Abstract Extracellular vesicle (EV)–based immunotherapeutics have emerged as promising strategy for treating diseases, and thus, a better understanding of the factors that regulate EV secretion and function can provide insights into developing advanced therapies. Here, we report that nutrient availability, even changes in individual nutrient components, may affect EV biogenesis and composition of immune cells [e.g., macrophages (Mφs)]. As a proof of concept, EVs from M1-Mφ under glutamine-depleted conditions (EV^GLN−) had higher yields, functional compositions, and immunostimulatory potential than EVs from conventional GLN-present medium (EV^GLN+). Mechanistically, the systemic metabolic rewiring (e.g., altered energy and redox metabolism) induced by GLN depletion resulted in up-regulated pathways related to EV biogenesis/cargo sorting (e.g., ESCRT) and immunostimulatory molecule production (e.g., NF-κB and STAT) in Mφs. This study highlights the importance of nutrient status in EV secretion and function, and optimizing metabolic states and/or integrating them with other engineering methods may advance the development of EV therapeutics. __________________________________________________________________ Metabolic rewiring by fine-tuning nutrient condition is a promising strategy for bioengineering EVs as therapeutics in diseases. INTRODUCTION Extracellular vesicles (EVs) are cell-secreted nanovesicles enclosed by lipid bilayer membranes that serve as key mediators of cell-cell communication ([44]1–[45]3). Immune cell–derived EVs play critical roles in regulating innate and adaptive immune processes (e.g., immune signaling and antigen presentation) by transferring diverse types of cargos (e.g., nucleic acids, proteins, and lipids) between cells. In recent years, the use of EV-based immunotherapies has emerged as a promising strategy for treating various immune-related diseases, such as autoimmune or inflammatory diseases, vaccinations, and cancers ([46]1, [47]4). For example, M1-like macrophage (Mφ)–derived EVs (M1-EVs) can enhance the effectiveness of cancer vaccines and chemotherapeutics ([48]5, [49]6). As cell-derived natural nanovesicles, EVs offer several advantages over other synthetic nanocarriers, such as lower toxicity, higher biocompatibility, and the ability to cross biological barriers. Currently, in vitro cell culture–based methods are commonly used to produce therapeutic EVs. However, the clinical translation of EV therapies is largely hindered by their low yield and unsatisfactory bioactivity ([50]3, [51]7). Although previous studies have explored methods (e.g., exogenous physical or drug stimuli) for increasing EV secretion or cargo levels to some extent ([52]7), the efficient production of highly bioactive EVs remains an enormous challenge in this field. A better understanding of the factors influencing EV secretion, cargo composition, and function can facilitate the manipulation EVs to improve their therapeutic potential. Thus, it is important to explore the specific factors that regulate EV biogenesis and the function of immune cells. Nutrients are indispensable substrates for supporting cell growth and various cellular processes both in vitro and in vivo. There is an abundant amount of studies indicating that metabolic status has a substantial impact on the phenotypes and functions of immune cells (e.g., Mφs and T cells). Immune cell behavior is influenced by changes in bioenergetic and material demands, including glucose (GLC), amino acids (AAs), and lipids ([53]8). For example, immunostimulatory M1-like Mφs (M1-Mφs) heavily rely on aerobic glycolysis while exhibiting a disrupted tricarboxylic acid (TCA) cycle to support rapid cell proliferation and cytokine production ([54]9). Immunosuppressive M2-like Mφs (M2-Mφs) exhibit an intact TCA cycle and predominantly use oxidative metabolism to regulate immune responses and promote tissue repair ([55]9). Additionally, α-ketoglutarate [αKG; mainly derived from glutamine (GLN)] deficiency can promote M1-Mφ polarization and enhance the production of inflammatory cytokines [e.g., interleukin-1β (IL-1β), IL-6, and tumor necrosis factor–α (TNF-α)] by activating the prolyl hydroxylase domain–mediated nuclear factor κB (NF-κB) pathway ([56]10). Several recent studies have shown that certain nutrient conditions, such as serum or GLC starvation, can induce EV release and/or alter EV compositions in different cells ([57]2, [58]7). For example, serum starvation for 20 hours can stimulate monocytes (THP-1 cells) to release more EVs containing TSG101 and active tissue factor than conventional culture conditions, and such EVs can promote endothelial thrombogenicity and apoptosis in human umbilical vein endothelial cells ([59]11). Additionally, cardiomyocytes subjected to GLC starvation for 48 hours secreted larger amounts of EVs that can stimulate the proliferation and angiogenesis of endothelial cells, and these effects may be due to the enrichment of EV proteins and microRNAs (miRNAs) related to metabolic regulation ([60]12). These findings suggest that changes in nutrient availability may affect the biogenesis and composition of EVs. However, the nutrient conditions and underlying mechanism that can enhance the yield and bioactivity of EVs in immune cells have not been fully elucidated. Here, we report that the nutrient state regulates both the biogenesis and function of EVs in Mφs and that fine-tuning the nutrient state is a potent strategy for bioengineering immune cell–derived EVs. Briefly, changes in individual nutrient components [e.g., GLC, AAs, and fatty acids (FAs)] in the medium altered the secretion of EVs from Mφs to some extent. As a proof of concept, GLN-depleted (GLN^−) conditions increased the yield, functional content, and immunostimulatory potency of EV preparations from M1-Mφs. GLN^− conditions up-regulated pathways related to EV biogenesis/cargo sorting and immunostimulatory factor production in M1-Mφs, and these effects were largely dependent on systemic metabolic rewiring of donor cells in response to the GLN^− state. In summary, these results highlight the importance of metabolic regulation in EV biogenesis and immune cell function and suggest that fine-tuning metabolic states represents a powerful strategy for engineering immune cell–derived EVs with enhanced therapeutic potential. RESULTS AND DISCUSSION Changes in nutrient conditions affect EV secretion in Mφs Increasing evidence indicates that the availability of nutrients in the environment can affect the metabolism and function of immune cells, and the most important nutrients include GLC, AAs, FAs, and vitamins ([61]8). Mφs are primary innate immune cells involved in various immune actions, such as defense against pathogens/danger signals, inflammation, and tissue repair ([62]3). Additionally, the diverse functions of Mφ-derived EVs (Mφ-EVs) are involved in the pathology of many diseases, such as infections, autoimmune disorders, and cancers ([63]3). Here, Mφs were selected as an example to explore the impacts of nutrient conditions on EV production in immune cells. EVs were isolated from the conditioned medium of mouse Mφs (RAW 264.7) using an ultracentrifugation method as previously reported ([64]13) ([65]Fig. 1A). The resulting Mφ-EV preparations exhibited a typical bilayer spherical structure with a size of approximately 150 nm, and they were positive for EV markers such as endosomal sorting complex needed for transport (ESCRT)–associated proteins (e.g., TSG101), chaperone proteins [e.g., heat shock protein 70 (HSP70)], and intraluminal vesicle (ILV) formation-related proteins (e.g., ALIX) ([66]Fig. 1B). Moreover, there were no obvious signals of the negative marker GM130, a protein from the Golgi apparatus, in these EV samples ([67]Fig. 1B). These results indicate that our EV preparations fulfill the fundamental criteria outlined in the Minimal Information Guidelines for Extracellular Vesicle Studies (MISEV2018), which advise confirming the presence of at least two EV-positive markers (e.g., TSG101 and ALIX) and the absence of at least one EV-negative marker (e.g., GM130) and conducting supplementary morphological analyses (e.g., membrane structure and size) ([68]14). Fig. 1. Effect of nutrient availability on EV secretion in Mφs. [69]Fig. 1. [70]Open in a new tab (A) Schematic illustration of the EV isolation and characterization process. (B) Representative TEM images of EV preparations (left), size distributions of EVs measured by NTA (middle), and Western blot analysis of EV markers (ALIX, HSP70, TSG101, and GM130; right). (C) Schematic illustration of major cellular nutrient components. (D) EV sizes determined by NTA from Mφs cultured in control medium (nl) or modified medium supplemented with 2.5% FBS (lo), 20% FBS (hi), GLC (9 mM, hi), OA (100 μM, hi), BCAA (5 mM, hi), or GLN (8 mM, hi) for 24 hours. (E) Relative EV yields of the different groups were quantified by normalizing the EV protein mass to the cell protein mass. (F) Western blot analysis of the expression of EV markers (HSP70 and TSG101) in equal amounts of EV samples with equal protein amounts (10 μg per panel). (G) Quantification of the expression of EV-positive markers (HSP70 and TSG101) in various groups. BSA was used as a carrier to prepare the OA solution, while BSA alone served as the vehicle control (n ≥ 3; **P < 0.01, *P < 0.05, ^NSP ≥ 0.05 versus the “nl” group). We then investigated the detailed effects of altered nutrient additions (GLC, AAs, or FAs) on the secretion of EVs from M0-Mφs ([71]Fig. 1C). M0-Mφs were initially cultured in control medium (complete RPMI 1640, widely used for Mφ culture) containing 10% fetal bovine serum (FBS), 11 mM GLC, and 2 mM GLN, modified medium containing lower (2.5%) or higher (20%) FBS, higher GLC (20 mM), higher GLN (8 mM), branched-chain amino acid (BCAA; 5 mM) supplementation, or oleic acid (OA; 100 μM) supplementation for 24 hours. The supernatants from each culture condition were collected for EV preparation. Because of the base RPMI 1640 (without FBS) already containing GLC (11 mM) and GLN (2 mM), medium with no or low GLC/GLN could not be included in this step. The impact of an individual nutrient on EV production will be assayed in the next steps using a specific medium containing no GLC or GLN. The results showed that the EV preparations from the control and altered nutrient conditions had similar mean sizes (~150 nm) ([72]Fig. 1D). Each method offers unique benefits and limitations in quantifying EVs based on size, concentration, and other characteristics ([73]15). Here, EV yield was assessed using the most common method, that is, determining the ratio of the protein concentration of EVs to the protein concentration of donor cells ([74]16). The results showed that changes in the individual nutrient contents altered the EV yield of M0-Mφs to some extent ([75]Fig. 1, E to G). The lower FBS, BCAA supplementation, and higher GLN conditions significantly reduced the secretion of EVs from M0-Mφs, compared to that in the control conditions, while OA supplementation slightly increased the EV secretion of these cells ([76]Fig. 1E). Neither higher FBS nor higher GLC concentrations significantly altered EV secretion compared to that in the control group ([77]Fig. 1E). Further validation tests also verified the elevated levels of EV yield in M0-Mφs from the no GLC (0 mM) group or no GLN (0 mM) group compared to those from the GLC groups (2 and 10 mM) and GLN groups (2 and 10 mM), respectively (fig. S1). The levels of protein markers of EV preparations in the different groups were also examined using equal total protein amounts. Compared with the control group, the levels of HSP70 and TSG101 in the lower FBS group, higher GLN group, BCAA group, and OA group were lower, while the expression of HSP70/TSG101 in the higher GLC group was not significantly different ([78]Fig. 1, F and G). Rapamycin (Rapa) affects BCAA metabolism by inhibiting mechanistic target of rapamycin (mTOR) signaling, which in turn affects the activity of multiple enzymes (e.g., ribosomal protein S6 kinase-β1) involved in BCAA metabolism ([79]17), and thus, the effect of Rapa on EV secretion of M0-Mφ was also evaluated. The BCAA plus Rapa group showed lower levels of HSP70 and TSG101 than the BCAA alone group (fig. S2A) but did not affect EV yield (fig. S2B), suggesting that BCAA metabolism may change the EV composition in M0-Mφs, but the detailed reasons are needed to be further researched in the future. Collectively, these results suggest that changes in nutrient status can affect EV secretion by Mφs. Because of the high heterogeneity of EVs, specific markers for distinguishing EV subtypes via different biogenetic routes (e.g., exosomes and ecotosomes) are not yet available, making it difficult to identify EV subtypes in EV preparations ([80]1, [81]18). Instead, MISEV2018 and current reports suggest categorizing EV preparations by their size, such as small EVs (~50 to 150 nm), medium-sized EVs (~200 to 800 nm), and large EVs (≥1 μm) ([82]18). Here, the prepared Mφ-EVs were found to be mainly small EVs, and the altered EV secretion is likely an overall effect induced by cell metabolic alterations in response to different nutrient conditions. Overall, these results suggest that nutrient states affect EV secretion by Mφs to some extent. Therefore, it is crucial to explore the optimal nutrient conditions for the efficient production of therapeutic EVs in diverse immune cells. We then aimed to examine the detailed effects of GLN availability on EV production, as GLN is one of the most abundant AAs in the blood and various tissues and is essential for immune cell functions ([83]19). GLN-depleted conditions enhanced EV secretion in Mφs Since GLN availability markedly affects the EV yield of M0-Mφs ([84]Fig. 1, E to G, and fig. S1), as a proof of concept, we chose a challenging condition (GLN depletion, GLN^−) to assay the specific impact of GLN catabolism on Mφ-EV secretion and function, since this method has been used to assay the effect of GLN metabolism on Mφ phenotype switching ([85]10). GLN depletion can completely shut down intracellular GLN metabolic flux, which is essential for the carbon and nitrogen necessary for anabolic metabolism. Glutaminolysis converts GLN to glutamate [via glutaminase 1 (GLS1)] for the TCA cycle, and to glycine and other substrates crucial for nucleotide (e.g., DNA and RNA) synthesis ([86]20). Thus, the functions of diverse immune cells (e.g., Mφs and T cells) heavily rely on GLN availability and consumption ([87]10). The effect of GLN^− conditions on Mφ cell growth and cell viability was first evaluated. The cell growth rates of the GLN^− group were not affected at 16 hours, were slightly reduced at 24 hours, and significantly declined at 48 hours compared to those of the GLN^+ group (control group) (fig. S3A). Similarly, compared with those in the GLN^+ group, the percentage of live cells in the GLN^− group displayed a minor decrease (less than 5%) at 24 hours (fig. S3B). These results suggest that both the cell growth and cell viability of most Mφs are minimally affected by GLN depletion for 24 hours and that the increase in EV production in this state is primarily due to metabolic alteration; thus, these conditions were chosen for the following experiments. Next, M0-Mφs were cultured in medium supplemented with (GLN^+, 2 mM) or without GLN (GLN^−) for 24 hours, after which resulting EVs were isolated and assayed ([88]Fig. 2A). EV preparations from the GLN^− and GLN^+ conditions exhibited similar characteristics in terms of mean size (~150 nm) ([89]Fig. 2B) and bilayer membrane structure (fig. S4A). Moreover, the EV yield (as indicated by the ratio of EV protein mass/cell protein mass or EV number/cell protein mass) of the GLN^− group was higher (~1.5-fold) than that of the GLN^+ group ([90]Fig. 2C). The levels of EV marker expression (HSP70, TSG101, and ALIX) were also higher in the GLN^− group than in the GLN^+ group ([91]Fig. 2, D and E), indicating that GLN^− conditions can promote the secretion of EV from M0-Mφs. To validate these results, M0-Mφs were treated with BPTES (a GLS1 inhibitor) in the presence of GLN ([92]Fig. 2F). Consistently, BPTES treatment also increased the EV yield (~1.4-fold increase in the EV number/cell protein mass ratio and ~2-fold increase in the EV protein mass/cell protein mass ratio) of M0-Mφ without altering the EV size distribution ([93]Fig. 2, G and H). Fig. 2. Effect of GLN conditions on Mφ-derived EV secretion. [94]Fig. 2. [95]Open in a new tab (A and B) Schematic illustrating GLN^− experiments in M0-Mφs and the sizes of EVs from M0-Mφs cultured with GLN (GLN^+, 2 mM) or without GLN (GLN^−) for 24 hours. (C) EV yield of different groups determined by the EV number/cell protein ratio (left) or the EV protein/cell protein ratio (right). (D and E) Western blot analysis and the expression of EV markers (HSP70, TSG101, and ALIX) in equal amounts of EVs (10 μg per panel, n = 4). (F and G) Schematic map of the GLN catabolism inhibition experiment and EV sizes from M0-Mφs cultured in GLN^+ medium with or without BPTES (10 nM) treatment for 24 hours. (H) EV yield of different groups determined by the EV number/cell protein ratio (left) or the EV protein/cell protein ratio (right). (I and J) Schematic illustrating GLN^− experiments in M1-Mφs and EV sizes from M1-Mφs cultured with GLN (GLN^+, 2 mM) or without GLN (GLN^−) for 24 hours. (K) EV yield of different groups determined by the EV number/cell protein ratio (left) or the EV protein/cell protein ratio (right) (n = 3; relative to the “GLN^+” group; ***P < 0.001, **P < 0.01, *P < 0.05, ^NSP ≥ 0.05 versus the GLN^+ group). Depending on the different phenotypes of the donor Mφs, Mφ-EVs may have stimulatory or inhibitory effects on immune function. For example, M1-EVs serve as immune booster agents that enhance antitumor efficacy ([96]21), while M2-EVs play an immunosuppressive role in attenuating inflammation ([97]13). Thus, the impact of GLN^− conditions on EV secretion of M1-Mφ was assessed, as GLN metabolism can support Mφ activation and desirable immune responses ([98]10). Lipopolysaccharide (LPS)–induced M1-Mφs were cultured in GLN-free RPMI 1640 supplemented with (GLN^+, 2 mM) or without GLN (GLN^−) for 24 hours ([99]Fig. 2I). EVs from both conditions exhibited comparable size distributions (~150 nm) and bilayer membrane structures (fig. S4B). The EV yield was significantly higher in the GLN^− group than in the GLN^+ group (~3-fold increase in the EV number/cell protein mass ratio, and ~2-fold increase in the EV protein mass/cell protein mass ratio) ([100]Fig. 2, J and K). These results collectively showed that limiting GLN availability (or inhibiting glutaminolysis) is sufficient to enhance EV production in both M0 and M1 Mφs. Previous studies have shown that M1-EVs exert superior therapeutic effects, such as cancer therapy, compared to M0-EVs, and this effect may be due to their distinct compositions ([101]3). For example, M1-EVs could move to lymph nodes and are mainly taken up by Mφs and dendritic cells, which subsequently enhances the cytotoxic T cell–mediated anticancer immune response in vivo ([102]5). Moreover, our results showed that there was no difference in EV yield between the M0-Mφ group and the M1-Mφ group under normal culture conditions (fig. S5). Considering the importance of future clinical translation, we therefore focused on M1-EVs due to their superior therapeutic effects (e.g., cancer therapy and anti-infections). GLN^− conditions enriched the functional composition of Mφ-EVs Immune cell–derived EVs play crucial roles in modulating immune functions for possible therapeutic use, and they affect the functions of target cells by delivering diverse surface molecules (e.g., proteins and lipids) and/or encapsulated cargos (e.g., proteins and RNAs) ([103]1). EVs can inherit biological signals from donor cells; thus, the functions of immune cell–derived EVs can vary due to their diverse compositions from donor cells under different pathological conditions ([104]18). For instance, Mφ-EVs from the early stage of sepsis displayed immunostimulatory effects (e.g., promoting leukocyte chemotaxis). This effect is associated with enriched cytokines and damage-associated molecular patterns (DAMPs), such as histones, high-mobility group box 1 (HMGB1), and HSPs. However, Mφ-EVs from the later stage of sepsis can also exert immunosuppressive effects (e.g., reducing serum proinflammatory cytokine levels and the expression of adhesion molecules on endothelial cells) due to the expression of CD14 [glycosylphosphatidylinositol (GPI)–anchored protein] ([105]22). These studies suggest that EV compositions (e.g., proteins) are strongly associated with their function, and globally profiling EV contents can provide insights into their biological roles. Thus, in addition to the increase in EV yield, we investigated whether GLN^− conditions can alter the composition of Mφ-EVs using liquid chromatography–tandem mass spectrometry (LC-MS/MS)–based proteomics ([106]Fig. 3A). Following a strict data quality process, we identified a total of 2282 proteins in our EV preparations (table S1). The principal components analysis (PCA) scatterplot and heatmap showed clear separation and an overall differential protein expression pattern between the EV^GLN+ group and the EV^GLN− group ([107]Fig. 3, B and C). A volcano plot identified 692 differentially expressed proteins (DEPs; P-adjusted < 0.05) in the EV^GLN− group compared to those in EV^GLN+ group ([108]Fig. 3D). The Gene Ontology (GO) enrichment analysis revealed that these DEPs were mainly enriched in pathways related to immune processes, such as regulation of the inflammatory response (e.g., Ddt, Tnfaip8l2, Lpl, IL-1β, and Ptgs2), cytokine production (e.g., Cdk9, Dek, Ddt, and Lmnb1), positive regulation of T cell activation (e.g., IL-1β, Cd81, and Igfbp2), and cell-cell adhesion (e.g., Tnfaip8l2, IL-1α, and Alox5) ([109]Fig. 3, E and F), suggesting that EV^GLN− may function in stimulating the immune response. Fig. 3. Effect of GLN conditions on M1-EV composition. [110]Fig. 3. [111]Open in a new tab (A) Schematic illustrating the EV proteomics experiments. (B) PCA scatterplot of different groups based on proteomic data showing the differences between different groups (n = 3). (C) Heatmap displaying variations in protein composition between the EV^GLN+ group and the EV^GLN− group (n = 3). (D) Volcano plots showing the DEPs (FC > 1.5 and P-adjusted < 0.05) between the EV^GLN+ group and the EV^GLN− group (n = 3). (E and F) GO enrichment analysis and interaction network analysis of DEPs in response to GLN conditions (FC > 1.5). (G) Heatmap illustrating specific immune processes related to DEPs in different groups (FC > 1.5). (H) qPCR analysis of cytokine (IL-1β, IL-6, and IL-10) gene expression in M1-Mφs or M1-EVs from the GLN^+ or GLN^− group (n = 3; ***P < 0.001, **P < 0.01, *P < 0.05, ^NSP ≥ 0.05 versus the GLN^+ group). To further uncover the potential immunoregulatory molecules enriched in the EV^GLN− preparations, the DEPs identified by GO analysis were subjected to hierarchical clustering analysis. As depicted in [112]Fig. 3G, compared with those in the EV^GLN+ preparations, EV^GLN− preparations exhibited higher levels of several immunoregulatory proteins, such as IL-1α, IL-1β, CDK9, Alox5, and Tnfaip8l2. Multiple molecules known to participate in immune regulation, which are involved in various immune processes, such as influencing cytokine production, orchestrating inflammatory responses, and facilitating innate immunity, were identified in EV^GLN− preparations. For example, cytokines (e.g., IL-1α and IL-1β) have been shown to induce M1-Mφ polarization and cytokine production by activating the NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways ([113]23). IL-1 can boost T cell proliferation by up-regulating IL-2 receptor (IL-2R) expression and inducing the NF-κB and phosphoinositide 3-kinase pathways ([114]24). IL-1β was shown to prompt chemokine secretion in keratinocytes and the subsequent recruitment of proinflammatory neutrophils, thus initiating a robust innate immune response to pathogens ([115]25). In addition to proteins, several other types of EV cargos, such as RNAs, may also contribute to the immunoregulatory roles of EVs. Thus, the changes in mRNAs related to immune action were also analyzed in donor cells and their EVs. Similarly, the results showed that both M1-Mφs and EVs from the GLN^− group had higher mRNA levels of immunostimulatory cytokines (e.g., IL-1β and TNF-α) and lower levels of immunosuppressive cytokines (e.g., IL-10) than those from the GLN^+ group ([116]Fig. 3H), suggesting that GLN^− conditions may alter the levels of diverse types of EV, such as proteins and RNAs. Overall, these results indicate that GLN^− conditions can promote the secretion and sorting of diverse immunostimulatory molecules into EVs, and that these EVs may serve as advanced therapeutics for immunoregulation. GLN^− conditions enhanced the immunostimulatory effects of Mφ-EVs Increasing evidence indicates that immune cell–derived EVs can be applied to modulate immune responses and address various immune-related diseases due to their ability to either initiate or suppress the immune system. For example, M1-EVs modified with NF-κB p50 small interfering RNA (siRNA) reduced the levels of M2 markers and cytokines (Arg1, TFG-β, IL-10, and IL-4) while increasing the levels of M1-related cytokines (IL-12p40 and IFN-γ), and thus enhanced antitumor immunity in a breast tumor model ([117]21, [118]26). The previous proteomic results ([119]Fig. 3, E to H) indicated that the EV^GLN− protein profile was highly enriched in immunostimulatory pathways (e.g., cytokine production, positive regulation of T cell activation, and cell-cell adhesion), indicating the potential for enhanced immune activation via these EVs. Having identified the enrichment of immunostimulatory molecules in M1-EVs, we sought to determine whether such EVs can be efficiently taken up by targeted cells to shed their cargos ([120]1). To assess the cellular uptake of EVs, purified DiD (a lipophilic fluorescent dye)–labeled EV^GLN− (DiD-EVs) were incubated with adherent Mφs (RAW 264.7) or suspension monocytes (THP-1) for 4 hours. Compared with those in the phosphate-buffered saline (PBS) (solvent control) or the Dye control group, much higher levels of DiD-positive signals were observed in Mφs or monocytes using flow cytometry analysis (FCA) (fig. S6), which indicated that EVs can be efficiently taken up by diverse immune cells. Systematically injected (intravenous) EVs are primarily taken up by innate immune cells (e.g., Mφs and monocytes), which subsequently recruit and activate many other cells ([121]1, [122]13). To better mimic this situation in vitro, we first investigated the immunomodulatory effects of such EVs on innate immune cells (e.g., Mφs and monocytes). EV^GLN+ or EV^GLN− was added into M2-Mφs induced by IL-4/transforming growth factor–β (TGF-β), and the results showed that both treatments notably reduced the expression of M2-related genes (e.g., Arg1, Mrc1, and TGF-β) in M2-Mφs ([123]Fig. 4, A and B). EV^GLN+ or EV^GLN− was also added to monocytes, and we found that EV^GLN− had higher potency to induce chemokine (e.g., Ccl2 and Cxcl2) expression than EV^GLN+ in monocytes ([124]Fig. 4, C and D). Consequently, supernatants (containing various chemokines and cytokines) from EV^GLN−-treated monocytes had a higher potential to induce chemotaxis in mouse splenocytes compared to those in the EV^GLN+ group ([125]Fig. 4E). Mammalian splenic tissue is rich in functional immune cells. In addition, compared with the EV^GLN+ treatment, EV^GLN− treatment exhibited a higher capacity to induce splenocyte migration in a Transwell system ([126]Fig. 4F). These results suggest that EV^GLN− may enhance cytokine release and chemotaxis of innate immune cells and could be used as a promising therapeutic for diseases. Moreover, the effects of EV^GLN− on T cell activation were also evaluated in mouse splenocytes, which contain high numbers of T cells and other leukocytes ([127]27). Mouse splenocytes were stimulated with concanavalin A (ConA), cotreated with EV^GLN− or EV^GLN+ for 72 hours, and then detected using FCA (fig. S7A). The FCA data were visualized using a t-distributed stochastic neighbor embedding (t-SNE) scatterplot ([128]Fig. 4H, left panel) as previously reported ([129]28). EV^GLN− treatment enhanced the ability to induce CD8^+ T cell (CD3^+CD8^+CD69^+) or CD4^+ T cell (CD3^+CD4^+CD69^+) activation in ConA-primed splenocytes compared to EV^GLN+ treatment ([130]Fig. 4H), indicating that EV^GLN− might improve the potential to potentiate T cell activation in vitro. Fig. 4. Immunostimulatory effects of M1-EVs from GLN^− conditions. [131]Fig. 4. [132]Open in a new tab (A and B) M2-Mφs were induced by IL-4/TGF-β (20 ng/ml each), and qPCR analysis was conducted to measure the expression of the M2 gene (Arg1, Mrc1, and TGF-β) expression in M2-Mφs treated with EV^GLN− (20 μg/ml) or EV^GLN+ (20 μg/ml) for 48 hours (n = 3; ***P < 0.001, **P < 0.01, ^##P < 0.01, ^###P < 0.01 versus the M2 group). (C and D) qPCR analysis of chemokine gene expression (Ccl2 and Cxcl2) in THP-1 monocytes treated with EV^GLN− preparations or EV^GLN+ preparations (20 μg/ml) for 24 hours (n = 3; ***P < 0.001, **P < 0.01 versus the “CON” group). (E and F) Chemotaxis evaluation of (E) conditioned culture medium from EV pretreated THP-1 cells and (F) EVs from splenocytes using a Transwell system, and the migrated cells in the lower chamber were counted using FCA (n = 3; **P < 0.01, *P < 0.05 versus the CON group). (G and H) Mouse splenocytes were treated with ConA or ConA plus EV^GLN− or EV^GLN+ (20 μg/ml) for 72 hours, and the populations of activated CD4^+ T cells and (CD3^+CD4^+CD69^+) activated CD8^+ T cells (CD3^+CD8^+CD69^+) were determined by FCA (n = 3). (I and J) Evaluation of immune responses in mice (n = 5) intravenously injected with EV^GLN− or EV^GLN+ (30 μg/mouse) for 4 hours and immune cell populations (F4/80^+ Mφs, Ly6C^+ monocytes, and Ly6G^+ neutrophils) in the spleen were analyzed using FCA (**P < 0.01, *P < 0.05 versus the CON group). To confirm these in vitro findings, we administered M1-EVs to normal mice and observed the effects of these M1-EVs on immune activation in vivo ([133]Fig. 4I). The in vivo distribution of M1-EVs was first assayed in normal mice by intravenously injecting Cy7 (a near-infrared dye)–labeled EV^GLN−. In line with the findings of previous reports ([134]13, [135]29), the imaging results revealed that EV^GLN− accumulated mainly in major organs, such as the liver, spleen, lung, kidney, and heart, of the mice at 4 hours after injection (fig. S8). Subsequently, the changes in populations of neutrophils, monocytes, and Mφs in response to EV injection were analyzed using FCA (fig. S9, A and B), since these cells are key innate cells that participate in various functions, such as host defense, inflammation, and tissue injury processes. Previous reports have indicated that systematically administered (intravenous) EVs are primarily taken up by innate immune cells, such as monocytes and Mφs ([136]30). Therefore, our in vitro results suggest that intravenously injected EV^GLN− may enhance the activation of the innate immune system in vivo. The spleen is a major immune organ that contains large numbers of innate immune cells, and populations of Ly6G^+ neutrophils, Ly6C^+ monocytes, and F4/80^+ Mφs in the spleen tissues of mice were detected at the acute phase (4 hours) after EV administration. As shown in [137]Fig. 4J, mice in the EV^GLN− group exhibited higher populations of Ly6C^+ monocytes, F4/80^+ Mφs, and Ly6G^+ neutrophils than those in the PBS or EV^GLN+ group, which corresponded with the findings obtained from the in vitro experiments. These findings were consistent with the in vitro findings. The enhanced ability of EV^GLN− treatment to boost innate immunity may provide additional beneficial outcomes in certain diseases, such as cancer and infectious diseases. For example, polarizing Mφs to a proinflammatory phenotype can reverse the immunosuppressive tumor environment and thus enhance the antitumor responses of Mφs in vivo ([138]31). Overall, our results suggest that, compared with conventional EV^GLN+, EV^GLN− has immunostimulatory effects in vivo, suggesting that it is a promising candidate for boosting immunostimulatory therapy. GLN^− conditions up-regulated EV biogenesis and cytokine synthesis pathways Although GLN^− conditions can increase EV secretion and immunostimulatory potency in M1-Mφs, the underlying mechanisms are unclear and need to be explored. Thus, RNA sequencing (RNA-seq) was conducted to profile the global changes in the gene expression of Mφs (table S2). We observed a clear separation and distinct gene expression patterns between the GLN^+ and GLN^− groups, as shown in the PCA plot and heatmap ([139]Fig. 5, A and B). The differentially expressed genes (DEGs; 5206 up-regulated and 3439 down-regulated) that were induced by GLN^− conditions were identified ([140]Fig. 5C). GO enrichment analysis revealed that the DEGs were involved in multiple pathways related to vesicle formation, such as the membrane, plasma membrane, cytoplasmic vesicle, and external side of the plasma membrane ([141]Fig. 5D). Furthermore, we found that the expression of numerous genes involved in the ESCRT subcomplexes, such as ESCRT-0 (e.g., STAM, AP4S1, and PICALM), ESCRT-I (e.g., TSG101, VPS37C, and VPS37B), ESCRT-II (e.g., VPS25 and SNF8), and ESCRT-III (e.g., CHMP4B, CHMP1A, and VPS4B), was up-regulated in M1-Mφs under GLN^− conditions compared to that in the GLN^+ group ([142]Fig. 5E). The gene profile of M0-Mφ under GLN^− conditions exhibited a similar pattern, with DEGs enriched in processes related to vesicle formation, including cytoplasmic vesicles, endosomes, and membranes (fig. S10, A and D, and table S3). Additionally, the expression levels of ESCRT genes, such as ESCRT-0 (e.g., STAM, AP4S1, and CLBA1), ESCRT-I (e.g., TSG101, VPS37C, and MVB12B), and ESCRT-III (e.g., CHMP4B, CHMP1A, and VPS4B), were also higher in M0-Mφs of the GLN^− group than in those of the GLN^+ group (fig. S10E). Notably, the ESCRT pathway plays crucial roles in the formation of ILVs and multimembrane endosomes (MVEs) by cooperating with the adenosine triphosphatase (ATPase) VPS4 to drive membrane scission or sealing ([143]18, [144]32), while depletion of ESCRT components was shown to reduce exosome biogenesis in Mono-Mac6 cells ([145]33). ESCRT-related proteins, such as TSG101 and ALIX, have been widely used as positive markers for EV characterization. Thus, our results indicate that the increased secretion of EVs under GLN^− conditions is at least partially attributed to the up-regulation of ESCRT pathway activity. Fig. 5. Effect of GLN^− conditions on EV biogenesis in M1-like Mφs. [146]Fig. 5. [147]Open in a new tab (A) PCA scatterplot based on RNA-seq data of M1-Mφs treated with GLN^+ (2 mM) or without GLN (GLN^−) for 24 hours. (B) Heatmap showing the different gene expression profiles in the different groups. (C) Volcano plot showing DEGs (P-adjusted < 0.05) between groups. (D) GO enrichment analysis of DEGs (FC > 1.5, top 10) in response to GLN^− conditions. (E) Schematic illustrating the ESCRT machinery and heatmap showing ESCRT gene expression in the different groups. (F to H) Representative images and quantification of TSG101^+ early endosomes, CD63^+ MVBs, and Lamp2b^+ late endosomes in M1-Mφs under GLN^− conditions (scale bar, 50 μm). (I) qPCR analysis of TSG101 and VPS4B gene expression in M1-Mφ under GLN^− conditions cotreated with GW4869 for 24 hours. (J) EV yield determined by the EV/cell protein ratio after GW4869 treatment. (K) qPCR analysis of cytokine gene expression (e.g., IL-6 and IL-1β) in EV^GLN− after GW4869 treatment (n = 3; **P < 0.01, *P < 0.05 versus the GLN^− group). To assess the changes in EV biogenesis processes under GLN^− conditions, we assessed endosome components during EV biogenesis in Mφs using immunofluorescence (IF) staining as previously reported ([148]32). The results showed that, compared with those from the GLN^+ condition, the GLN^− condition increased the number of TSG101^+ early endosomes, the number of CD63^+ multivesicular bodies (MVBs), and the number of Lamp2b^+ late endosomes formed in Mφs ([149]Fig. 5, F to H), indicating an overall enhancement of EV biogenesis under GLN^− conditions. Additionally, M1-Mφs under GLN^− conditions were treated with GW4869, a neutral sphingomyelinase inhibitor known to block exosome generation by inhibiting ceramide-mediated pathways ([150]34). GW4869 treatment reduced the expression of several ESCRT-related genes (e.g., TSG101 and VPS4B) in M1-Mφs and increased the expression of cytokines (e.g., IL-6 and IL-1β) in M1-EVs. In addition, the GLN^− group had a slightly decreased EV yield compared with that of the GLN^− group ([151]Fig. 5, I to K). It has been proposed that the generation of different EV subtypes may involve diverse intracellular compartments and distinct EV biogenesis routes, and the ESCRT machinery is involved in both exosome and ecotosome biogenesis ([152]35). Moreover, studies have reported that GW4869 can reduce exosome secretion but increase ecotosome secretion ([153]35). Therefore, these results suggest that the enhanced EV biogenesis and cargo (e.g., cytokines) sorting in Mφs under GLN^− conditions partially rely on the ESCRT pathway. However, its impact on different EV subtypes needs further investigation in future studies. Having shown that GLN^− conditions increase the yield of EVs and facilitate the production of diverse immunostimulatory molecules, as well as their sorting into EVs, KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway enrichment analysis was also conducted to elucidate the underlying mechanisms involved. The results showed that many DEGs of M1-Mφ under GLN^− conditions were also enriched in pathways related to cytokine production and immune responses, such as the Janus kinase (JAK)–signal transducer and activator of transcription (STAT) pathway (e.g., CREB3L4, IL6, IL11, and LTA), the NF-κB pathway (e.g., IL1R1, TNFRSF9, JAK2, PTEGS2, IL27, and AHR), TNF signaling pathway (e.g., GBP3, IL1β, TNFSF1B, and IL33), cytokine–cytokine receptor interaction (e.g., GDF9, MMP13, IL13RA2, GBP7, and EDN1), and T[H]17 cell differentiation (e.g., IL-1α, LCN2, and FOS) ([154]Fig. 6, A to C). KEGG analysis also revealed that the DEGs in M0-Mφs under GLN^− conditions were enriched in pathways related to immune responses, such as cytokine–cytokine receptor interaction, the TNF signaling pathway, the JAK-STAT pathway, and the NF-κB pathway (fig. S10F). Several of these pathways (e.g., STAT and NF-κB) have been shown to play crucial roles in mediating various immune processes, such as cytokine and chemokine synthesis, inflammasome activation, and immune cell recruitment (e.g., monocytes, Mφs, and T cells) ([155]36, [156]37). For example, in endothelial cells treated with monocyte-derived EV treatment, the NF-κB, Toll-like receptor 4 (TLR4), and myeloid differentiation primary response gene 88 (MyD88) pathways were activated, leading to up-regulated expression of adhesion molecules (e.g., ICAM1 and VCAM1) and cytokines (e.g., IL1β and IL6) ([157]38). Further, quantitative real-time polymerase chain reaction (qPCR) assays confirmed the expression of genes associated with the STAT and NF-κB pathways (e.g., Stat4, Stat6, IL-12B, and Nlrp3) in the GLN^− group compared to that in the GLN^+ group ([158]Fig. 6D). Moreover, M1-Mφ in the GLN^− group showed higher protein expression levels of p-NF-κB and NLRP3 than those in the GLN^+ group ([159]Fig. 6E). Similarly, compared with those in the GLN^+ group, M1-Mφ in the GLN^− group released higher levels of immunostimulatory cytokines (e.g., TNF-α and IL-6) and chemokines (e.g., CCL2) and lower levels of immunosuppressive cytokines (e.g., IL-10) ([160]Fig. 6F). To verify the vital role of the identified cytokine synthesis pathway (e.g., NF-κB) in this process, M1-Mφs under GLN^− conditions were cotreated with dehydroxymethylepoxyquinomicin (DHMEQ; an NF-κB pathway inhibitor) ([161]39), and the changes in cytokine expression in parent cells and EVs were assayed. As shown in [162]Fig. 6 (G and H), compared with those in the GLN^− group, the levels of immunostimulatory cytokines (e.g., IL-6 and IL-1β) in M1-Mφ and cytokine mRNA (e.g., IL-6 and IL-1β) in M1-EVs in the GLN^− group were reduced by NF-κB inhibitor treatment, suggesting that the GLN^− state can induce cytokine synthesis and sorting process. Fig. 6. Effect of GLN^− conditions on immunoregulatory cargo synthesis in Mφs. [163]Fig. 6. [164]Open in a new tab (A) KEGG enrichment analysis of DEGs (FC > 1.5) in M1-Mφs under GLN^− conditions for 24 hours. (B) Heatmap showing the differential expression levels of immune-related genes in the different groups. (C) Interaction network analysis of DEGs in response to GLN^− conditions. (D) qPCR analysis of Stat4, Stat6, IL-12B, and Nlrp3 gene expression in Mφs from the different groups. (E) Western blot analysis and quantification of inflammatory pathway proteins (NF-κB and NLRP3) in Mφs from the different groups (n = 3). (F) Luminex assay measuring cytokine (TNF-α, IL-6, and IL-10) and chemokine (CCL2) levels in culture medium from M1-Mφs cultured under GLN^+ or GLN^− conditions for 24 hours. (G) qPCR analysis of cytokine gene expression (IL-1β and IL-6) in M1-Mφs under GLN^− conditions cotreated with DHMEQ for 24 hours. (H) qPCR analysis of cytokine gene expression (e.g., IL-6 and IL-1β) in EV^GLN− after DHMEQ treatment. (I and J) Representative images and quantification of the colocalization of cytokines (IL-1β) with HSP70^+ or TSG101^+ endosomes in M1-Mφs under GLN^− conditions (scale bar, 50 μm) (n = 3; ***P < 0.001, *P < 0.05 versus the GLN^+ group). Recent reports have highlighted the involvement of both active and passive mechanisms in the EV cargo sorting process. The active sorting process relies on the EV biogenesis machinery (e.g., ESCRT), which captures and clusters cargos, while passive sorting occurs based on the cellular levels of cargos and the rate of EV production ([165]18). To determine the role of sorting mechanisms under GLN^− conditions, we detected the colocalization of EV biogenesis markers and cytokines in M1-Mφs. As shown in [166]Fig. 6 (I and J), we observed increased colocalization of cytokines (IL-1β, green) and EV biogenesis and trafficking markers (TSG101 and HSP70, red) in cells from the GLN^− group compared to those from the GLN^+ group. This finding suggested that the enrichment of immunostimulatory cytokines in EV^GLN− may be partly due to the enhanced activity of the sorting machinery and increased cytokine production in these Mφs. In addition to cargo sorting, the ESCRT machinery can also affect the rate of EV biogenesis, and thus, the passive sorting process might also contribute to the enrichment of cytokines in EV^GLN− ([167]32). Together, these results indicate that GLN^− conditions promote EV secretion and the synthesis and sorting of immunostimulatory cargos into EVs in M1-Mφs, and these effects are mediated by the up-regulation of pathways involved in EV biogenesis (e.g., ESCRT) and cytokine production (e.g., STAT and NF-κB). GLN^− conditions improved EV yield and function through metabolic rewiring To further explore the detailed metabolic alterations contributing to EV secretion in the GLN^− state, the changes in the metabolic profiles of M1-Mφ were analyzed using LC-MS/MS–based targeted metabolomics (table S4). PCA and heatmap analysis revealed distinct metabolite profiles between the GLN^+ group and the GLN^− group ([168]Fig. 7, A and B), suggesting that GLN^− induces systemic metabolic rearrangement in Mφs. Different metabolites (12 up-regulated and 19 down-regulated) between the GLN^+ group and the GLN^− group were identified ([169]Fig. 7C). Further KEGG enrichment analysis revealed that the differentially expressed metabolites were mainly involved in pathways related to cellular energy and redox metabolism, such as the TCA cycle, GSH [glutathione (reduced form)] metabolism, glycolysis, and glycine metabolism ([170]Fig. 7D). The GLN^− group exhibited lower levels of metabolites related to glutaminolysis (e.g., αKG, glutamate, and GSH) (fig. S11A), as well as the TCA cycle (e.g., fumarate, succinate, and citrate) (fig. S11B), and higher levels of glycolysis and pentose phosphate pathway (PPP) (e.g., G-6-P and F-6-P) (fig. S11C) and nucleotide metabolism [e.g., uridine diphosphate (UDP) and guanosine monophosphate (GMP)] (fig. S11D) than the GLN^+ group ([171]Fig. 7E and fig. S5E). GLN is an essential precursor of intracellular antioxidants, such as glutathione (GSH) and NADPH [reduced form of nicotinamide adenine dinucleotide phosphate (NADP^+)], while GLN^− can lead to impaired antioxidant capacity and elevated levels of reactive oxygen species (ROS) ([172]9). GLN can be converted to αKG through glutaminolysis and enter the TCA cycle and glutathione (GSH) synthesis to mitigate ROS ([173]20). Similarly, we observed that M1-Mφs in the GLN^− group exhibited a lower glutathione/oxidized glutathione (GSH/GSSG) ratio and nicotinamide adenine dinucleotide/reduced NAD^+ (NAD^+/NADH) ratio and a greater reduction in the nicotinamide adenine dinucleotide phosphate/NADP^+ (NADPH/NADP^+) ratio than those in the GLN^− group ([174]Fig. 7F). NAD^+, NADPH, and GSH are essential cofactors for maintaining cellular energy metabolism and redox homeostasis. For example, NAD^+ can be reduced to NADH to regulate mitochondrial energy metabolism and redox homeostasis ([175]40). GSH can be oxidized to GSSG and act as a key scavenger of intracellular ROS. NADPH, an electron donor, is indispensable for converting GSSG to GSH ([176]40). Unlike GSH, NADPH is mainly generated from GLC via the PPP ([177]41), suggesting that the increase in glycolysis and the increase in the PPP in M1-Mφ under GLN^− conditions are likely adaptive effects that aim to maintain cellular energy and redox balance. These results collectively suggest that GLN^− conditions may induce systemic metabolic rewiring in M1-Mφs, leading to oxidative metabolism defects, redox imbalance, and enhanced glycolysis/PPP pathway activity (fig. S12). Fig. 7. Systemic metabolic rewiring of Mφs under GLN^− conditions. [178]Fig. 7. [179]Open in a new tab (A) Cells were cultured in medium supplemented with GLN (2 mM, GLN^+) or without GLN (GLN^−) for 24 hours, and a PCA scatterplot was generated based on the metabolomic data of the different groups (n = 5). (B) Heatmap showing different metabolites in different groups (n = 5). (C) Volcano plots illustrating differentially abundant metabolites (P-adjusted < 0.05) between groups (n = 5). (D) Metabolic pathway enrichment of different metabolites under GLN conditions. (E) Heatmap showing detailed differentially abundant metabolites between groups (n = 5). (F) Measurements of cellular GSH/GSSG ratio (left), NADPH/NADP^+ ratio (middle), and NAD^+/NADH (right) ratio in the different groups (n = 5). (G) Pie charts depicting the number (top) and sublocalization (bottom) of mitochondrial DEGs (mito-DEGs) revealed by RNA-seq analysis between the GLN^+ and GLN^− groups (n = 3). (H) KEGG enrichment analysis of mito-DEGs and the top 10 affected pathways (n = 3). (I) Heatmap showing mito-DEGs (FC > 2 and P-adjusted < 0.05) between groups (n = 3). (J) Measurements of mitochondrial ROS, mitochondrial membrane potential, and ATP levels in M1-Mφs under GLN^+ conditions or GLN^− conditions for 24 hours (**P < 0.01, *P < 0.05, ^NSP > 0.05 versus the GLN^+ group). Mitochondria play a central role in cellular energy production [adenosine triphosphate (ATP)] and redox metabolism and are also the primary source of intracellular ROS (~90%, mtROS) ([180]42). To better understand the impact of GLN depletion on mitochondrial function, we analyzed the changes in mitochondrial genes using MitoCarta2.0 ([181]43). The results showed that 6.28% (543 of 8645) of the DEGs whose expression was induced by GLN^− conditions were mitochondrial genes, 48.25% of which were related to the mitochondrial matrix and 30.57% to the mitochondrial inner membrane ([182]Fig. 7G). In line with the metabolomic results, GLN^− conditions affected multiple metabolic processes in M1-Mφ, such as metabolic pathways (e.g., SDHD, ACAT1, IDH2, COX7B, and GLUD1), ROS (e.g., ATP5, SDHC, COX7B, and SOD1), oxidative phosphorylation (e.g., ATP5G1, SDHD, SDHC, COX7B, and COX10), and the TCA cycle (e.g., PDHA1, CS, IDH3A, PCK2, and FH1) ([183]Fig. 7, H and I). Perturbations in energy and redox metabolism have been associated with cytokine production and EV secretion in both immune and nonimmune cells ([184]7), and oxidative stress can affect the production of EVs by redirecting their destinations (e.g., the lysosomal pathway). For instance, mitochondrial respiratory defects and mtROS overproduction under hyperglycemic conditions induce NF-κB/p38 MAPK pathway and cytokine (e.g., TNF-α and IL-6) production in Mφs ([185]41). Likewise, mitochondrial stress caused by mitochondrial Lon overexpression promoted the secretion of EVs harboring mtDNA in cancer cells ([186]44). Consistently, M1-Mφs in the GLN^− group exhibited impaired mitochondrial oxidative metabolism, as indicated by increased mtROS and decreased mitochondrial membrane potential and ATP compared to those in the GLN^+ group ([187]Fig. 7J). These results suggest that the up-regulated pathways involved in EV biogenesis and cytokine production in M1-Mφ under GLN^− conditions may be linked to the rewiring of mitochondrial energy and redox metabolism. Next, we assessed whether restoring GLN metabolism through anaplerosis could partially reverse the increase in EV secretion and cytokine production in M1-Mφs ([188]Fig. 8, A and E). αKG is a metabolite of glutaminolysis and a vital intermediate of the TCA cycle, and N-acetylcysteine (NAC) is a vital precursor of GSH that can mitigate excessive intracellular ROS ([189]20, [190]45). Thus, M1-Mφs under GLN^− conditions were supplemented with αKG or NAC. The expression levels of immunostimulatory molecules (e.g., IL-1β, IL-6, and IL-12) were lower in the αKG supplementation group, compared to those in the GLN^− alone group, but the EV yield was not affected in M1-Mφs under GLN^− conditions ([191]Fig. 8, B to D). A possible explanation is that GLN regulates cellular functions via diverse metabolic pathways, and αKG may mainly function in certain aspects (e.g., energy metabolism), which could partially affect EV production (e.g., composition) in Mφs. On the other hand, NAC supplementation reduced the expression levels of immunostimulatory cytokines (e.g., IL-1β, IL-6, and IL-12B) and the yield of EVs in M1-Mφs under GLN^− conditions ([192]Fig. 8, E to G). Neither αKG nor NAC supplementation altered the EV size distribution of M1-Mφs ([193]Fig. 8, C and F). However, neither αKG nor NAC supplementation changed the EV production of M1-Mφs under GLN^+ conditions, and cellular cytokine (IL-1β, IL-6) synthesis was not affected by NAC treatment (fig. S13). Notably, αKG treatment reduced cytokine synthesis in M1-Mφs under GLN^+ conditions (fig. S13), supporting the findings of previous reports, which indicated that excessive αKG can promote M2-Mφ activation ([194]10). These results confirmed that GLN^− conditions induced cellular metabolism rewiring and play vital roles in both promoting both EV secretion and stimulating the sorting of immunostimulatory cytokines into M1-EVs. GLN^− conditions induce mitochondrial stress (defects in the TCA cycle and GSH metabolism) accompanied by increased utilization of GLC and the PPP pathway in M1-Mφs (fig. S14), which subsequently up-regulates pathways related to cytokine production (e.g., NF-κB and STAT), EV biogenesis, and cargo sorting (e.g., ESCRT), ultimately resulting in an increase in the release of EVs containing abundant immunoregulatory contents from such cells. Fig. 8. Effect of GLN metabolism anaplerosis on EV secretion and cytokine synthesis in Mφs. [195]Fig. 8. [196]Open in a new tab (A and B) Schematic illustrating the αKG supplementation experiment. M1-Mφs were cultured in GLN medium supplemented with or without αKG (0.8 mM) for 24 hours, after which the gene expression levels of immunostimulatory molecules (IL-1β, IL-6, and IL-12B) were analyzed via qPCR analysis (n = 3). (C) EV sizes in the different groups (n = 3). (D) EV yields determined by the EV number/cell protein ratio (left) or the EV protein/cell protein ratio (right) in the different groups (n = 3). (E and F) Schematic illustrating the NAC treatment experiment. M1-Mφs were cultured in GLN medium supplemented with or without NAC (0.5 μM) for 24 hours, and the gene expression of immunostimulatory molecules (IL-1β, IL-6, and IL-12B) was analyzed using qPCR analysis (n = 3). (G) EV sizes in the different groups (n = 5). (H) EV yields determined by EV number/cell protein ratio (left) or EV protein/cell protein ratio (right) in the different groups (n = 5) (**P < 0.01, *P < 0.05, ^NSP > 0.05 versus the “GLN^−LPS^+” group). Although our results have elucidated the role of nutrient availability in regulating EV secretion, composition, and function, several questions remain elusive and need to be addressed in future studies. For example, we only investigated whether GLN^− conditions increased functional EV secretion in M1-Mφs, but whether modulating GLN metabolism could promote EV secretion in M2-Mφs has not been determined. It is unclear whether the increased EV secretion in M2-Mφs is dependent on metabolic alterations and signaling pathways similar to those observed in M1-Mφs. Mφs with different phenotypes exhibit diverse metabolic signatures and nutrient demands ([197]3); thus, it is necessary to evaluate the impacts of specific nutrient changes on EV secretion and function during M2-EV production. The metabolic states of immune cells are diverse, and various immune cells may respond differently to the same nutrient conditions ([198]19). Thus, the optimal conditions for EV production in Mφs may not be applicable to other immune cell types, necessitating careful assessment of the nutrient requirements for each specific immune cell. Additionally, while certain methods of engineered EV production (e.g., preconditioning and genetic modification) have been shown to increase EV secretion or cargo levels ([199]46, [200]47), the potential enhancement of these effects through optimizing nutrient conditions has not been explored. Thus, further investigations are needed to develop advanced EV-based immunotherapeutics by integrating conventional approaches and metabolic (nutrient) modulation. Nevertheless, our study highlights the significance of nutrient availability in regulating functional EV secretion. Therefore, fine-tuning nutrient conditions should be carefully considered in future studies concerning therapeutic EV production for diverse disease treatments. In summary, the nutrient status of immune cells (e.g., Mφs) can regulate both the secretion and function of EVs from immune cells. Precisely modulating the metabolic states of donor cells is an effective strategy for enhancing the production of EVs with valuable bioactivities ([201]Fig. 9). Changes in the availability of an individual nutrient component (e.g., GLN depletion) in the medium are sufficient to induce systemic metabolic reprogramming and activate pathways related to EV biogenesis and cargo synthesis in donor cells. As a result, the yield, functional composition, and immunoregulatory potency of the produced EVs improved. These findings emphasize the importance of reconsidering the optimal metabolic state when generating therapeutic EVs. This topic has been overlooked in many previous studies. Furthermore, as indispensable substrates for cell growth, nutrients can be readily integrated with many other techniques to synergistically enhance the production of functional EVs. This study highlights the potential of fine-tuning nutrient conditions as a promising strategy for tailored EV engineering and provides valuable insights into the development of advanced EV-based therapeutics for various diseases. Fig. 9. Nutrient availability can regulate EV secretion and function in immune cells. [202]Fig. 9. [203]Open in a new tab Fine-tuning nutrient availability–induced metabolic rewiring is a basic and promising strategy for bioengineering EVs as advanced therapeutics for the treatment of diverse diseases. (A) GLN^− depletion efficiently promotes EV biogenesis and cargo packaging via systemic metabolic rewiring of Mφs. (B) EVs derived from Mφs under GLN^− conditions (EV^GLN−) exhibit favorable immunostimulatory potential. MATERIALS AND METHODS Cell culture Mouse Mφ RAW 264.7 cells and the human monocytic THP-1 cells were cultured in control RPMI 1640 (31800105, Gibco, La Quinta, CA, USA) supplemented with 10% heat-inactivated FBS (Gibco), penicillin (50 U/ml), and streptomycin (50 U/ml). M0-Mφs were stimulated with LPS (40 ng/ml, Sigma-Aldrich, St. Louis, MO, USA) for M1 activation and with mouse recombinant IL-4 and TGF-β (20 ng/ml each, Novoprotein, Shanghai, China) for M2 activation. To block glutaminolysis, M1-Mφs were treated with BPTES (10 nM, CSN Pharm, Coquitlam, BC, Canada) for 24 hours. For GLN metabolism anaplerosis, M1-Mφs cultured in GLN^+ or GLN^− medium were supplemented with dimethyl αKG (0.8 mM, Sigma-Aldrich) or NAC (0.5 μM, Beyotime, Beijing, China) for 24 hours. Isolation of cell-secreted EVs EV-depleted FBS was prepared using an ultracentrifugation (120,000g, 16 hours, 4°C) method as previously reported ([204]48). M0-Mφs or M1-Mφs were cultured in RPMI 1640 supplemented with EV-depleted FBS and subjected to the indicated nutrient conditions for 24 hours. Then, the culture medium was collected and subjected to EV isolation via a differential centrifugation method ([205]16). In brief, the medium was centrifuged at 300g for 10 min and at 2000g for 30 min at 4°C to remove cells and other debris, followed by ultracentrifugation at 110,000g for 90 min at 4°C in an SW32Ti rotor (Beckman Coulter, Brea, CA, USA) to obtain EVs. After washing with PBS, the EV pellets were recentrifuged at 110,000g for 90 min at 4°C. The purified EVs were resuspended in PBS and stored at −80°C until further use. Transmission electron microscopy The morphology of the EVs was evaluated using TEM (HT7800, Hitachi Ltd., Tokyo, Japan) as previously described ([206]3). EV samples were diluted with PBS and dropped on a carbon-coated copper grid. To remove unattached EVs, the grid was washed with pure water. After staining with 2% phosphotungstic acid, the grids were air-dried and subjected to TEM. Nanoparticle tracking analysis Nanoparticle tracking analysis (NTA) of the EV samples was carried out using a Zeta View PMX 120 (Particle Metrix, Meerbusch, Germany) as previously described ([207]3). Briefly, EVs were diluted (v/v, 1:2000) with pure water, and their size distributions and particle concentrations were analyzed using the following parameters (minimum area 5, maximum area 1000, and minimum brightness 20) at 25°C. EV quantification The protein concentrations of the EV samples were quantified using Pierce BCA Protein Assay Kits (Thermo Fisher Scientific, Carlsbad, CA, USA). The EV yield of each group was assessed using the ratio of EV protein or particles relative to total protein in the donor cell as previously reported ([208]49). Western blotting Protein extraction from the samples was carried out using radioimmunoprecipitation assay (RIPA) buffer containing protease and phosphatase inhibitors (Calbiochem, San Diego, CA, USA). The protein concentration of samples was quantified using a BCA kit. Protein electrophoresis was performed using 10% SDS–polyacrylamide gel electrophoresis (SDS-PAGE), followed by protein transfer from the gel to a nitrocellulose membrane (NC; Cytiva, Germany). The membranes were blocked in 5% nonfat milk and incubated with mouse anti-ALIX (12422-1-AP, Proteintech, Wuhan, China), mouse anti-HSP70 (A0284, ABclonal, Wuhan, China), mouse anti-TSG101 (14497-1-AP, Proteintech), mouse anti-GM130 (A11408, ABClonal), mouse anti–pNF-κB (WL02169, Wanleibio, China), mouse anti–NF-κB (10745-1-AP, Proteintech), mouse anti-NLRP3 (15101, Cell Signaling Technology), and mouse anti–β-actin (AC026, ABclonal) at 4°C overnight. The membranes were washed and incubated with a secondary antibody conjugated with horseradish peroxidase (HRP) at 37°C for 1 hour. Protein signals were detected with a chemiluminescence kit (MedChemExpress, Shanghai, China). Protein quantification was carried out using ImageJ software. Modification of cellular nutrient conditions Mφs were cultured in control RPMI 1640 supplemented with 10% FBS, 11 mM GLC, and 2 mM GLN or in modified medium supplemented with altered nutrients, including FBS, GLC, AAs, and FAs. In brief, a stock solution of each nutrient was prepared and subsequently added to cells, after which the final concentration of each nutrient was considered lower (2.5%, v/v) FBS or higher (20%, v/v) FBS, higher concentrations of GLC (20 mM, Sigma-Aldrich), higher concentrations of GLN (8 mM, GlutaMAX, Gibco), BCAAs (5 mM), or OA (100 μM, Aladdin, Shanghai, China) dissolved in FA-free bovine serum albumin (BSA; Solarbio, Beijing, China). The BCAA stock solution was prepared as a mixture of leucine, isoleucine, and valine at 100 mM (Sigma-Aldrich), and BSA alone was used as a control in the OA supplementation experiments. In concentration course tests of GLC or GLN, cells were cultured in GLC-free RPMI (11879020, Gibco) supplemented with GLC (0, 2, and 10 mM; A2494001, Gibco) or GLN-free RPMI supplemented with GLN (0, 2, and 10 mM) for 24 hours. In the case of GLN-depleted conditions, cells were cultured in GLN-free RPMI 1640 (21870076, Gibco) supplemented with (GLN^+, 2 mM) or without GLN for 24 hours (GLN^−). After incubation for the indicated times, the cells or conditioned medium from each group was collected for further tests. LC-MS/MS–based proteomic analysis of EVs Total EV proteins were extracted from the EV samples and quantified using a BCA kit (Thermo Fisher Scientific). Protein samples (100 μg) were taken for reductive alkylation and digested with trypsin overnight at 37°C. The peptides were subjected to desalting and vacuum-dried, and the concentrations were determined by a peptide quantification kit (Thermo Fisher Scientific). Then, 30 μg of trypsin-digested peptides was analyzed by an EASY nLC-1200 system (Thermo Fisher Scientific, USA) coupled with a timsTOF Pro2 (Bruker, Germany) mass spectrometer at Majorbio Bio-Pharm Technology Co. Ltd. (Shanghai, China). Briefly, the tryptic peptides were separated by an ultrahigh-performance liquid phase system subjected to a capillary ion source and then analyzed by timsTOF Pro2 at a flow rate of 250 nl/min (the electrospray voltage was 1.5 kV). The peptide parent ions and their secondary fragments were detected and analyzed using high-resolution time of flight. MS/MS spectra were searched using MaxQuant version 2.0.3.1 software. Bioinformatic analysis of the proteomic data was performed with a free online platform ([209]https://www.omicsolution.org/wkomics/main/). DEPs with fold change (FC) > 1.5 and P-adjusted < 0.05 were identified using a free online platform ([210]https://www.omicsolution.org/wkomics/main/) with SAMR analysis ([211]50). PCA plots, volcano plots, and heatmaps were generated using a free online platform ([212]https://www.omicsolution.org/wkomics/main/). GO analysis of the DEPs was performed using STRING ([213]https://cn.string-db.org/) and the Metascape tool (v3.5) ([214]51), and false discovery rate (FDR) < 0.05 was considered to indicate statistical significance. Quantitative real-time polymerase chain reaction Total RNA was extracted from cells using TRIzol reagent (Gibco), and cDNA was synthesized with a commercial kit (Vazyme, Nanjing, China). qPCR was performed with SYBR Green (Vazyme) on a CFX96 real-time PCR detection system (Bio-Rad, Hercules, CA, USA). The primers used in this study are listed in table S5. The data were analyzed by Bio-Rad CFX Manager software, and the relative FC in mRNA levels was determined using the ∆∆C[t] method with GAPDH or β-actin as the internal reference gene. Impact of EV preparations on M2-Mφs M2-Mφs were induced by stimulating RAW 264.7 cells with mouse recombinant IL-4 and TGF-β (20 ng/ml each, Novoprotein, Shanghai, China) for 48 hours. M2-Mφs were treated with PBS, EV^GLN− preparations (20 μg/ml), or EV^GLN+ preparations (20 μg/ml) for 48 hours, after which the expression of M2 genes (Arg1, Mrc1, and TGF-β) in the cells was detected via a qPCR assay. Impact of EV preparations on chemotaxis THP-1 monocytes were treated with PBS, EV^GLN− preparations (20 μg/ml), or EV^GLN+ preparations (20 μg/ml) for 24 hours, after which the gene expression of chemokines (Cxcl2 and Ccl2) in the cells was assayed via qPCR. The supernatants of each group were collected and concentrated using ultrafiltration tubes (3-kDa molecular weight cutoff, Sigma-Aldrich). Mouse splenocytes (mainly consisting of monocytes and lymphocytes) were isolated from 8-week-old male C57BL/6 mouse spleens using density gradient medium (Ficoll-Paque, Cytiva, Marlborough, USA) according to the manufacturer’s protocols. In a Transwell system (5-μm pore size, Labselect, Hefei, China), mouse mononuclear cells and concentrated medium (with equal protein amounts) were added to the upper and lower chambers, respectively. After 4 hours of incubation, the migrated cells in the lower chamber were collected and counted using flow cytometry (Beckman Coulter). To evaluate the direct chemotaxis effect of EVs, mouse splenocytes were cultured with PBS, EV^GLN+ preparations (10 μg/ml), or EV^GLN− preparations (10 μg/ml) in the upper chambers for 4 hours, and the migrated cells in the lower chamber were collected and counted using a flow cytometer (Cytoflex, Beckman Coulter). Impact of EV preparations on T cells Mouse mononuclear cells were treated with ConA (2 μM, Thermo Fisher Scientific), ConA plus EV^GLN− preparations (10 μg/ml), or EV^GLN+ preparations (10 μg/ml) for 72 hours (n = 3). After treatment, cells from each group were collected and stained with fluorescein isothiocyanate (FITC)–conjugated anti-mouse CD3e (553061, BD, Brea, CA, USA, USA), peridinin chlorophyll protein (PerCP)/Cyanine5.5-conjugated anti-mouse CD4 (100434, BioLegend), phycoerythrin (PE)–Cy7–conjugated anti-mouse CD8a (552887, BD), and allophycocyanin (APC)–conjugated anti-mouse CD69 (APC-65105, Proteintech, Wuhan, China) for 30 min. After washing with PBS, the stained cells were analyzed using a flow cytometer (LSRFortessa, BD Biosciences). Immune responses to EV preparations in vivo All animal experiments were approved by the Animal Care and Use Committee of West China Hospital, Sichuan University (permit no. 20230330008) and were performed according to the guidelines of the National Institutes of Health (NIH). BALB/c mice (male, 8 weeks) were purchased from Byrness Weil Biotech Ltd. (Chengdu, China) and housed in a pathogen-free facility. Normal mice were randomly divided into three groups (n = 5) and intravenously injected with 100 μl of PBS, EV^GLN− preparations (30 μg each mouse), or EV^GLN+ preparations (30 μg each mouse). At 4 hours after injection, the mice were anaesthetized, and the spleens were collected and prepared as single-cell suspensions to detect immune cells. In brief, mouse spleen tissues were ground in PBS and then passed through a 70-μm filter. The red blood cells in spleen samples were removed using a Mouse Erythrocyte Lysing Kit (Solarbio, Beijing, China). The single-cell suspension was stained with FVS (564997, BD), APC-Cy7 rat anti-mouse CD45 (557659, BD Pharmingen), PE rat anti-mouse F4/80 (565410, BD), APC-conjugated anti-mouse Ly6G (560599, BD), and BV421-conjugated anti-mouse Ly6C (562727, BD) for 30 min. After washing with PBS, the stained cells were analyzed using a flow cytometer (LSRFortessa). RNA-seq analysis of cells M1 or M0 Mφs cultured in GLN^+ medium or GLN^− medium for 24 hours were collected for RNA-seq analysis. In brief, total RNA was extracted from tissue samples using TRIzol, and genomic DNA was removed using deoxyribonuclease (DNase) I (Takara, Shiga, Japan). The RNA-seq transcriptome library was constructed using a TruSeq RNA Sample Preparation Kit (Illumina, San Diego, CA, USA), and the paired-end RNA-seq library was sequenced on an Illumina HiSeq Xten/NovaSeq 6000 sequencer by Shanghai Majorbio Biopharm Technology Co. Ltd. (Shanghai, China). The expression levels of the transcripts were calculated using the transcripts per million reads (TPM) method. DEGs (FC > 1.5 and P-adjusted < 0.05) were considered significant. Venn plots, PCA plots, volcano plots, and heatmaps were generated using a free online platform ([215]https://www.omicsolution.org/wkomics/main/). GO and KEGG analyses of the DEGs were performed with DAVID Bioinformatics Resources ([216]https://david.ncifcrf.gov/home.jsp) and visualized using a free online platform ([217]https://www.omicsolution.org/wkomics/main/). IF staining After different treatments, the cells were fixed (4% paraformaldehyde for 10 min), permeabilized (0.3% Triton X-100 for 10 min), and blocked (1% BSA for 1 hour). Then, the fixed cells were incubated overnight at 4°C with mouse anti-TSG101 (14497-1-AP, Proteintech), mouse anti-CD63 (25682-1-AP, Proteintech), mouse anti-Lamp2b (27823-1-AP, Proteintech), mouse HSP70 (A0284, ABclonal), and mouse anti–IL-1β (A11369, ABclonal), followed by incubation with donkey anti-rabbit immunoglobulin G (IgG) H&L (DyLight 550) (ab96892, Abcam), tetramethyl rhodamine isothiocyanate (TRITC)–conjugated secondary antibody (2294985, Thermo Fisher Scientific), or FITC-conjugated secondary antibody (abs20139, Absin, Shanghai, China) as needed at 37°C for 1 hour. DAPI (4′,6-diamidino-2-phenylindole) dyes were used to label the nuclei. After washing with PBS, the cells were observed under a fluorescence microscope (Zeiss, Axio Imager Z2, Oberkochen, Germany). Luminex assay The concentrations of cytokines/chemokines, including TNF-α, CCL2, IL-6, and IL-10, in the culture medium were analyzed using a mouse magnetic Luminex assay (R&D Systems, Wiesbaden-Nordenstadt, Germany) as previously described ([218]48). Briefly, the diluted culture medium samples were incubated with a premixed cocktail of antibody-conjugated beads in a 96-well plate for 2 hours, followed by incubation with a cocktail of biotinylated antibodies for 1 hour and streptavidin-PE (SA-PE) for 30 min. The sample-loaded plate was washed with buffer, and the protein concentration was measured on a Luminex 200 analyzer (Luminex, Austin, TX, USA) according to the manufacturer’s instructions. Pathway inhibition experiments M1-Mφs under GLN depletion conditions were treated with GW4869 (20 μM, HY-19363, MedChemExpress, Monmouth Junction, NJ, USA) or DHMEQ (2.5 μg/ml, HY-14645, MedChemExpress) for 24 hours. After incubation for the indicated times, the cells or EVs from each group were collected for further tests. LC-MS/MS–based targeted metabolomics The intracellular metabolites of Mφs were extracted using methanol/H[2]O solution as previously described ([219]52). After washing with PBS, the cells in the wells of a six-well plate were immediately extracted with buffer (1 ml, methanol/H[2]O = 8:2, v/v) and incubated at −20°C for 30 min. After centrifugation (13,000 rpm for 20 min at 4°C), 0.7 ml of the supernatant was collected and dried in a drier (Eppendorf, Fisher Scientific, Pittsburgh, PA) at 30°C for 4 hours. The dried samples were reconstituted in 100 μl of hydrophilic interaction chromatography (HILIC) solution. Targeted MS-based metabolomics was performed using an Agilent 1260 LC (Agilent Technologies, Santa Clara, CA) coupled with an AB Sciex Qtrap 5500 MS (AB Sciex, Toronto, Canada) system. The sample solution was injected into the LC-MS/MS system and analyzed under positive (2 μl of sample solution) and negative (10 μl of sample solution) ion modes. The mobile phase, gradient conditions, and MS parameters were set up as previously described ([220]52). Multiple reaction monitoring (MRM) mode was used to detect multiple metabolites. The metabolites were normalized to the cellular protein, and FC > 1.5 and P-adjusted < 0.05 were considered significant. PCA plots and heatmaps were generated using a free online platform ([221]https://www.omicsolution.org/wkomics/main/). The differentially expressed metabolites were analyzed and visualized using MetaboAnalyst (version 5.0, [222]https://www.metaboanalyst.ca/MetaboAnalyst/ModuleView.xhtml). Mitochondrial membrane potential assay The mitochondrial membrane potential of the cell samples was analyzed using a fluorescent tetramethylrhodamine methyl ester probe (TMRM, Sigma-Aldrich). After different treatments, the cells were stained with 20 nM TMRM for 15 min at 37°C. The stained cells were washed with PBS, and the fluorescence was measured using a flow cytometer (LSRFortessa). Mitochondrial ROS measurement Intracellular levels of mitochondrial ROS were analyzed using the MitoSOX Red mitochondrial superoxide indicator (MitoSOX, Thermo Fisher Scientific). After different treatments, the cells were stained with 5 μM MitoSOX for 20 min at 37°C. The stained cells were washed with PBS, and the fluorescence was measured using a flow cytometer (LSRFortessa). Intracellular ATP measurement ATP was measured using a bioluminescence assay kit (Beyotime Biotechnology, Shanghai, China). After different treatments, the cells were lysed with 200 μl of lysis buffer provided with the kit. The supernatant was collected by centrifugation at 12,000g for 5 min at 4°C. The concentration of ATP was determined by mixing 100 μl of luciferase reagent and 20 μl of supernatant to catalyze the production of luminescence from ATP and luciferin. The luminescence of each sample was measured using a microreader (Tecan Group Ltd., Switzerland). Statistical analysis All the data are presented as the means ± SDs and were analyzed using GraphPad software (version 8.0.2, IBM Corporation, USA) with one-way analysis of variance (ANOVA) or Student’s t test; P < 0.05 was considered to indicate a significant difference. The data were obtained from at least three biological replicates, and “NS” represents the number of independent samples for each group. Acknowledgments