Graphical abstract graphic file with name fx1.jpg [39]Open in a new tab Highlights * • MAC4 shows potent antibacterial activity against multiple bacterial strains * • MAC4 prevents biofilm formation and disrupts bacterial adhesion * • MAC4 targets bacterial membranes, metabolism, and DNA * • MAC4 reduces lung inflammation and bacterial load in in vivo studies __________________________________________________________________ Pharmacology; Medical Microbiology; Immunology Introduction Bacterial infections remain a critical global health challenge, with Escherichia coli being a prominent example of a pathogen causing a wide spectrum of diseases, from mild gastroenteritis to life-threatening conditions such as septicemia and hemolytic uremic syndrome.[40]^1^,[41]^2^,[42]^3^,[43]^4^,[44]^5 The burden of E. coli infections on healthcare systems worldwide is substantial, leading to increased morbidity and mortality.[45]^6^,[46]^7 Current treatment strategies, heavily dependent on antibiotics, face mounting challenges due to antimicrobial resistance (AMR), with some bacterial strains exhibiting resistance to multiple drug classes.[47]^8^,[48]^9^,[49]^10 This crisis necessitates the urgent development of novel therapeutic approaches that can effectively combat bacterial infections while minimizing resistance development.[50]^11 One particularly challenging aspect of bacterial pathogenesis is biofilm formation - structured communities of microorganisms encased in a self-produced polymer matrix.[51]^12^,[52]^13 Within biofilms, bacteria exhibit altered phenotypes with enhanced resistance to antimicrobial agents and host immune defenses.[53]^14 For species such as E. coli, biofilm formation significantly increases virulence and complicates treatment outcomes.[54]^15^,[55]^16 The ability of biofilms to resist conventional antibiotics highlights the need for novel therapeutic strategies that can both prevent biofilm formation and eliminate established biofilms. Traditional Chinese Medicine (TCM) offers a promising avenue for discovering novel antimicrobial compounds. Recent studies have demonstrated that natural compounds derived from traditional medicines possess significant antimicrobial and anti-inflammatory properties.[56]^17^,[57]^18^,[58]^19 For instance, Rashed and Butnariu (2014) identified potent antimicrobial compounds in Bauhinia racemosa,[59]^11 while similar studies on Eriobotrya japonica revealed bioactive compounds with both antimicrobial and antioxidant properties.[60]^20 These findings suggest that traditional medicine could be a valuable source of novel therapeutic agents against bacterial infections. Our metabolomic analysis of Squama manitis, a TCM staple traditionally used for infectious diseases, revealed 78 distinct metabolites.[61]^21 Through systematic screening and analysis, we identified a novel Malic Acid Combination (MAC4), comprising malic acid, fumaric acid, glycine, and hippuric acid, that showed promising antibacterial efficacy.[62]^22^,[63]^23 MAC4 represents a convergence of traditional medicine and modern scientific approaches, distinguishing itself from conventional synthetic antimicrobials through its natural origin and complex composition. The present study aims to evaluate MAC4’s dual antimicrobial and immunomodulatory properties through comprehensive in vitro and in vivo studies. Using a multi-faceted approach combining Minimum Inhibitory Concentration (MIC) assays, biofilm formation analyses, whole-transcriptome sequencing, and a mouse model of E. coli-induced lung infection, we investigated both MAC4’s direct antibacterial effects and its ability to modulate host immune responses. This comprehensive evaluation addresses critical gaps in current antimicrobial research by exploring the compound’s capacity to combat bacterial infections while potentially minimizing inflammatory damage. Our findings could provide valuable insights into developing novel therapeutic strategies against bacterial infections, particularly those complicated by biofilm formation and inflammatory responses, contributing to the global effort to combat antimicrobial resistance through natural product discovery. Results Broad-spectrum antibacterial action of malic acid combination against key pathogens To evaluate the antibacterial potential of MAC4, we assessed its efficacy against four critical pathogenic bacteria: E. coli, S. aureus, P. aeruginosa, and S. marcescens. Comprehensive analyses, including minimum inhibitory concentration (MIC), growth curve assessment, and minimum bactericidal concentration (MBC), were conducted with varying concentrations of MAC4 ([64]Figures 1A–1D). At optimally determined concentrations (0.1C for E. coli and S. aureus; 0.2C for P. aeruginosa and S. marcescens), MAC4 exhibited significant inhibitory effects. The MICs were established at these concentrations, with growth inhibition rates at the MIC showing notable efficacy: 80% for E. coli, 67% for S. aureus, 71% for P. aeruginosa, and 73% for S. marcescens ([65]Figures 1A–1D). In 24-h antimicrobial assays, ciprofloxacin (CIP) demonstrated excellent antibacterial activity with inhibition rates exceeding 97% against all four tested strains (E. coli, S. aureus, P. aeruginosa, and S. marcescens), compared to MAC4’s inhibition rates of 67–80%. Longitudinal monitoring over 24 h at the MIC revealed that MAC4 consistently suppressed bacterial proliferation across all tested species, affirming its potent inhibitory capability ([66]Figures 1E–1H). Figure 1. [67]Figure 1 [68]Open in a new tab MAC4 exhibits broad-spectrum antibacterial activity (A–D) Minimum inhibitory concentration (MIC) of MAC4 and CIP against E. coli, S. aureus, P. aeruginosa, and S. marcescens determined by broth microdilution assay after 24-h exposure. Data points represent mean ± SEM. (n = 3 independent experiments). (E–H) Growth curves of E. coli, S. aureus, P. aeruginosa, and S. marcescens treated with various concentrations of MAC4 and CIP over time. Data points represent mean ± SEM. (n = 3 independent experiments). (I) Antibacterial activity of MAC4 against a panel of Gram-positive and Gram-negative bacteria. To further elucidate the bactericidal properties of MAC4, MBC assays were conducted at multiples of the MIC (1MIC, 2MIC, and 4MIC). The bactericidal thresholds for E. coli and S. aureus were identified at 4MIC (0.4C), highlighting a robust bactericidal effect. Conversely, for P. aeruginosa and S. marcescens, the effective MBC was at 1MIC (0.2C), underscoring the compound’s broad-spectrum bactericidal properties against diverse bacterial strains ([69]Figure 1I). The findings from this study underscore the remarkable antibacterial activity of MAC4 against a spectrum of pathogenic bacteria, presenting a notable inhibitory and bactericidal capacity. The ability of MAC4 to effectively inhibit and eradicate bacterial cells at specific concentrations suggests its utility as a potent broad-spectrum antibacterial agent. These results not only affirm MAC4’s therapeutic potential but also contribute to the global effort in combating bacterial infections, offering a promising avenue for the development of antimicrobial strategies. Disrupting the shield: malic acid combination’s multidimensional assault on E. coli and S. aureus biofilms, morphology, and genetic integrity To evaluate the potential of MAC4 in inhibiting biofilm formation by both E. coli and S. aureus, we conducted biofilm formation assays, metabolic activity of biofilms assays, and scanning electron microscopy (SEM) analysis. Biofilm formation was evaluated using a crystal violet staining method, which quantitatively measures the total biomass of the biofilm. The absorbance at 595 nm was used to assess biofilm mass after treatment with MAC4, and the inhibition was calculated as a percentage of the untreated control. Our results revealed that the biofilm formation of both E. coli and S. aureus was inhibited across all tested concentrations of MIC when compared to untreated controls ([70]Figures 2A and 2B). Notably, treatment groups with concentrations at 2-fold and 4-fold the MIC of MAC4 resulted in a marked inhibition of biofilm formation. We observed a positive correlation between higher concentrations of MAC4 and greater inhibition of biofilm formation in both bacterial species. This outcome aligns with the results of metabolic activity within the biofilms as determined by the MTT assay, which evaluates the metabolic activity of live cells within the biofilm. After treatment with MAC4 at 2MIC and 4MIC concentrations, the metabolic activity of the biofilms decreased by over 94.97%, suggesting a reduction in vitality or biofilm density ([71]Figure 2C). Interestingly, MAC4 showed comparable efficacy to ciprofloxacin at 2MIC concentration, particularly in biofilm formation inhibition and metabolic activity reduction assays ([72]Figures 2A–2C). Both compounds achieved >90% reduction in biofilm metabolic activity, suggesting MAC4’s particular effectiveness against bacterial biofilms. Figure 2. [73]Figure 2 [74]Open in a new tab MAC4 inhibits biofilm formation in E. coli and S. aureus (A, B) Inhibition of E. coli and S. aureus biofilm formation by different concentrations of MAC4 and CIP, assessed by crystal violet (CV) staining. Data represent mean ± SEM (n = 3 independent experiments; ^∗∗p < 0.01, ^∗∗∗p < 0.001, ^∗∗∗∗p < 0.0001, t-test). (C) Metabolic activity of E. coli and S. aureus biofilms treated with MAC4 and CIP, determined by MTT assay. Data represent mean ± SEM. (n = 3 independent experiments; ^∗∗p < 0.01, ^∗∗∗∗p < 0.0001, t-test). (D) Scanning electron microscopy (SEM) images of E. coli and S. aureus biofilms after 24 h treatment with 0.1× MIC MAC4 compared to untreated controls. Representative scale bars: 50 μm (left column), 5 μm (middle column), and 1 μm (right column). (E) TEM images of E. coli and S. aureus treated with 0.1× MIC MAC4 compared to untreated controls. MAC4 treatment resulted in 1. uneven cytoplasm distribution, 2. blurred cell edges, 3. content overflow, 4. increased cell edge transparency, 5. plasmolysis, and 6. DNA condensation (red arrows). Representative scale bars: 1 μm (left column), 500 nm (middle column), and 200 nm (right column). Further SEM analysis showed a reduction in the biofilm density and disruption of the cell structure of both E. coli and S. aureus after MAC4 treatment when compared to control groups. ([75]Figure 2D). Notably, in the E. coli control group, bacterial cells were connected by filamentous structures, likely composed of extracellular DNA (eDNA) or matrix fibers,[76]^24 components of the biofilm matrix that provide a framework for bacterial survival and motility.[77]^25^,[78]^26 However, we observed a significant reduction in the connectivity of these structures in E. coli and S. aureus after MAC4 treatment ([79]Figure 2D). This suggests a possible direct action of MAC4 on the eDNA, leading to its denaturation and inactivation, as well as affecting the synthesis of matrix fibers, thereby diminishing their connectivity. These findings indicate that MAC4 exhibits a capacity to effectively mitigate biofilm formation and may disrupt interbacterial interactions within biofilms. Morphological alterations in E. coli and S. aureus cells Transmission electron microscopy (TEM) was employed to discern the ultrastructural impacts of MAC4 on E. coli and S. aureus. Comparative analysis between untreated controls and MAC4-treated bacteria unveiled stark morphological contrasts. In controls, E. coli and S. aureus preserved their characteristic rod and cocci shapes, respectively, with intact cell walls, clearly defined membranes, and homogeneously dense cytoplasm ([80]Figure 2E). Conversely, MAC4 treatment instigated significant morphological disruptions, including uneven cytoplasm distribution, edge blurring, content overflow, increased cell edge transparency, plasmolysis, and DNA condensation. These structural alterations suggest compromised cellular integrity, potentially hindering critical bacterial functions, diminishing viability, and leading to cell destruction. Reduced bacterial adhesion post-malic acid combination treatment Further investigations into MAC4’s effects on bacterial adhesion employed FITC-labeled E. coli to quantify adhesion efficiency to human HaCaT cells before and after MAC4 exposure. Our results demonstrated a dramatic decrease in E. coli adhesion post-treatment ([81]Figure 3A). Initially, untreated E. coli showed a 6.85% adhesion rate to HaCaT cells, which significantly dropped to 0.81% following MAC4 treatment, marking an 8.5-fold reduction in adhesion capability. This pronounced decline in adhesion efficiency after 24 h of MAC4 exposure underscores a substantial impairment in the bacterial adhesion process. Figure 3. [82]Figure 3 [83]Open in a new tab MAC4 induces reduces bacterial adhesion and alters gene expression in E. coli (A) Adhesion rate of E. coli to HaCaT cells following MAC4 treatment (mean ± SEM, n = 3 independent experiments; ^∗∗∗∗p < 0.0001, t-test). (B) Volcano plot of DEGs in MAC4-treated E. coli. Gray dots: non-significant genes; red dots: upregulated DEGs; blue dots: downregulated DEGs. Horizontal axis: log2(fold change); vertical axis: -log10(q-value). (C) GO term enrichment and (D) KEGG pathway analysis of downregulated DEGs in MAC4-treated E. coli. (E) Correlation between RNA-seq and RT-PCR data for selected DEGs. R^2: coefficient of determination for the linear regression. Transcriptomic insights unveil malic acid combinations antimicrobial mechanism against E. coli To study how bacteria respond to MAC4 at the transcriptomic level, we sequenced and compared the gene expression profiles of MAC4-treated E. coli cells to untreated controls, using four biological replicates for each. We identified 1,150 differentially expressed genes (DEGs), with 490 being upregulated and 660 downregulated ([84]Figure 3B and [85]Table S1). Gene Ontology (GO) enrichment for downregulated genes revealed a focus on membrane-related components, specifically integral components of the membrane (GO:0005886), plasma membrane (GO:0005886), and outer membrane (GO:0019867) ([86]Figure 3C). This suggests the MAC4’s targeted disruption of membrane integrity and function. Moreover, downregulated DEGs showed enrichment in DNA binding and recombination activities, indicating an impairment in genetic processes critical for bacterial survival and adaptation. KEGG pathway analysis via Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed significant enrichment in pathways such as Glycerophospholipid metabolism (ko00564), ABC transporters (ko02010), DNA replication (ko03030), Lipopolysaccharide biosynthesis (ko00540), and Bacterial invasion of epithelial cells (ko05100) ([87]Figure 3D). Notably, the ABC transporter pathway, crucial for nutrient import and toxin export,[88]^27 emerged as a major target of downregulation, alongside pathways involved in energy metabolism and biofilm formation. These insights suggest that MAC4’s bactericidal mechanism encompasses broad-spectrum interference with essential bacterial functions, from membrane integrity to energy metabolism and genetic processes. To ascertain the accuracy of our DEG findings, we conducted RT-qPCR validation on a random subset of 10 genes, employing the rrsG (16S rRNA gene) as a normalization control. The high correlation coefficient (R^2 = 0.77) between RT-qPCR and RNA-Seq data validates the reliability of our identified DEGs ([89]Figure 3E and [90]Table S2). Our study elucidates the complex antimicrobial mechanism of MAC4, demonstrating its dual action in not only preventing biofilm formation and bacterial adhesion but also disrupting bacterial morphology and cellular processes. The observed transcriptomic alterations further reveal MAC4’s capacity to target critical pathways essential for bacterial viability and communication. Malic acid combination modulates immune responses and preserves lung integrity against E. coli infection To assess MAC4’s antibacterial effectiveness in vivo, we utilized an ELI model in mice. Intratracheal inoculation with E. coli at a concentration of 1 × 10^9 CFU/mL was followed by the administration of MAC4 at dosages ranging from 0.5 to 1.5 g/kg/d. Lung bacterial counts, conducted three days post-infection, revealed a significant decrease in bacterial load in mice treated with MAC4 compared to controls ([91]Figure 4A). This demonstrates MAC4’s potent antibacterial activity in a live animal model, providing a promising outlook for its therapeutic application. Figure 4. [92]Figure 4 [93]Open in a new tab MAC4 treatment reduces bacterial load and modulates inflammatory cytokine expression in an E. coli-induced pneumonia (ELI) mouse model (A) Bacterial load in lung tissues of ELI mice treated with MAC4 or CIP. Data represent mean ± SEM (n = 6 mice per group; compared with control group, ^###p < 0.001, t-test; compared with model group, ^∗∗∗p < 0.001, t-test). (B) Representative hematoxylin and eosin (HE) stained lung sections displaying histopathological changes. Scale bars: 100 μm for the upper row of images and 200 μm for the lower row of magnified images. (C) mRNA levels of inflammatory cytokines IL-10, IL-6, IL-1β, and TNF-α in mouse lungs analyzed by RT-qPCR, normalized to GAPDH. Data represent mean ± SEM (n = 4 mice per group; ^∗∗p < 0.01; t-test). (D) Protein expression levels of IL-10, IL-6, IL-1β, and TNF-α detected by western blotting, with β-Actin as a loading control. Quantification of normalized protein expression levels is shown on the right. Data represent mean ± SD (n = 4 mice per group; ^∗p < 0.05, ^∗∗p < 0.01, ^∗∗∗p < 0.001, ^∗∗∗∗p < 0.0001; t-test). Further insights were gained through the histopathological examination of lung tissues to evaluate structural changes and inflammation levels. Normal lung architecture characterized the control group, with intact alveolar spaces and minimal inflammatory cell infiltration ([94]Figure 4B). In contrast, the model group displayed signs of infection-induced alterations, such as increased inflammatory cell infiltration (evident as dense accumulations of dark purple cells, indicated by blue arrows), obscured alveolar spaces indicative of edema or exudate, and thickened alveolar walls reflecting inflammation. Treatment with MAC4, similar to ciprofloxacin (a known drug to treat pneumonia), showed a notable improvement in lung tissue condition. All treated groups exhibited reduced cellularity, with a clear dose-dependent effect observed in the MAC4-treated groups. The extent of inflammatory cell infiltration was determined by the presence and density of dark purple cell clusters, which were substantially reduced in the treatment groups, particularly at medium and high doses of MAC4. All treated groups displayed clearer alveolar structures, and diminished vascular and bronchiole wall thickening, with MAC4 treatment at medium and high doses markedly alleviating inflammation (as evidenced by the near-absence of dark cell clusters) and preserving lung integrity in the E. coli-induced pneumonia mouse model. Analysis of endogenous lung RNA levels of inflammatory markers in an ELI model showed a significant decrease in pro-inflammatory cytokines (IL-6, IL-1β, TNF-α) following MAC4 treatment ([95]Figures 4C and 4D and [96]Data S1). However, the increase in the anti-inflammatory cytokine IL-10 was not statistically significant (p > 0.05). These results, supported by both RT-PCR and western blot analyses, suggest that MAC4 not only acts as an antibacterial agent but also modulates the host immune response, reducing inflammation. Altogether, our data showed that MAC4 demonstrates potential as an effective antibacterial and immunomodulatory agent for bacterial pneumonia, with implications for managing various bacterial lung infections and associated inflammation. Malic acid combination-induced immunomodulation enhances host defense against lung infection: transcriptomic evidence To investigate the host response to MAC4 treatment in an E. coli-induced pneumonia (ELI) mouse model, we performed transcriptomic analysis of lung tissues before and after MAC4 administration. RNA-Seq analysis identified 535 differentially expressed genes (DEGs), with 310 upregulated and 225 downregulated genes post-MAC4 treatment ([97]Figure 5A and [98]Table S3). The validation of selected DEGs via RT-qPCR demonstrated a strong positive correlation (R^2 = 0.85) with RNA-Seq data, confirming the reliability of the transcriptomic findings ([99]Table S4 and [100]Figure 5F). Notably, MAC4 treatment upregulated genes involved in immune response and neutrophil production, such as Csf3 and Alox15, suggesting an enhancement of host defense mechanisms[101]^28 Concurrently, the downregulation of genes such as Ccl7 and Adtrp indicated a strategic modulation of immune responses, potentially reducing excessive inflammation and tissue damage. Figure 5. [102]Figure 5 [103]Open in a new tab MAC4 treatment alters gene expression in the ELI mouse model (A) Volcano plot of DEGs in lung tissues of MAC4-treated ELI mice compared to controls. Red dots: upregulated genes; blue dots: downregulated genes. The top 10 up-and-down-regulated genes by fold change are labeled. GO enrichment analysis of (B) upregulated and (C) downregulated DEGs. KEGG pathway enrichment analysis of (D) upregulated and (E) downregulated DEGs. (F) Correlation between RNA-Seq and RT-qPCR data for selected DEGs. R2: coefficient of determination for the linear regression. Data points represent mean ± SEM. (n = 4 independent experiments). Gene enrichment analyses revealed that upregulated genes were predominantly involved in immune response processes (GO:0002376 and GO:0045087), transcriptional regulation (GO:0006357 and GO:0000122), and metabolic adjustments (GO:0006629) essential for infection control ([104]Figure 5B). Conversely, downregulated genes were associated with cell adhesion (GO:0007155) and apoptotic pathways (GO:0006915), suggesting mechanisms to restrict pathogen dissemination and cell death ([105]Figure 5C). The decreased expression in inflammatory response (GO:0006954) could potentially reduce post-infection inflammation. Moreover, alterations in the lipid metabolic process (GO:0006629) may affect the integrity and functionality of cell membranes, crucial for defending against pathogen invasion ([106]Figure 5C). KEGG pathway analysis identified upregulation in immune-related pathways, including cytokine-cytokine receptor interaction (mmu04060) and Toll-like receptor signaling (mmu04620), indicating enhanced immune response ([107]Figure 5D). Downregulation of pathways such as the PI3K-Akt signaling pathway (mmu04151) suggested reduced inflammatory cell activation and survival, contributing to decreased inflammation ([108]Figure 5E). Altogether, our data demonstrate that MAC4 inhibits pathogen proliferation and modulates the host’s immune response during lung infection, highlighting its therapeutic potential against bacterial pathogens. Discussion Bacterial infections pose a significant global health challenge, particularly due to increasing antibiotic resistance and the limitations of current therapeutic approaches. Our study introduces MAC4, a novel therapeutic compound derived from traditional Chinese medicine, which demonstrates both potent antibacterial activity and immunomodulatory effects. This dual-action mechanism distinguishes MAC4 from conventional antibiotics, which typically focus solely on bacterial elimination. MAC4’s broad-spectrum activity is particularly noteworthy when compared to conventional antibiotics, which often target specific bacterial groups. The concentration-dependent bactericidal effect of MAC4 suggests a mechanism of action that could potentially overcome antibiotic resistance, a growing concern in current antimicrobial therapies.[109]^29 Moreover, MAC4’s ability to disrupt biofilm formation addresses a critical challenge in infection treatment, as biofilms are major contributors to antibiotic resistance and treatment failure.[110]^30^,[111]^31^,[112]^32 Mechanistically, our transcriptomic analysis revealed that MAC4 targets multiple cellular processes simultaneously. The observed downregulation of genes (e.g., evgA, yihL, fimB, fimE) involved in membrane components, DNA binding, and recombination activities indicates that MAC4 may target these crucial processes, leading to the disruption of bacterial cell integrity, impaired DNA replication and repair, and ultimately, cell death.[113]^33^,[114]^34 Furthermore, the downregulation of metabolic pathways, such as glycerophospholipid metabolism and ABC transporters, suggests that MAC4 may interfere with energy production and nutrient acquisition, hindering bacterial growth and survival.[115]^35^,[116]^36 These findings suggest MAC4 acts through multiple complementary mechanisms affecting bacterial cellular processes. Our structural investigations have provided additional mechanistic insights. TEM analysis demonstrated clear morphological alterations, including disrupted cell wall integrity, altered cytoplasmic distribution, and DNA condensation. Similarly, SEM analysis showed significant disruption of biofilm architecture and bacterial connectivity. These structural changes are further supported by functional evidence, including an 8.5-fold decrease in bacterial adhesion capacity, significant inhibition of biofilm formation, and reduced metabolic activity in biofilms by more than 94.97%. The immunomodulatory properties of MAC4 represent a particularly promising aspect of its therapeutic potential. Our findings demonstrate that MAC4 can effectively balance the host’s pro- and anti-inflammatory responses, as evidenced by the modulation of cytokines and immune cell receptors. The upregulation of genes associated with cytokine signaling (e.g., Csf3, Il4i1, Ccl4, Tnf)[117]^37^,[118]^38^,[119]^39 and immune cell receptor function (e.g., Cd180, Fcrl1, Fcer1g)[120]^40^,[121]^41 suggests that MAC4 promotes host immune responses. Additionally, MAC4 downregulated genes associated with cell adhesion molecules (e.g., Itga3, Itga8),[122]^42 extracellular matrix (e.g., Col12a1, Col6a1),[123]^43 and cell cycle/apoptosis (e.g., Bcl2l1),[124]^44 suggesting the potential suppression of excessive inflammation and tissue damage. These findings suggest that MAC4’s therapeutic effect extends beyond direct bacterial killing, encompassing a strategic modulation of host immune responses to favor recovery and minimize collateral tissue damage. The implications of our findings extend beyond acute bacterial infections. MAC4’s antibacterial, antibiofilm, and anti-inflammatory properties make it particularly promising for treating conditions where infection and inflammation are interlinked, such as chronic infections or inflammatory diseases.[125]^45^,[126]^46^,[127]^47 By reducing bacterial load, preventing infection-induced inflammatory responses, and inhibiting biofilm formation, MAC4 may help mitigate disease progression. However, given the complex nature of many such conditions, which often involve multiple factors,[128]^48^,[129]^49^,[130]^50 additional therapeutic approaches targeting these contributing factors may be necessary to complement MAC4’s effects. In conclusion, MAC4 represents a promising therapeutic agent that addresses both the direct challenges of bacterial infection and the associated inflammatory responses. Its mechanism of action, combined with its origin in traditional medicine, exemplifies the potential of integrating ancient medical knowledge with modern scientific approaches. Further development of MAC4 could lead to more effective treatments for bacterial infections and inflammatory conditions, particularly in cases where conventional antibiotics alone may be insufficient. Limitations of the study Several limitations of our study warrant consideration. First, while we tested MAC4 against key pathogens, future research should evaluate its efficacy against a broader spectrum of bacteria, particularly drug-resistant strains. Second, although we demonstrated MAC4’s immunomodulatory effects, the complete mechanisms underlying its interaction with host immune pathways require further investigation. Third, all experiments were conducted using six-week-old female ICR mice. The exclusive use of female mice may limit the generalizability of our findings. Long-term studies in animal models of chronic conditions would be valuable in assessing MAC4’s safety and efficacy for extended use. Future research should focus on First structure-activity relationship studies to optimize MAC4’s composition. Second, the exploration of synergies with conventional antibiotics for combination therapies. Third, deeper mechanistic studies using knockout mutants, the investigation of the potential synergistic effects between MAC4 components, metabolomic profiling (to track cellular metabolic changes), and advanced imaging techniques for studying membrane integrity. Fourth, the inclusion of both male and female animal models to evaluate the influence of sex on MAC4’s efficacy and safety. These studies would facilitate MAC4’s development toward clinical applications. Resource availability Lead contact Requests for further information and resources should be directed to and will be fulfilled by the lead contact, Siew Woh Choo (cwoh@wku.edu.cn). Materials availability This study did not generate new unique reagents. Data and code availability * • Data: Raw sequence reads data have been deposited at NCBI Sequence Read Archive (SRA) and are publicly available as of the date of publication. Accession numbers are listed in the [131]key resources table. * • Code: This article does not report the original code. * • All other requests: Any additional information required to reanalyze the data reported will be shared by the [132]lead contact upon request. Acknowledgments