing assay In 48-well plates, cells (4 × 10^4 cells/well) were plated. After serum-free media starvation of 24 h, cells were scratched using a 200 µL pipette tip and washed with PBS to remove the dead cells. HUVEC and HaCaT cells were treated with a medium containing LGG-EVs (50 ng/µL) in 1% FBS for 24 h or 48 h, respectively, and then photographed. The wounded area was measured with ImageJ. Each was conducted in triplicate. The migration ratio was calculated as migration area (%) = (Initial wound area-remaining area at observation point)/ Initial wound area× 100. Transwell assay HUVEC was suspended in starvation medium and plated into the apical chamber of 24-well plates (8.0 μm pore filters, Corning, 3464) at a density of 4 × 10^4 cells/well. Meanwhile, 500 µL culture medium containing 10%FBS with or without LGG-EVs (50 ng/µL) was added to the basolateral chamber. After 24 h, cells on the lower side of the filter membrane were fixed, and stained with crystal violet (Sangon Biotech, E607309). The migrating cells were observed under an optical microscope (Nikon, Japan) and analyzed with ImageJ. Tube formation assay In 96-well plates, Matrigel (50 µL per well, Corning, 354230) was added. After polymerization, HUVECs were overlaid on Martigel-coated wells at a density of 4 × 10^4 cells/well, followed by exposure to LGG-EVs (50 ng/µL) or PBS for 6 h. Tube formation was visualized with microscopy (Nikon, Eclipse Ti2E). ImageJ software was used to quantify the tube meshes. miRNA sequencing and analysis The high-throughput sequencing and analysis of miRNAs in LGG-EVs was conducted as previously described [[74]25]. TruSeq Small RNA Sample Prep Kits (Illumina, San Diego, USA) were used to develop the miRNA sequencing library. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis were conducted using R package. Inhibitor transfection miR-21-5p inhibitor and corresponding control were purchased from RiboBio (Guangzhou, China). Cells were transfected with miR-21-5p inhibitor (100 nmol/L) or the relevant negative control. The transfection was conducted in HUVEC and HaCaT cells using Lipofectamine 3000 (Invitrogen, L3000008) as per the recommendations. The downstream experiments were performed after twenty-four hours of transfection. Western blot Proteins were prepared with radioimmunoprecipitation (RIPA) buffer (Beyotime, P0013K) containing PMSF (Beyotime, ST506) and phosphatase inhibitor cocktails. BCA Kits (Beyotime, P0010) were used to quantify proteins. Western blot was performed as described previously [[75]26]. ImageJ was employed to analyze the signal intensity (n = 3 independent replicas). The protein expressions were normalized to the GAPDH. The primary antibodies included p-AKT (CST, 9271, 1:1000), AKT (CST, 4691,1:1000), HIF1α (NOVUS, NB100-449, 1:2000), and GAPDH (Proteintech, 60004,1:50000). Statistical analysis All experimental data were expressed as mean ± s.d. from at least three independent replicas. We used Graph Pad Prism software for data analysis. Statistical Significance was determined using one-way ANOVA or Student’s t-test. P values of less than 0.05 were considered significant differences (*P < 0.05; **P < 0.01; ***P < 0.001; # P > 0.05). Results LGG-EV isolation and characterization LGG-EVs were isolated from the culture supernatants of LGG (Fig. [76]1A). TEM analysis revealed that LGG-EVs exhibited a typical sphere-shaped feature (Fig. [77]1B), consistent with previously described EVs from mammalian cells [[78]27]. NTA analysis showed that the particle size of LGG-EVs ranged from 30 to 200 nm in diameter, with the main peak observed around 180 nm (Fig. [79]1C). However, unlike mammalian-derived EVs, specific marker for BEV characterization is still lacking [[80]5, [81]28]. Zeta potential analysis is a common method to evaluate the stability of EVs, and the zeta potential value around or less than − 20 mV indicates good stability [[82]29]. As shown in Fig. [83]1D, the zeta potential of LGG-EVs was about − 19.4 mV, indicative of their high stability. These results collectively indicate that LGG-EVs possess a similar structure and size to EVs from other mammalian sources. Fig. 1. [84]Fig. 1 [85]Open in a new tab LGG-EV isolation and characterization. A Diagram describing the isolation of LGG-EVs. B Representative TEM images of LGG-EVs. Scale bar = 100 nm. C NTA to monitor the size of LGG-EVs. D Quantification of the zeta potential of LGG-EVs (n = 3). TEM, transmission electron microscopy; NTA, nanoparticle tracking analysis In vivo safety of LGG-EVs We then assessed the in vivo safety of LGG-EVs through subcutaneous injection into mice once a day for 14 days. H&E staining results revealed that compared with Control, there were no obvious pathological changes in major organs, including heart, kidney, liver, lung, and spleen, after LGG-EV treatment (Supplementary Fig. [86]1), supporting the notion that LGG-EVs are well-tolerated. This observation aligns with reports of favorable safety profiles for LGG-EVs and EVs from other bacteria [[87]25, [88]30]. These findings provide both a theoretical and experimental foundation for the clinical application of LGG-EVs. LGG-EV application accelerates cutaneous wound healing The retention and internalization are crucial for EV functions. To determine the contributions of LGG-EVs to wound healing, we firstly examined the retention and internalization of LGG-EVs in skin tissues after wound administration. LGG-EVs were labeled with Cy5, a red fluorescent dye [[89]25], and topically injected into the tissues around the wounds. Fluorescence microscopy analysis revealed that abundant Cy5-labeled LGG-EVs were detected and maintained in the perinuclear region of both skin epidermal keratinocytes and dermal cells at 24 h and 48 h after injection (Supplementary Fig. [90]2A). We further detected the capability of key dermal cell types to internalize LGG-EVs. To this end, we evaluated the co-localization of endothelial cells marked by CD31 and macrophages marked by F4/80 with Cy5 using immunostaining. We found that numerous CD31^+ or F4/80^ + cells were also with Cy5 signals, indicating that both cell types can internalize LGG-EVs (Supplementary Fig. [91]2B). These results confirmed the internalization of the LGG-EVs by resident wound cells and their retention in vivo. Subsequently, we investigated the impact of LGG-EVs on wound healing. Full-thickness skin wounds (6 mm punch) were inflicted in the back skin of C57BL/6 wildtype mice. LGG-EVs or Control vehicle (PBS) were subcutaneously injected into the edges of wounds once a day (Fig. [92]2A). The wound-healing kinetics were visualized and quantified at corresponding day 4 and day 7 post-injury. Macroscopical analysis of digital photographs showed that LGG-EVs treated wounds were smaller than in the Control group at day 7 (Fig. [93]2B-D). Wound tissues were collected at day 4 and 7 post-injury, which represents the inflammatory or proliferative phases of wound healing [[94]31]. H&E-stained analysis revealed shorter wound widths at days 4 and 7 post-wounding after LGG-EV treatment (Fig. [95]2E and F). As expected, the LGG-EV treatment group showed a longer hyperproliferative epithelial tongue on day 4 (Fig. [96]2E and G), indicative of accelerated re-epithelialization. Given the nearly enclosed epidermis in the LGG-EV group, we did not count the epithelial tongue on day 7. In addition to epidermal re-epithelialization, granulation tissue formation and ECM deposition are necessary for timely wound closure [[97]32]. LGG-EVs treated wounds exhibited thicker granulation tissue (gt) on day 4 and day 7 (Fig. [98]2E and H). Collagen synthesis, a hallmark of granulation tissue formation, increases to fill the wound gap during the early phase of healing [[99]32]. Therefore, Masson staining was performed to test the effect of LGG-EVs on collagen content. Compared with Control wounds, LGG-EVs treated wounds had more collagen content on day 7 (Fig. [100]2I and Supplementary Fig. [101]3), indicative of the enhanced granulation tissue formation. Altogether, these data suggest that LGG-EVs could accelerate the wound healing process in vivo. Fig. 2. [102]Fig. 2 [103]Open in a new tab LGG-EV application accelerates cutaneous wound healing. A Schematic of full-thickness wounds in mouse skin (n = 5 mice per time point). Either LGG-EVs or PBS were subcutaneously injected into skin around wounds once a day. On day 4 and day 7 post-wounding, the wounds were collected. B Representative time-lapse photographs of wounds treated with PBS or LGG-EVs at indicated day post-wounding. Scale bar = 2 mm. C Representative wound edge traces were established on each condition according to wound photographs. D Quantification of the wound size at indicated days (n = 5). E H & E images of the wounds on day 4 and day 7 (n = 5). The black dotted line indicates epithelial tongue. Scale bar = 500 μm. F-H Quantification of the scar widths (F) and granulation tissue (gt) thickness (H) on day 4 and day 7, as well as the length of the epithelial tongue (G) on day 4 (n = 5). I Representative Masson’s trichrome staining of wounds on day 7 (n = 5). Scale bar = 100 μm. he, hyperproliferative epithelium LGG-EV application enhances wound cell proliferation and angiogenesis In light of the enhanced re-epithelialization and vascularized granulation tissue formation after LGG-EV treatment, we reasoned that LGG-EVs may play a role in wound cell proliferation and angiogenesis processes. To test this hypothesis, we performed immunofluorescence staining for Ki67, a marker of proliferative cells, as well as CD31 in wounds. Compared to Control wounds on day 7, LGG-EV treatment significantly increased Ki67^+ cells within epithelium as well as dermal compartments (Fig. [104]3A-C), which may explain the increased re-epithelialization rate and granulation tissue content (Fig. [105]2E-I). Meanwhile, LGG-EVs treated wounds displayed a much higher proportion of CD31 positive tissue, indicative of more blood vessels within the granulation tissue compared with the Control wounds on day 7 (Fig. [106]3D and E). Furthermore, a proportion of CD31 positive cells from Control wounds displayed as singlets, indicative of the initial stage of angiogenesis. VEGF-A is one of the most important proangiogenic factors that promote the formation of new blood vessels [[107]33]. Therefore, we further determined the gene level of Vegfa in wound tissues on day 7 post-injury. As expected, Vegfa expression was remarkably increased in the LGG-EVs group (Fig. [108]3F). Hypoxia-inducible factor 1α (HIF1α), a transcription factor, plays a crucial role in VEGF expression and angiogenesis during wound healing [[109]34]. Given the enhanced angiogenesis and Vegfa expression, we next investigated the influence of LGG-EVs on the expression of HIF1α. qRT-PCR results showed that the LGG-EVs group exhibited markedly higher Hif1a expression than the Control group (Fig. [110]3F). Taken together, our findings suggest that LGG-EVs accelerated the wound healing program, both in terms of cell proliferation and angiogenesis. Fig. 3. [111]Fig. 3 [112]Open in a new tab LGG-EV application enhances wound cell proliferation and angiogenesis. A Immunofluorescent images of Ki67 (red staining) and DAPI (blue) in the wounds on day 7 (n = 5). Scale bar = 100 μm. B, C Quantification of Ki67 positive cells in the basal layer of epidermis (B), and dermis (C). D Immunofluorescent images of CD31 (red) and DAPI (blue) in the wounds on day 7 (n = 5). Scale bar = 100 μm. E Quantification of CD31 positive area in the wounds. F RT-qPCR analysis of Hif1α and Vegfa in the wounds on day 7(n = 5). gt, granulation tissue LGG-EVs enhance the proliferation and migration of endothelial cells and keratinocytes Wound healing is an intricate process, which relates to the interplay of different cells, such as endothelial cells and keratinocytes [[113]32]. Given that endothelial cells and keratinocytes are key to angiogenesis and re-epithelization, respectively, we tested the effect of LGG-EVs on these cells. To this end, we first asked whether LGG-EVs could be taken by endothelial cells (HUVEC) and keratinocytes (HaCaT). LGG-EVs were labeled with Cy5, and subsequently added to both cells. The internalization of LGG-EVs was observed 6 h after treatment. Fluorescence microscopy analysis showed that red fluorescence-labeled EVs had been largely internalized by HUVEC (Fig. [114]4A) and HaCaT (Fig. [115]4B). Fig. 4. [116]Fig. 4 [117]Open in a new tab LGG-EVs enhance the proliferation and migration of endothelial cells and keratinocytes. A, B Internalization of LGG-EVs labeled with Cy5 (red) by HUVEC (A) and HaCaT (B). Scale bar = 25 μm. C, D Representative images of Ki67 (green) immunofluorescence staining in HUVEC (C) and HaCaT (D) with or without LGG-EVs treatment for 24 h (n = 3). Scale bar = 50 μm. E, F Scratch wound-healing assay examining the migratory abilities of HUVEC (E) and HaCaT (F) after exposure to LGG-EVs or Control vehicle in starvation medium for 24 h and 48 h, respectively (n = 3). Scale bar = 200 μm. G Transwell assay examining the migratory abilities of HUVEC after exposure to LGG-EVs or control vehicle for 24 h. Quantification of the corresponding migration cells (right, n = 3). Scale bar = 200 μm. H Tube formation of HUVEC after exposure to LGG-EVs or control vehicle for 6 h (n = 3). Scale bar = 200 μm. Quantitative analysis of the number of meshes (right) using Image J In response to injury, endothelial cells and keratinocytes undergo a series of cellular activities, including proliferation and migration to support healing processes [[118]32]. Hence, we next investigated the effect of LGG-EVs on the functions of endothelial cells and keratinocytes via proliferation assay and mobilization assay. Ki67 staining assay demonstrated that the proliferation of HUVEC (Fig. [119]4C) and HaCaT (Fig. [120]4D) was significantly increased after exposure to LGG-EVs. Analysis of the mobilization ability of cells using scratch assays and transwell assays displayed that compared with that of Control, LGG-EVs enhanced the closure of cell-free gaps of HUVEC (Fig. [121]4E) and HaCaT (Fig. [122]4F), and migration of HUVEC (Fig. [123]4G). Furthermore, we also examined the vessel-forming ability of HUVECs using tube formation assay. As shown in Fig. [124]4H, compared to the Control, LGG-EVs exhibited a significantly increased number of meshes, suggesting that LGG-EVs promoted angiogenesis. In conclusion, these results indicate that LGG-EVs can endow keratinocytes and endothelial cells with enhanced capabilities, including proliferation, migration, as well as vessel formation for endothelial cells. Profiling of LGG-EV miRNAs Although EVs derived from other bacteria have been demonstrated to regulate the wound healing process [[125]13], their effective contents remain to be elucidated. The EV cargos consist of various miRNAs, proteins, lipids, and other nucleic acid compositions. Among them, miRNA is one of the most abundant, and many of the biological activities of EVs have been considered to be related to miRNAs [[126]35]. To investigate the key miRNAs in LGG-EVs contributing to wound healing, miRNA sequencing was conducted. miRNA identification was performed based on miRBase and the genome and EST database. A total of 494 miRNAs have been identified, including 193 known miRNAs and 301 novel miRNAs. Heatmap showed that many miRNAs related to cell proliferation, migration, and angiogenesis (miR-21-5P, miR-25-3p, miR-92a-3p etc.) were highly expressed in LGG-EVs (Fig. [127]5A). In addition, KEGG pathway enrichment analysis and GO analysis of these miRNAs were performed to verify the pathways and biological functions potentially involved. KEGG analysis displayed that miRNAs were engaged in signal transduction of PI3K-AKT, metabolic, HIF-1, and VEGF pathways (Fig. [128]5B), all of which were reported to be associated with wound closure [[129]36–[130]38]. In terms of biological process (BP), miRNAs were mainly involved in cell proliferation, cycle and migration, angiogenesis, cell differentiation, and extracellular matrix organization (Fig. [131]5C). As for cellular component (CC), miRNAs participated in regulating membrane composition, cytosol, nucleus, and cell junction (Fig. [132]5D). In molecular function (MF) analysis, they were associated with the binding of proteins, nucleotide, ATP, and cadherin, as well as kinase activity (Fig. [133]5E). These results were consistent with the pro-healing effect of LGG-EVs observed in vivo and in vitro. Fig. 5. [134]Fig. 5 [135]Open in a new tab Profiling of LGG-EV miRNAs. A Heatmap showing the top miRNAs in LGG-EVs. n = 3. B KEGG pathway enrichment analysis of the top miRNAs in LGG-EVs. C-E GO analysis of the top miRNAs of LGG-EVs in BP, CC, MF. F The expression of five key miRNAs in LGG-EVs. G The predicted key target gene network of the five key miRNAs in LGG-EVs. KEGG: Kyoto Encyclopedia of Genes and Genomes; BP, biological process; CC, cellular component; MF, molecular function Of importance, there are multiple miRNAs involved in cell proliferation, migration, and angiogenesis among the top 20 miRNAs, such as let-7i-5p [[136]39, [137]40], miR-21-5p [[138]41–[139]43], miR92a-3p [[140]44, [141]45], miR-122-5p [[142]46, [143]47], and miR182-5p [[144]48, [145]49] (Fig. [146]5F). Cytoscape was used to visualize the target genes and networks of these five miRNAs (Fig. [147]5G and Supplementary Fig. [148]4A). Of importance, the network revealed multiple target genes associated with energy metabolism, such as PDK4, SLC2A3, LAMP2 and CDK13 [[149]50–[150]53], indicative the relationship between LGG-EV targets with metabolism. Furthermore, GO and KEGG analysis of these five miRNAs were also performed. KEGG analysis also showed that these five miRNAs were enriched in PI3K-AKT, HIF-1, MAPK, and metabolic signaling pathways (Supplementary Fig. [151]4B), which is consistent with cytoscape analysis. GO analysis suggested that these five miRNAs contributed to positive regulation of transcription, protein and cell differentiation (Supplementary Fig. [152]4C), nucleus, an integral component of membrane, cytosol, and Golgi apparatus in the category of CC (Supplementary Fig. [153]4D), and the binding of protein, nucleotide, and ATP, as well as hydrolase activity and DNA transcription factor (Supplementary Fig. [154]4E). Collectively, the results revealed that the miRNA cargo in LGG-EVs has the potential to benefit wound healing. Of note, the miRNA database associated with BEVs is currently deficient, and deserves further investigation. LGG-EVs shuttle miR-21-5p into endothelial cells and keratinocytes Having demonstrated the miRNA contents of LGG-EVs, we next investigated the functional miRNAs that mediate the pro-wound healing effects. Since miR-21-5p displayed the second highest expression level and had positive roles in regulating cell functions and angiogenesis [[155]41–[156]43], all of which give rise to the pro-healing properties, we thus selected miR-21-5p as a possible candidate miRNA. We first confirmed the transfer of miR-21-5p into target cells. HUVEC and HaCaT were administered with LGG-EVs for 6 h followed by qRT-PCR analysis. In both cells, the expression of miR-21-5p was markedly increased after exposure to LGG-EVs (Fig. [157]6A and B). These data demonstrated that miR-21-5p can be shuttled by LGG-EVs to recipient cells to regulate gene expression. Fig. 6. [158]Fig. 6 [159]Open in a new tab LGG-EVs activate the functions of endothelial cells and keratinocytes via transferring miR-21-5p. A, B The expression of miR-21-5p in HUVEC (A) and HaCaT (B) after treatment with or without LGG-EVs for 6 h (n = 3). C, D Representative images of Ki67 (green) and DAPI (blue) immunofluorescence staining. HUVEC (C) and HaCaT (D) were pre-transfected with miRi for 24 h, followed by treatment with LGG-EVs or PBS (Control) for 24 h (n = 3). Scale bar = 50 μm. E-H Scratch wound-healing assay examining the migration. HUVEC (E) or HaCaT (G) was transfected with miRi for 24 h, followed by exposure to LGG-EVs or vehicle control for 24 h or 48 h, respectively (n = 3). Scale bar = 200 μm. F, H Quantification of cell migration ratio in E and G, respectively. miRi: miR-21-5p inhibitor LGG-EVs activate the functions of endothelial cells and keratinocytes via transferring miR-21-5p To investigate if the pro-healing effects of LGG-EVs were mediated by miR-21-5p, we transfected a miRi that specifically reduces miR-21-5p before LGG-EVs treatment in HUVEC and HaCaT. LGG-EVs promoted cell proliferation and this effect was abolished by miRi as judged by Ki67 staining (Fig. [160]6C and D, Supplementary Fig. [161]5). Furthermore, after miR-21-5p inhibition, the pro-migratory effects of LGG-EVs were attenuated, though not completely abolished in both cells (Fig. [162]6E-H). Intriguingly, LGG-EVs supplemented with miRi still displayed a higher migration ratio than control group (Fig. [163]6E-H), indicating that EVs have complex contents and other cargos in EVs may also contribute to cell migration. These findings showed that the effects of LGG-EVs on endothelial cells and keratinocytes were, at least in part, mediated through miR-21-5p. The activation of the AKT/HIF1α pathways is involved in response to LGG-EV stimulation We next asked which signaling pathway was affected by LGG-EVs during wound healing. The wound healing process is associated with the activation of various signaling pathways, such as PI3K-AKT and HIF1α [[164]36, [165]37]. PI3K-AKT and HIF1α are the key pathways involved in keratinocyte proliferation and migration, as well as angiogenesis. Previous reports also suggested that miR-21-5p might modulate migration through AKT signaling by targeting PTEN, a known negative regulator of PI3K-AKT and HIF1α signaling pathways [[166]54, [167]55]. Combining with above KEGG analysis (Fig. [168]5B), we hence investigated the influence of LGG-EVs on these two pathways. As shown in Fig. [169]7, LGG-EVs remarkably increased the levels of phosphorylation of AKT (p-AKT), an indicator of PI3K-AKT activation, and HIF1α in HUVEC (Fig. [170]7A-C) and HaCaT (Fig. [171]7D-F). We also found that the miRi abolished the activation induced by LGG-EVs (Fig. [172]7A-F). Therefore, LGG-EVs enhanced the function of endothelial cells and keratinocytes by miR-21-5p mediated PI3K-AKT/HIF1α signaling activation. Fig. 7. [173]Fig. 7 [174]Open in a new tab The activation of the AKT/HIF1α pathways is involved in response to LGG-EV stimulation. A-F Western Blot analysis of HIF1α, AKT, and p-AKT levels. HUVEC (A-C) and HaCaT (D-F) were transfected with miRi for 24 h, followed by exposure to LGG-EVs or PBS (Control) for 24 h (n = 3). miRi, miR-21-5p inhibitor Discussion Despite considerable research efforts, timely wound healing or regeneration remains an area of highly unmet medical need that requires novel therapeutics. In this study, we investigated the therapeutic effects and potential mechanisms of EVs released by probiotic LGG on cutaneous wound healing. We provided evidence for the first time that LGG-EVs facilitated wound closure, improved pro-healing-associated biomarkers, including enhanced angiogenesis and re-epithelialization capacities. Furthermore, we also revealed that LGG-EVs can be internalized by endothelial cells and keratinocytes, activate the PI3K-AKT and HIF1α signaling pathways, and enhance cell proliferation and migration as well as angiogenesis properties by shuttling miR-21-5p. In recent years, the crosstalk between gut microbiota and skin has received significant attention from scientists and clinicians [[175]56, [176]57]. Particularly, the gut microbiome participates in regulating homeostasis and pathogenesis via “gut-skin-axis” [[177]57]. Clinical trials have shown promising results with improved skin conditions following oral and topical probiotic interventions [[178]57, [179]58], indicating the potential of probiotics in dermatology. EVs are lipid bilayer particles loaded with bioactive molecules that transfer biological signals between local or distant organs. Interestingly, emerging evidence supports that EVs from gut bacteria function as mediators, facilitating the transport of bioactive components into the skin, regulating various biological processes, and exhibiting therapeutic effects in the disease therapy field [[180]59]. For instance, Wu et al. showed that Lactobacillus druckerii-derived EVs can accelerate wound closure as well as inhibit scar formation [[181]13], conferring regenerative capability. Fuhrmann et al. demonstrated that EVs from Lactobacilli increased cell migration and inhibited inflammation in vitro, thus improving cutaneous wound healing [[182]60]. Lactobacillus plantarum-derived EVs protect against ischemic brain injury [[183]61]. Interestingly, in a very recent study, Kuhn et al.. embedded EVs from Lactobacillus plantarum and Lactobacillus casei, respectively, into hydrogel and furtherly demonstrated their anti-inflammatory effects in vitro and capability to accelerate wound healing in vivo [[184]62]. However, to our knowledge, no study has reported the effects and underlying mechanisms of LGG-EVs concerning wound healing. In our study, we found that LGG-EVs can promote cutaneous wound healing, as evidenced by faster wound closure, higher rates of re-epithelialization, and more vessel formation. Therefore, we propose that gut probiotics-derived EVs may be an innovative therapeutic strategy in regenerative medicine, deserving further investigation. Notably, several EVs have entered into clinical trials (ClinicalTrials.gov.), underscoring their significant potential. However, these EVs are typically derived from mammalian cells, which face challenges such as poor and slow yield, high economic investment, and medical ethics concerns. BEVs can overcome these limitations as they are easily accessible and produced by fed-batch fermentation protocols. Importantly, manipulation of bacterial EVs is feasible, and this work opens new possible areas of study, such as engineered LGG-EVs to enhance therapeutic effects. BEVs also encapsulate novel bioactive molecules to regulate recipient cells [[185]11]. Among the most abundant loads of EVs, miRNAs play an important role in regulating biological activities by transferring them into recipient cells. Our miRNA sequencing revealed that miRNAs associated with cell proliferation, migration, and angiogenesis were enriched in LGG-EVs (Fig. [186]5). In line with previously reported engineered LGG-EVs [[187]25], we observed that miR-21-5p was also enriched in naïve LGG-EVs, with the second-highest expression level in our assay. This supports the notion that physicochemical modifications of EVs did not significantly change their components [[188]63]. In addition, while this study predominantly concentrates on miRNAs, we recognize the potential impact of other LGG-EV cargos. Future investigations are needed to provide a comprehensive and in-depth analysis of the bioactive components encapsulated in LGG-EVs and their respective functions. miR-21-5p may be the key regulator in LGG-EVs associated with the improved functions of keratinocytes and endothelial cells [[189]41–[190]43]. LGG-EVs contained abundant miR-21-5p. After incubation of LGG-EVs for a short time (6 h), the expression of miR-21-5p was significantly increased in those two cells, indicating that miR-21-5p can be shuttled from LGG-EVs to recipient cells. By using specific inhibitor of miR-21-5p, we were able to identify the therapeutic effects, as observed that inhibition of miR-21-5p attenuated LGG-EVs effects on endothelial cells and keratinocytes, thereby highlighting the key role of miR-21-5p in LGG-EVs. miR-21-5p is a vascular-specific member of the family of miRNA precursors that mediates angiogenesis, invasion, and metastasis of tumors, including skin tumors [[191]64]. Intriguingly, miR-21 expression is increased after skin injury [[192]65], while the expression of miR-21-5p is lower in diabetic mouse skin wounds [[193]66], suggesting its potential role in chronic wounds. Therefore, further study is needed to investigate the role of miR-21-5p and LGG-EVs in diabetic wounds and impaired healing conditions. In addition, multiple studies also have investigated the beneficial effects of miR-21-5p on the functional properties of endothelial cells, fibroblasts, and tumor cells [[194]41–[195]43]. For example, miR-21-5p has been reported to promote the proliferation and migration of endothelial cells and fibroblasts in vitro and angiogenesis in tumor microenvironment [[196]41–[197]43]. In normal and keloid keratinocytes, miR-21-5p overexpression induced the functional activity of these keratinocytes [[198]54]. However, few studies have illustrated the functions of miR-21-5p in wound re-epithelialization and revascularization. Our findings further consolidated that miR-21-5p up-regulation may promote wound healing. Multiple signaling pathways have been involved in wound healing processes. KEGG analysis of key pathways associated with miRNAs in LGG-EVs demonstrated the enrichment in PI3K-AKT and HIF1α pathways, both of which are required for timely wound healing. Consistent with the finding, LGG-EVs upregulated HIF1α and p-AKT, indicating the activated PI3K-AKT and HIF1α signals and miR-21-5p inhibition attenuated the activation effect, which means that LGG-EVs regulated the functional properties, at least in part, by inducing PI3K-AKT and HIF1α activity via miR-21-5p. Conclusion In summary, we demonstrate that probiotic LGG-derived EVs are enriched with miR-21-5p, which enhance the functions of endothelial cells and keratinocytes by metabolic signaling rewiring, and subsequently promote re-epithelization and angiogenesis, thereby accelerating cutaneous wound healing. Our findings support the intriguing hypothesis that probiotic EVs-mediated signals can promote and facilitate the restoration of tissue integrity after injury in various aspects. Future experiments will determine whether these EVs can facilitate wound healing in pathological conditions and ultimately expand the therapeutic benefits, potentially leading to their application in clinical settings. Moreover, further pre-clinical in vivo studies are necessary to elucidate the innovative mechanisms of the signal exchange between probiotics and skin, specifically focusing on gut microbiota-derived EVs. Electronic supplementary material Below is the link to the electronic supplementary material. [199]Supplementary Material 1^ (3.3MB, docx) Acknowledgements