Carolina E Portero, Claire Smith, Yuxi Zhou, M Raquel Marchán-Rivadeneira, Shiyong Wu, Yong Han
{"title":"Construction of Synthetic Probiotic Bacteria for In Situ Delivery of Anti-SARS-CoV-2 Nanobodies.","authors":"Carolina E Portero, Claire Smith, Yuxi Zhou, M Raquel Marchán-Rivadeneira, Shiyong Wu, Yong Han","doi":"10.1007/s12602-025-10758-1","DOIUrl":"https://doi.org/10.1007/s12602-025-10758-1","url":null,"abstract":"<p><p>SARS-CoV-2 viral infection can be inhibited by blocking the interaction between the viral spike protein and the human receptor angiotensin-converting enzyme 2 (hACE2). The development of specific spike inhibitors using nanobodies, the antigen-binding region of llamas' antibodies, arose as a promising therapeutic method against SARS-CoV-2. However, one limitation of nanobodies is that they cannot be used directly in the human body due to their susceptibility to degradation. Bacteria-based delivery systems provide site-specific targeted action that can circumvent nanobody degradation. Here, we report the development of a genetically modified bacterium expressing anti-SARS-CoV-2 nanobodies that can inhibit the interaction between the hACE2 receptor and the receptor-binding domain (RBD) of the spike protein. Lactococcus lactis, a human symbiont probiotic bacterium, was selected to express nanobodies attached to their cell surface. Our data shows that FLAG-tagged anti-SARS-CoV-2 nanobodies were detected on the cell surface of recombinant L. lactis strains by flow cytometry and immunofluorescence without permeabilization. Furthermore, nanobodies are functional and can bind the RBD region from the spike protein in a dose-dependent manner. Inhibition of the hACE2-RBD interaction in cellular assays was quantified using a pseudotype lentivirus that mimics SARS-CoV-2 in an adaptation of the neutralization assay. Our results suggested that the recombinant bacteria can inhibit viral infectivity in more than 50% compared with a control without bacteria in a neutralization assay. These outcomes suggest that the engineered strain can be used in the future as a new therapeutic tool in COVID-19 prevention.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145034292","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}
Alice Njolke Mafe, Obiekezie Obumneme Smart, Great Iruoghene Edo, Patrick Othuke Akpoghelie, Tayser Sumer Gaaz, Emad Yousif, Endurance Fegor Isoje, Ufuoma Augustina Igbuku, Shams A Ismael, Arthur Efeoghene Athan Essaghah, Dina S Ahmed, Dilber Uzun Ozsahin, Huzaifa Umar
{"title":"Genesis, Health Benefits, and Future Perspectives of Probiotics: Exploring Endogenous and Exogenous Classes, Innovations, and Research Gaps.","authors":"Alice Njolke Mafe, Obiekezie Obumneme Smart, Great Iruoghene Edo, Patrick Othuke Akpoghelie, Tayser Sumer Gaaz, Emad Yousif, Endurance Fegor Isoje, Ufuoma Augustina Igbuku, Shams A Ismael, Arthur Efeoghene Athan Essaghah, Dina S Ahmed, Dilber Uzun Ozsahin, Huzaifa Umar","doi":"10.1007/s12602-025-10756-3","DOIUrl":"https://doi.org/10.1007/s12602-025-10756-3","url":null,"abstract":"<p><p>Probiotics are live beneficial microorganisms that confer health benefits to the host when administered in adequate amounts, have gained considerable scientific and commercial interest for their ability to support gut health, strengthen immunity, and reduce disease risk. This review traces the genesis of probiotic science from its origins in traditional fermented foods to contemporary clinical applications, offering a conceptual understanding of its evolution. A clear distinction is drawn between endogenous probiotics, naturally resident in the human microbiome, and exogenous probiotics, introduced via dietary supplements and functional foods. The broad spectrum of documented health benefits is examined, encompassing digestive, immune, metabolic, neurological, and dermatological outcomes. Recent innovations are highlighted, including bioengineered probiotic strains with targeted therapeutic functions, the integration of probiotics with prebiotics as synbiotics, and advanced delivery systems such as microencapsulation and nanotechnology. Special attention is given to regulatory frameworks, with global comparisons and a focused case study on Argentina, alongside a structured roadmap for translating research into market-ready products. The review also addresses inclusivity in probiotic use, emphasizing safety considerations across diverse populations, and underscores the strain-specific nature of probiotic effects. Current challenges such as commercialization gaps, regulatory inconsistencies, and underexplored applications in non-digestive health domains are critically discussed. The conclusion calls for interdisciplinary collaboration among microbiologists, nutritionists, clinicians, and technologists to accelerate innovation, ensure equitable access, and maximize the potential of probiotics in promoting health and preventing disease.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145030389","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":"Effects and Mechanisms of Lactiplantibacillus plantarum G83 on Enterotoxigenic Escherichia coli (ETEC)-Induced Intestinal Inflammation.","authors":"Chunjia Li, Miao Wang, Lixiao Duan, Jinge Xin, Xueqin Ni, Dong Zeng, Bangyuan Wu","doi":"10.1007/s12602-025-10712-1","DOIUrl":"https://doi.org/10.1007/s12602-025-10712-1","url":null,"abstract":"<p><p>Enterotoxigenic Escherichia coli (ETEC) is a prevalent intestinal pathogen that significantly impacts both human and animal health. G83, isolated from giant panda feces, has demonstrated notable probiotic properties. In this study, C57BL/6 J mice were randomly divided into Control, ETEC, and G83 groups. Experimental included monitoring body weight, assessing fecal occult blood, histopathological examination of ileal tissues, and quantification of antioxidant markers (SOD, T-AOC, MDA) in ileal tissues. Furthermore, real-time quantitative PCR was utilized to determine mRNA expression levels of inflammatory cytokines (TNF-α, IL-17, IL-10), tight junction proteins (Claudin, ZO-1, Occludin), mucin (Muc2), and lysozyme (Lyz-1). Transcriptomic bioinformatics analysis and 16S rRNA sequencing were integrated to characterize host gene expression profiles and gut microbial compositional dynamics, respectively. The results revealed that G83 alleviated ETEC-induced weight loss, reduced fecal occult blood, and mitigated ileal structural injuries. Additionally, G83 significantly enhanced intestinal antioxidant capacity by increasing T-AOC and SOD levels. Mechanistically, G83 downregulated pro-inflammatory cytokines TNF-α and IL-17 and the levels of Muc2 and Lyz1, while upregulating the expression of tight junction proteins ZO-1, Claudin, and Occludin. Transcriptomic analysis suggests that ETEC triggers inflammasome activation and initiates inflammatory responses by significantly upregulating Aim2. Conversely, G83 exerts protective effects by modulating the immune regulatory network-specifically, by significantly downregulating C3 expression to activate the complement system and participating in mucosal immune remodeling. Enrichment analysis reveals that G83 alleviates ETEC-induced intestinal inflammation primarily by inhibiting the NF-κB pathway and enhancing the intestinal IgA immune network. Additionally, 16S rRNA analysis indicates that G83 may improve ETEC-induced alterations in microbial community structure by increasing the abundance of beneficial bacteria (e.g., Lactobacillus), thereby further ameliorating impairment of intestinal microbial barrier function in mice. These findings provide a scientific basis for using G83 to ameliorate ETEC-mediated intestinal inflammation.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145024019","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":"Trp-containing Peptides with Therapeutic Potential for Cutibacterium acnes Infection.","authors":"Mengmiao Li, Zhuyun Liu, Jialu He, Jiahuan Jiang, Dejing Shang, Weibing Dong","doi":"10.1007/s12602-025-10749-2","DOIUrl":"https://doi.org/10.1007/s12602-025-10749-2","url":null,"abstract":"<p><p>Cutibacterium acnes (C. acnes, formerly classified as Propionibacterium acnes) is a Gram-positive bacterium that contributes to the development of acne vulgaris, resulting in inflammation and pustule formation on the skin. In this study, we developed and synthesized a series of antimicrobial peptides (AMPs) that are derived from the skin secretion of Rana chensinensis. Screening by MIC assay, Trp-containing peptides had potent anti-bacterial activity against C. acnes. The effect of Trp-containing peptides on the morphology was detected by scanning electron microscopy (SEM) and atomic force microscopy (AFM). The cell surface of C. acnes that was treated with Trp-containing peptides appeared rough and irregular. Further investigations into the peptides effects on cell membranes were conducted via zeta potential, Disc3(5) and Live/Dead assays. The results revealed that the AMPs bound to the cell surface, induced the disruption of the cell membranes and ultimately led to cell death. The leakage of cellular contents like nucleic acid, K<sup>+</sup> and Ca<sup>2+</sup> also confirmed the membrane-damaging effect of Trp-containing peptides. In addition, Trp-containing peptides suppressed biofilm formation and disrupted mature biofilms of C. acnes. Among the Trp-containing peptides, I4WL5W with lowest cytotoxic effect displayed dual anti-bacterial and anti-inflammatory effects in acne mice; it decreased the bacterial load and inhibited the levels of TNF-α, IL-6 and IL-1β in the infected skin. This study demonstrated that Trp-containing peptides might be promising candidates for treating acne.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145024111","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":"Natural and synthetic antimicrobial peptides against anaerobic bacteria: A review of activities and mechanisms of action.","authors":"Reza Ghasemi, Asieh Taji, Haniyeh Norouzi, Maryam Koleini, Ahmad Mosadegh, Hamid Heidari","doi":"10.1007/s12602-025-10730-z","DOIUrl":"https://doi.org/10.1007/s12602-025-10730-z","url":null,"abstract":"<p><p>Anaerobic bacteria cause a wide range of infections, varying from mild to severe, whether localized, implant-associated, or invasive, often leading to high morbidity and mortality. These infections are challenging to manage due to antimicrobial resistance against common antibiotics such as carbapenems and nitroimidazoles. The empirical use of antibiotics has contributed to the emergence of resistant organisms, making the identification and development of new antibiotics increasingly difficult. This highlights the need for adjuvant treatments. Antimicrobial peptides (AMPs) have garnered attention as promising agents in combating resistant bacteria. Researchers are interested in AMPs due to their broad-spectrum activity, multiple mechanisms of action, and reduced likelihood of inducing resistance. Moreover, AMPs can enhance the effectiveness of conventional antibiotics through synergistic effects. Researchers suggest that AMPs could play a valuable role in the development of new drugs targeting anaerobic pathogens. Exploring the properties and functions of these peptides represents a significant step toward alternative therapies. This paper reviews natural and synthetic AMPs derived from various sources that target different cellular components and functions of anaerobic bacteria, including Actinomyces, Fusobacterium, Bacteroides, Porphyromonas, Prevotella, Clostridium, Clostridioides, Lactobacillus, Peptostreptococcus, Propionibacterium, Bifidobacterium, Veillonella, and Tannerella. The described AMPs act on the cell membrane, peptidoglycan, lipopolysaccharide (LPS), biofilms, ion channels, toxins, DNA, transcription, and translation. The data indicate that these peptides demonstrate significant potential against anaerobes, presenting a promising strategy for combating antibiotic resistance.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145024027","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":"Comparative Genome Analysis and Characterization of Lacticaseibacillus Paracasei NKN344 Strain Isolated from Curd of Buffalo Milk Reared on Brackish Water Lagoons of the Eastern Indian Coast.","authors":"Amit Gaurav, Harpreet Singh, Mahesh Dige, Manishi Mukesh, Naveen Kumar Navani","doi":"10.1007/s12602-025-10754-5","DOIUrl":"https://doi.org/10.1007/s12602-025-10754-5","url":null,"abstract":"<p><p>Ethnic fermented foods represent a significant repository for discovering novel probiotic entities. These fermented foods, entrenched in indigenous practices, have conserved a distinct microbiota through generations. Exploration of these fermented foods could yield microbial consortia capable of transforming human health. However, comprehensive research into the probiotic attributes and quality analysis is necessary before its usage as biotherapeutics. In the current study, Chilika curd - an ethnic fermented curd originating from Odisha was explored to isolate novel probiotic strains. A detailed phenotypic and genomic characterization of a novel Lacticaseibacillus paracasei strain was conducted. Host-probiotic interactions were assessed using the Caenorhabditis elegans model. Lacticaseibacillus paracasei NKN344 exhibited robust survival under various physiochemical stresses, such as in vitro simulated gut environment and in vivo Caenorhabditis elegans intestinal model. Additionally, an in-depth bioinformatic analysis revealed the metabolic prowess of Lacticaseibacillus paracasei NKN344, including a few bacteriocin-encoding operons. Lastly, the production of active bacteriocin by Lacticaseibacillus paracasei NKN344 was validated, showing inhibitory activity against Bacillus cereus, a major food spoilage bacterium. Results of the current study proved that Lacticaseibacillus paracasei NKN344 isolated from Chilika curd has promising probiotic properties and seems favorable for its use in functional fermented foods.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145024051","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":"Synergistic Effects of Bacillus subtilis and Bacillus amyloliquefaciens on Growth, Immunity, Enzymatic Indices, Immune Gene Expression, and Survival of Labeo rohita Fry in a Biofloc Culture System.","authors":"Pushpa Choudhary, Debaprasad Rath, Shweta Priyadarshini Dash, Rakhi Kumari, Chinmayee Muduli, Rakesh Das","doi":"10.1007/s12602-025-10733-w","DOIUrl":"https://doi.org/10.1007/s12602-025-10733-w","url":null,"abstract":"<p><p>This study investigates the effects of probiotics Bacillus subtilis and Bacillus amyloliquefaciens on Labeo rohita fry within a biofloc culture system (BFC). The experimental design consisted of four treatment groups: control (BFC only), T1 (BFC + B. subtilis), T2 (BFC + B. amyloliquefaciens), and T3 (BFC + B. subtilis + B. amyloliquefaciens), each in triplicate. Molasses was added to maintain an optimal C:N ratio of 15:1. Over an 11-week period, the inclusion of probiotics resulted in significant improvements in weight gain, survival, digestive enzyme activities, and immune responses, with the T3 group showing the most pronounced effects. Probiotic supplementation significantly enhanced immune parameters, liver antioxidative activities, and expression of pro-inflammatory cytokines and antioxidant molecules, particularly in the T3 group. Additionally, there was a non-significant difference between concentration of glucose and cortisol levels in treatments and control; however, the control group showed highest glucose and cortisol among treatments. After the challenge study with Aeromonas hydrophila, the highest relative percentage survival (RPS) was observed in T3 (50%), followed by T2 (33.34%) and T1 (16.67%). These findings underscore the synergistic benefits of using a combination of probiotics in biofloc systems for enhancing the growth, immunity, and survival of L. rohita fry.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145006532","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}
Milena Rosa Lopes, Rosa Direito, Elen Landgraf Guiguer, Vitor Cavallari Strozze Catharin, Tereza Lais Menegucci Zutin, Claudio José Rubira, Virgínia Maria Cavallari Strozze Catharin, Kátia Portero Sloan, Lance Alan Sloan, José Luiz Yanaguizawa Junior, Lucas Fornari Laurindo, Sandra Maria Barbalho, Ricardo de Alvares Goulart
{"title":"Bridging the Gut Microbiota and the Brain, Kidney, and Cardiovascular Health: The Role of Probiotics.","authors":"Milena Rosa Lopes, Rosa Direito, Elen Landgraf Guiguer, Vitor Cavallari Strozze Catharin, Tereza Lais Menegucci Zutin, Claudio José Rubira, Virgínia Maria Cavallari Strozze Catharin, Kátia Portero Sloan, Lance Alan Sloan, José Luiz Yanaguizawa Junior, Lucas Fornari Laurindo, Sandra Maria Barbalho, Ricardo de Alvares Goulart","doi":"10.1007/s12602-025-10680-6","DOIUrl":"https://doi.org/10.1007/s12602-025-10680-6","url":null,"abstract":"<p><p>The symbiosis between intestinal bacteria and the human body's physiological processes can modulate health. The intestinal microbiota is linked to the development of neurotrophic factors; therefore, it is increasingly related to the modulation of nervous system pathologies. Moreover, microbiota can interfere with inflammation and oxidative stress, which are closely linked to cardiovascular risk factors and several other inflammatory conditions, such as kidney and neurodegenerative diseases. Probiotics are live microorganisms that help regulate and maintain healthy microbiota; thus, they can help prevent these diseases. Due to these reasons, this review aimed to evaluate the effects of probiotics on the gut, kidneys, brain, and heart homeostasis. Clinical trials showed several positive results with the treatment. In the brain, probiotics reduce depressive symptoms (decreases in HAMA, GAD-7, and BDI-II scales), improving patients' sleep quality and fatigue, enhancing cognitive subscales while slowing brain atrophy, and reducing IL-6 levels in the central areas, also modulating REM delta power to reduce high-frequency brain waves. Probiotics can also reduce cardiovascular risk factors, such as inflammation. Probiotics can also benefit the heart by decreasing TMAO, LDL-c, TG, CRP, MDA, TNF-α, IL-6, and urea levels, improving dyslipidemia and toxin profiles. Probiotics also increase HDL-c, ApoE, and insulin sensitivity, decreasing BMI, body fat, and the risk of developing chronic hyperglycemia while increasing lean mass. Besides, probiotic supplementation helped reduce toxic uremic toxins (serum urea) and sodium levels, bringing benefits to the kidneys, and improve energy/amino acid metabolism. Probiotics can also modulate and enhance kidney function due to decreased pro-inflammatory TGFβ-1 and TNF-α levels and RUNX2. Furthermore, enhanced gastrointestinal motility and diversity have been reported using specific bacteria. Although probiotics can bring several health benefits, there are still challenges regarding these supplements, such as dose, frequency, and pharmaceutical formula. Therefore, new studies are welcome to deepen the understanding of these microorganisms.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145006579","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}
João Xavier da Silva Neto, Laís Oliveira Leite, Ana Paula Apolinário da Silva, Maria Izabel Florindo Guedes, Daniele de Oliveira Bezerra de Sousa
{"title":"Advancing the Understanding of the Mo-CBP<sub>2</sub> Protein: Unraveling Its Antifungal and Antibiofilm Properties Against Candida Species.","authors":"João Xavier da Silva Neto, Laís Oliveira Leite, Ana Paula Apolinário da Silva, Maria Izabel Florindo Guedes, Daniele de Oliveira Bezerra de Sousa","doi":"10.1007/s12602-025-10744-7","DOIUrl":"https://doi.org/10.1007/s12602-025-10744-7","url":null,"abstract":"<p><p>Candida spp. are opportunistic fungi capable of forming biofilms, a key factor contributing to their resistance to conventional antifungals. This highlights the need for novel compounds with distinct mechanisms of action to combat fungal infections. This study aimed to evaluate the antifungal activity of Mo-CBP<sub>2</sub>, a chitin-binding lectin from Moringa oleifera seeds, against Candida albicans and Candida tropicalis, with a specific focus on its effects on planktonic cells and biofilms, and to investigate its mechanism of action. Mo-CBP<sub>2</sub> was purified via affinity and ion-exchange chromatography. Antifungal activity was assessed using microdilution, CFU counts, and MTT assays for planktonic cells, and crystal violet staining for biomass quantification in both early-stage and mature biofilms. Mechanistic studies included aggregation assays, sorbitol protection tests, and ergosterol quantification. Against planktonic cells, Mo-CBP<sub>2</sub> exhibited potent antifungal activity, with MIC₅₀ values ranging from 20-45 μM, reducing metabolic activity and CFUs by up to 90%. Additionally, it promoted aggregation of fungal cells, indicating interaction with cell wall components, and showed a fungistatic profile. Regarding biofilms, Mo-CBP<sub>2</sub> significantly inhibited biomass formation in both initial adhesion and mature stages (CIB₅₀ = 20 μM), with greater efficacy than nystatin. Mechanistic assays revealed that its antibiofilm effect is independent of ergosterol biosynthesis or cell wall synthesis pathways, as no alterations were observed in the presence of sorbitol or in sterol content. Mo-CBP<sub>2</sub> presents strong antifungal and antibiofilm activities against Candida spp., acting through a novel, non-conventional mechanism.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145001364","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}
Cuijiao Feng, Yue Zhao, Qiuwen He, Jie Yu, Zhihong Sun
{"title":"Bifidobacterium breve B2798 and Its Heat-Killed Cells Alleviate Inflammation in Rats with DSS Model by Modulating Gut Microbiota.","authors":"Cuijiao Feng, Yue Zhao, Qiuwen He, Jie Yu, Zhihong Sun","doi":"10.1007/s12602-025-10648-6","DOIUrl":"https://doi.org/10.1007/s12602-025-10648-6","url":null,"abstract":"<p><p>While probiotics are widely recognized for their adjunctive benefits in ulcerative colitis treatment, the therapeutic potential of heat-killed cells remains underexplored. This study directly compared the efficacy of Bifidobacterium breve B2798 probiotics (LB group) and their heat-killed counterparts (DB group) in alleviating dextran sulfate sodium (DSS)-induced colitis in rats. Over a 21-day intervention, both treatments significantly mitigated colitis symptoms, including weight loss, colon damage, and splenomegaly, with heat-killed cells demonstrating superior histological improvement over live probiotics. Serum analysis revealed that both interventions normalized DSS-induced cytokine dysregulation, reducing pro-inflammatory markers and elevating anti-inflammatory. Although α-diversity remained stable, β-diversity analysis indicated distinct gut microbiota restructuring. Heat-killed cells uniquely enriched butyrate-producing Alistipes spp. and Parabacteroides distasonis, while probiotics upregulated Mucispirillum schaedleri and Odoribacter splanchnicus. Metabolomic profiling identified shared elevation of anti-inflammatory metabolites (linoleic acid, isorhamnetin) in both groups, yet heat-killed cells exhibited stronger modulation of metabolic pathways, including TCA cycle activation and pantothenate biosynthesis suppression. Correlation networks highlighted species-specific microbiota-metabolite-cytokine interactions, with Mucispirillum schaedleri and Barnesiella intestinihominis negatively associated with inflammatory markers (MPO, TNF-α). These findings demonstrate that while both live and heat-killed B. breve B2798 alleviate colitis, heat-killed cells exert enhanced regulatory effects on gut microbiota composition, metabolic pathways, and inflammatory responses, offering a safer alternative for inflammatory bowel disease management. Further mechanistic studies are warranted to validate these preclinical insights.</p>","PeriodicalId":20506,"journal":{"name":"Probiotics and Antimicrobial Proteins","volume":" ","pages":""},"PeriodicalIF":4.4,"publicationDate":"2025-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145001370","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}