{"title":"Limosilactobacillus fermentum 21 N1 Mitigates Candida albicans-Induced Vulvovaginal Candidiasis and Suppresses NLRP3 Inflammasome Activation in a Mouse Model.","authors":"Eun Yeong Lim, Gun-Dong Kim, Geun-Hye Hong, So-Young Lee, So-Young Lee, Hee Soon Shin","doi":"10.1007/s12602-025-10619-x","DOIUrl":"https://doi.org/10.1007/s12602-025-10619-x","url":null,"abstract":"<p><p>Vulvovaginal candidiasis (VVC), primarily caused by Candida albicans, is a common fungal infection that typically causes inflammation and discomfort. Current antifungal treatments, such as fluconazole, are associated with adverse effects and drug resistance, highlighting the need for alternative therapies. In this study, we investigated the antifungal efficacy of Limosilactobacillus fermentum 21N1 (LF21) against C. albicans using both in vitro assays and a murine model of VVC. A spotting assay assessed the inhibitory activity of LF21 against C. albicans. A murine VVC model was established by intravaginally inoculating C. albicans. Fungal burden, inflammation, and histopathological changes were assessed using colony counting, H&E, and PAS staining. Immunofluorescence and Western blot analyses quantified the expression of NLRP3 inflammasome and pyroptosis-related proteins. In vitro experiments using a spot formation assay revealed that LF21significantly inhibited C. albicans growth. Additionally, oral administration of LF21 alleviated VVC symptoms in mice inoculated with C. albicans, reducing the vaginal fungal burden and IL-1β levels. Histological analysis revealed reduced polymorphonuclear neutrophil infiltration and decreased C. albicans presence in the vaginal tissues of LF21-treated mice. LF21 treatment significantly reduced the expression of NLR family pyrin domain containing 3 (NLRP3) inflammasome components and pyroptosis-related proteins, including gasdermin-D, which were upregulated in response to the inoculation of C. albicans. These findings suggest that LF21 exerts antifungal effects by modulating the NLRP3 inflammasome and consequently reducing inflammation, indicating its potential as a novel therapeutic agent for treating VVC.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Probiotics and Postbiotics Derived from Saline/Marine Plant-Based Feedstocks.","authors":"Stanislav Rudnyckyj, Mette Hedegaard Thomsen","doi":"10.1007/s12602-025-10617-z","DOIUrl":"https://doi.org/10.1007/s12602-025-10617-z","url":null,"abstract":"<p><p>The growing demand for the sustainable and cost-effective production of probiotics and postbiotics has highlighted the potential of saline and marine plants as novel substrates. These plants, including seaweeds and halophytes, are abundant and nutrient-rich and require minimal resources, making them ideal candidates for green biorefineries. The incorporation of saline plant-based feedstocks could lower media costs and environmental impact, as these plants do not require arable land or freshwater while contributing to carbon sequestration and sustainable farming. The development of integrated biorefineries could drive economic feasibility by facilitating cost-effective probiotic and postbiotic production. However, challenges such as high salt content and lignocellulosic composition may complicate microbial fermentation. This review examines recent advancements in leveraging naturally salt-tolerant probiotics and efficient bioconversion methods to address these challenges. It explores the nutritional profiles of saline plants, their prebiotic potential, and their synergetic compatibility with diverse probiotic strains, including probiotic bacteria and fungi and their metabolites. Additionally, the review discusses state-of-the-art fermentation techniques tailored to saline plant-based substrates and the possible advantages of saline feedstocks for probiotics and postbiotics production through biorefinery pathways. The work highlights the transformative potential of saline and marine plant-derived probiotics and postbiotics in health supplementation and biotechnological innovation, contributing to biorefinery development within a circular economy framework.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abdallah S I Hassan, Ahmed R Henawy, Youssef A Saied, Karen A Garas, Omar M Shahat, Asmaa A Halema
{"title":"Direct-fed Microbials (DFM) and Poultry Genomics: A Synergistic Approach to Sustainable Antibiotic Free Farming.","authors":"Abdallah S I Hassan, Ahmed R Henawy, Youssef A Saied, Karen A Garas, Omar M Shahat, Asmaa A Halema","doi":"10.1007/s12602-025-10618-y","DOIUrl":"https://doi.org/10.1007/s12602-025-10618-y","url":null,"abstract":"<p><p>Improper usage of antibiotics in poultry production is a great threat to the ecosystem because their residues can enter into the food chain or leach into soil or water systems and increase antibiotic resistance risks. Hence, direct-fed microbials (DFMs) have gained recognition as a sustainable and viable alternative to antibiotics in poultry production, capitalizing on the relationship between microbial genetics, host genomics, and gut microbiota. This review delves into the genetic and host genomic mechanisms through DFMs effects including the enhancement of nutrient metabolism, modulation of gut microbiota and strengthening of the host immunity. The revolution of multi-omics has participated in the identification of probiotic strains with desirable traits and revealed their impact on host gene expression, particularly in genes related to intestinal health, such as tight junction proteins and mucins. Furthermore, the review summarizes the benefits of using DFMs in poultry production, the factors affecting their efficacy and their challenges and limitations. Future research integrating host and microbial genomics, along with precision microbiome engineering, holds promise for maximizing the potential of DFMs in advancing sustainable poultry farming practices.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144294813","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Development and Techno-functional Characterization of Beyaz Cheese Fortified with Walnut (Juglans regia L.) Leaf Powder and Lactobacillus acidophilus LA-5.","authors":"Mehmet Ali Salık, Songül Çakmakçı","doi":"10.1007/s12602-025-10629-9","DOIUrl":"https://doi.org/10.1007/s12602-025-10629-9","url":null,"abstract":"<p><p>This study aimed to produce a new functional Beyaz cheese variety with the addition of different ratios (0.5% and 1%) of walnut leaf powder (WLP) and probiotic bacteria (Lactobacillus acidophilus LA-5.). The probiotic shelf-life and techno-functional properties of Beyaz cheeses were investigated at 30-day intervals during ripening in brine (120 days at + 4 °C). The highest acidity, ash, salt, Fe, Cu, Mn, a*, b*, H°, and C* values were found in the WLP-supplemented cheeses compared with those in the control without probiotic (C) and probiotic control (PC) cheeses (p < 0.01). The count of L. acidophilus LA-5 in probiotic cheeses decreased to less than 10<sup>6</sup> colony-forming units (CFU)/g after the 60th day of storage. Compared with PC (77.52%), WLP increased the viability rate (82.04-86.52%) over the 60-day shelf-life period. The total phenolic content (TPC), total flavonoid content (TFC), chlorophyll a and chlorophyll b contents, and total antioxidant activity (DPPH•, CUPRAC, and FRAP assays) of the cheeses significantly increased with the addition of 0.5 and 1% WLP (p < 0.01). The addition of WLP increased proteolysis (WSN, pH 4.6‒SN, TCA‒SN and PTA-SN) and lipolysis (p < 0.01). The general acceptability of cheese samples was PC > C > PWL<sub>0.5%</sub> > PWL<sub>1%</sub>. Consequently, a new variety of functional Beyaz cheese with a probiotic shelf life of 60 days that is rich in bioactive compounds and microminerals (Fe, Cu and Mn) and natural for healthy nutrition can be produced with the addition of L. acidophilus LA-5 and 0.5% WLP.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144294812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Immunoenhancing Effect of Lactobacillus Kefiranofaciens ZW18 Postbiotic On Cyclophosphamide-induced Immunosuppressed Mice.","authors":"Hanxue Xu, Xinyuan Li, Weitao Geng, Longgang Jia, Yanping Wang, Jinju Wang","doi":"10.1007/s12602-025-10630-2","DOIUrl":"https://doi.org/10.1007/s12602-025-10630-2","url":null,"abstract":"<p><p>Lactobacillus kefiranofaciens, a lactic acid bacterium isolated from kefir grains, has various probiotic functions, including antioxidant activity, improvement of intestinal health, and immunomodulation. However, the effect of its postbiotic on immune modulation has been rarely reported. In this study, the immunomodulatory activities of the Lactobacillus kefiranofaciens ZW18 postbiotic (Post-ZW18) were investigated both in vitro and in vivo. We discovered that Post-ZW18 increased the phagocytosis of RAW264.7 cells (P > 0.05) and promoted the secretion of cytokines, including NO, IL-6, and TNF-α (P < 0.05), thereby activating the immune response in macrophages in vitro. In vivo, Post-ZW18 reversed cyclophosphamide (CTX)-induced weight loss and organ damage in immunosuppressed mice (P < 0.05), and enhanced the immune response by activating the TLR2/NF-κB signaling pathway, promoting T cell activation and differentiation, and modulating the production of cytokines such as TNF-α, IL-6, and INF-γ (P < 0.05). Furthermore, Post-ZW18 remodeled the intestinal flora, increased the Firmicutes/Bacteroidetes (F/B) ratio (P > 0.05), and upregulated the abundance of bacteria associated with immune activation, specifically increasing unclassified_Lachnospiraceae, Odoribacter, and Lachnospiraceae_NK4A136_group (P < 0.05). In summary, this study provided evidence that Post-ZW18 may serve as a novel dietary supplement for immune regulation.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144286394","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Novel Approaches in Establishing Chemical Food Safety Based on the Detoxification Capacity of Probiotics and Postbiotics: A Critical Review.","authors":"Amin Abbasi, Elham Sheykhsaran, Negin Hosseinzadeh, Marjan Bazdar, Mohsen Hamehjani, Baharak Aghapour, Saeedeh Shojaee-Aliabadi","doi":"10.1007/s12602-025-10615-1","DOIUrl":"https://doi.org/10.1007/s12602-025-10615-1","url":null,"abstract":"<p><p>Environmental and chemical contaminants from industrial and agricultural sources increasingly threaten food safety worldwide. These contaminants, including bacterial toxins (e.g., botulinum toxin), mycotoxins (e.g., aflatoxin, ochratoxin), pesticides, heavy metals, and microplastics, pose significant health and economic risks. Emerging evidence highlights that chronic exposure to such xenobiotics disrupts gut microbiota structure and function, adversely affecting host health. While traditional physicochemical detoxification methods exist, they often require impractical conditions. Biological detoxification via probiotics and their metabolites (postbiotics) has gained attention as a practical and cost-effective alternative. Numerous studies concur that probiotics can bind, biotransform, or sequester xenobiotics, thereby mitigating toxicity and restoring microbiota balance. However, variations in strain efficacy, binding mechanisms, and in vivo performance indicate areas needing further exploration. This review critically synthesizes current knowledge on probiotic-mediated detoxification mechanisms, their interaction with xenobiotics and the gut microbiome, and host responses, highlighting consensus, discrepancies, and gaps to inform future research and applications.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144286395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ji Hye Choi, Sehyeon Song, Min Ji Jang, Md Ariful Haque, Hye Eun Lee, Da Hui Kim, Yeo Ju Kim, Ja Won Cho, Jin Seok Moon, Keon Heo, Myeong Soo Park, Seockmo Ku
{"title":"A Randomized, Double-Blind, Placebo-Controlled Study on Probiotic Treatment for Halitosis: Novel Insights into Glucose and Phosphorus Metabolism.","authors":"Ji Hye Choi, Sehyeon Song, Min Ji Jang, Md Ariful Haque, Hye Eun Lee, Da Hui Kim, Yeo Ju Kim, Ja Won Cho, Jin Seok Moon, Keon Heo, Myeong Soo Park, Seockmo Ku","doi":"10.1007/s12602-025-10603-5","DOIUrl":"https://doi.org/10.1007/s12602-025-10603-5","url":null,"abstract":"<p><p>Halitosis, or bad breath, is associated with oral microbial imbalances and the production of volatile sulfur compounds (VSCs). While existing treatments target pathogenic bacteria or oral health indicators, they may not address the underlying systemic complexity. This study explored the efficacy of Complex OK oral probiotics containing Lactobacillus gasseri HHuMIN D and L. paracasei OK in mitigating halitosis by evaluating VSC levels and metabolic markers. A 12-week, randomized, double-blind, placebo-controlled clinical trial was conducted involving 80 participants, 70 of whom completed the study in South Korea (KCT0009894). The participants were selected based on the presence of 2 of 3 pathogenic oral bacteria (F. nucleatum, P. gingivalis, and P. intermedia) and baseline VSCs > 2.0 ng/10 mL. Exclusion criteria included systemic diseases, recent antibiotic/probiotic use, and severe dental conditions. Oral health, VSCs, and metabolic markers were assessed using paired t-tests, ANCOVA, and Wilcoxon rank-sum tests. Despite unchanged oral health indicators and levels of harmful bacteria, probiotic supplementation showed efficacy in maintaining microbial balance. Significant reductions in H₂S and total VSCs were observed in the experimental group compared to the placebo (P < 0.05). No significant changes were observed in oral health indices or levels of harmful oral bacteria, but the experimental group showed a significant decrease in blood glucose (P = 0.009) and an increase in phosphorus levels (P < 0.05). This study provides the first published evidence linking systemic metabolic regulation to halitosis reduction, suggesting that probiotics mitigate bad breath through glucose and phosphorus metabolism rather than by direct bacterial inhibition. Further research is needed to confirm these findings and to explore the underlying mechanisms.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144286392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aleksander Gryciuk, Małgorzata Milner-Krawczyk, Marta Rogalska, Artur Krzysztof Banach, Ewa Sitkiewicz, Magdalena Bakun, Magdalena Edyta Świadek, Jolanta Mierzejewska
{"title":"Characteristics of Two Saccharomyces cerevisiae Strains and Their Extracellular Vesicles as New Candidates for Probiotics.","authors":"Aleksander Gryciuk, Małgorzata Milner-Krawczyk, Marta Rogalska, Artur Krzysztof Banach, Ewa Sitkiewicz, Magdalena Bakun, Magdalena Edyta Świadek, Jolanta Mierzejewska","doi":"10.1007/s12602-025-10624-0","DOIUrl":"https://doi.org/10.1007/s12602-025-10624-0","url":null,"abstract":"<p><p>There is a huge disparity between the number of bacterial and yeast probiotics in favor of the former. The latest reports indicate that extracellular vehicles (EVs) play a significant role in probiotic mechanisms. In the present work, we compared the probiotic properties of Saccharomyces cerevisiae strains (WUT3 and WUT151), which have never been previously characterized in this context, with commercial probiotic yeast-Saccharomyces cerevisiae var. boulardii CNCM-745. Notably, WUT3 and WUT151 reacted more mildly to the unfavorable simulated environment of saliva, stomach, small, and large intestines. As a result, we confirmed that WUT3 and WUT151 were superior to S. boulardii in terms of probiotic properties. Then, we performed a complex analysis of their EVs, isolated by a multistep filtration process. The nanoparticle tracing analysis showed no significant difference in the diameter of the vesicles between the strains. MTT studies confirmed that EVs are not toxic against normal human colorectal cell lines CCD-18 Co and CCD 841 CoN. However, toxicity was observed against the HT-29 cancer line. By staining EVs with Nile Red, we successfully visualized EVs-cell interactions. Finally, we explored the profile of proteins transported with the EVs, identifying a significant overrepresentation of extracellular proteins. Based on comparison with other proteomic data, we selected marker proteins for S. cerevisiae EVs. This knowledge will be helpful for further studies on tracking the transfer of the protein cargo of yeast EVs to human cells using, for instance, specific antibodies to these marker proteins.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144286393","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Elizabeth Annie George, Aniket Naha, H Soundharya, Janardhan Pallavi, Anushka Menon, Anand Anbarasu, Sudha Ramaiah
{"title":"Pharmacokinetics Screening, Molecular Docking, and Dynamics Simulations Revealed Novel Antimicrobial Peptide NKLF2 Mutants as Potent Inhibitors of Mycobacterium tuberculosis.","authors":"Elizabeth Annie George, Aniket Naha, H Soundharya, Janardhan Pallavi, Anushka Menon, Anand Anbarasu, Sudha Ramaiah","doi":"10.1007/s12602-025-10608-0","DOIUrl":"https://doi.org/10.1007/s12602-025-10608-0","url":null,"abstract":"<p><p>The increasing threat of multidrug-resistant Mycobacterium tuberculosis (Mtb) underscores the urgent need for novel therapeutics that can circumvent existing resistance mechanisms. The clinical ineffectiveness of current treatment regimens propelled the exploration for alternative antimicrobials with minimal toxicity and multi-target specificity. This study aimed to design and optimize antimicrobial peptides (AMPs) targeting crucial Mtb enzymes, namely, arabinosyltransferase C, DNA gyrase, 30S ribosomal protein S1, and enoyl-[acyl-carrier-protein] reductase. Curated set of 92 natural peptides, exhibiting high positive charge and reported efficacy against Mtb-H37Rv were screened and evaluated for pharmacokinetic properties. To enhance efficacy and overcome the intrinsic limitations of cationic AMPs, a mutant library was generated. Among them, NKLF2 and its mutants (M16C and M16I) demonstrated improved antibacterial efficacy (~ 5%) and favorable pharmacological profiles with no predicted toxicity. Molecular docking revealed enhanced binding affinities of the mutants across multiple targets. Notably, NKLF2_M16I exhibited enhancements of 9.71% and 7.63% in binding affinities against 4NNI and 5VRL respectively, while NKLF2_M16C achieved increments of 5.4% and 4.37% against 4G3N and 3PTY. Intermolecular interaction profiling identified hydrogen bonds, salt bridges, and hydrophobic interactions with the crucial active site residues of each target. Validations through coarse-grained, molecular, and essential dynamics simulations revealed minimal residue-level fluctuations, stable backbone profile, and minimized energy cluster basins ensuring compactness and stability of the protein-peptide docked complexes. These in silico findings open new avenues for further experimental validations and suggest that NKLF2 mutants in combination with conventional anti-TB drugs could pave the path towards the development of effective therapeutics in combating Mtb.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144267175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Anti-Inflammatory Effect and Gut Health of Levilactobacillus brevis Strains in Macrophage and Intestinal Cells.","authors":"Su-Jin Min, Eun-Soo Lee, Hyun-Joo Yoon, Na-Kyoung Lee, Hyun-Dong Paik","doi":"10.1007/s12602-025-10621-3","DOIUrl":"https://doi.org/10.1007/s12602-025-10621-3","url":null,"abstract":"<p><p>This study evaluated the potential of Levilactobacillus brevis strains (20080 and G1) to alleviate intestinal inflammation and support gut health. Chronic inflammatory conditions are often associated with gut microbiota dysbiosis, suggesting a potential role for probiotics in its management. The anti-inflammatory effects of L. brevis strains were assessed in cellular models of systemic and intestinal inflammation using RAW 264.7 macrophages and HT-29 epithelial cells. In lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages, L. brevis strains suppressed pro-inflammatory cytokines and nitric oxide (NO) production while enhancing anti-inflammatory responses. Furthermore, the expression levels of prostaglandin E<sub>2</sub> and leukotriene B4, which are key mediators of inflammatory diseases, particularly arthritis, were significantly reduced. Additionally, these strains effectively inhibited the activation of nuclear factor-κB, activator protein-1, and mitogen-activated protein kinase pathways, which are key regulators of inflammatory responses. In HT-29 cells, treatment with these strains led to a reduction in NO production upon sodium nitroprusside (SNP) stimulation. Under LPS stimulation, these strains suppressed the expression of pro-inflammatory cytokines and enhanced the expression of tight junction and mucin genes. In conclusion, L. brevis strains exhibited anti-inflammatory effects and gut-protective effects against intestinal inflammation, supporting their potential as therapeutic agents for inflammatory diseases.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144258823","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}