1
|
Steinert RE, Rehman A, Sadabad MS, Milanese A, Wittwer-Schegg J, Burton JP, Spooren A. Microbial micronutrient sharing, gut redox balance and keystone taxa as a basis for a new perspective to solutions targeting health from the gut. Gut Microbes 2025; 17:2477816. [PMID: 40090884 PMCID: PMC11913388 DOI: 10.1080/19490976.2025.2477816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025] Open
Abstract
In health, the gut microbiome functions as a stable ecosystem maintaining overall balance and ensuring its own survival against environmental stressors through complex microbial interaction. This balance and protection from stressors is maintained through interactions both within the bacterial ecosystem as well as with its host. As a consequence, the gut microbiome plays a critical role in various physiological processes including maintaining the structure and function of the gut barrier, educating the gut immune system, and modulating the gut motor, digestive/absorptive, as well as neuroendocrine system all of which are crucial for human health and disease pathogenesis. Pre- and probiotics, widely available and clinically established, offer various health benefits primarily by beneficially modulating the gut microbiome. However, their clinical outcomes can vary significantly due to differences in host physiology, diets, individual microbiome compositions, and other environmental factors. This perspective paper highlights emerging scientific insights into the importance of microbial micronutrient sharing, gut redox balance, keystone species, and the gut barrier in maintaining a diverse and functional microbial ecosystem, and their relevance to human health. We propose a novel approach that targets microbial ecosystems and keystone taxa performance by supplying microbial micronutrients in the form of colon-delivered vitamins, and precision prebiotics [e.g. human milk oligosaccharides (HMOs) or synthetic glycans] as components of precisely tailored ingredient combinations to optimize human health. Such a strategy may effectively support and stabilize microbial ecosystems, providing a more robust and consistent approach across various individuals and environmental conditions, thus, overcoming the limitations of current single biotic solutions.
Collapse
Affiliation(s)
- Robert E. Steinert
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
- Department of Surgery and Transplantation, University Hospital Zurich (USZ) and University of Zurich (UZH), Zürich, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| | | | - Alessio Milanese
- Data Science, Science & Research, Dsm-Firmenich, Delft, Netherlands
| | | | - Jeremy P. Burton
- Department of Microbiology and Immunology, The University of Western Ontario, London, Canada
| | - Anneleen Spooren
- Health, Nutrition & Care (HNC), Dsm-Firmenich, Kaiseraugst, Switzerland
| |
Collapse
|
2
|
Schoonakker MP, van Peet PG, van den Burg EL, Numans ME, Ducarmon QR, Pijl H, Wiese M. Impact of dietary carbohydrate, fat or protein restriction on the human gut microbiome: a systematic review. Nutr Res Rev 2025; 38:238-255. [PMID: 38602133 DOI: 10.1017/s0954422424000131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Restriction of dietary carbohydrates, fat and/or protein is often used to reduce body weight and/or treat (metabolic) diseases. Since diet is a key modulator of the human gut microbiome, which plays an important role in health and disease, this review aims to provide an overview of current knowledge of the effects of macronutrient-restricted diets on gut microbial composition and metabolites. A structured search strategy was performed in several databases. After screening for inclusion and exclusion criteria, thirty-six articles could be included. Data are included in the results only when supported by at least three independent studies to enhance the reliability of our conclusions. Low-carbohydrate (<30 energy%) diets tended to induce a decrease in the relative abundance of several health-promoting bacteria, including Bifidobacterium, as well as a reduction in short-chain fatty acid (SCFA) levels in faeces. In contrast, low-fat diets (<30 energy%) increased alpha diversity, faecal SCFA levels and abundance of some beneficial bacteria, including Faecalibacterium prausnitzii. There were insufficient data to draw conclusions concerning the effects of low-protein (<10 energy%) diets on gut microbiota. Although the data of included studies unveil possible benefits of low-fat and potential drawbacks of low-carbohydrate diets for human gut microbiota, the diversity in study designs made it difficult to draw firm conclusions. Using a more uniform methodology in design, sample processing and sharing raw sequence data could foster our understanding of the effects of macronutrient restriction on gut microbiota composition and metabolic dynamics relevant to health. This systematic review was registered at https://www.crd.york.ac.uk/prospero as CRD42020156929.
Collapse
Affiliation(s)
- Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Quinten R Ducarmon
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
| | - Maria Wiese
- Department of Medical Microbiology, Leiden University Medical Centre (LUMC), Leiden, The Netherlands
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
3
|
Das S, Konwar BK. The vital role of Lactobacillus sp. in vaginal health: Implications for enhanced prophylactic research. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10574-7. [PMID: 40402415 DOI: 10.1007/s12602-025-10574-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Genital opening or vaginal canal (VC) in women has different embryological origin than the entire female reproductive system. The microenvironment of the VC post puberty is everchanging due to the hormonal fluctuations in a woman's body. However, the vaginal canal maintains a defined microbiota with higher population of inherent lactic acid bacteria (LAB) species under healthy conditions. The preservation of the beneficial flora in the genital area is dependent on genetic, social, and habitual factors. The understanding and practice of these factors prevents the commencement of various vaginal infections/vaginitis. Common vaginal infections have known antibiotic treatments; however, treatments of few infections are still unavailable. The study of sexual health is scarce in India due to social and economic factors, with less number of studies from various parts of the country. Thus, there is a necessity of new-age prophylactic solutions in such developing countries. This review highlights the origin of the female reproductive system and describes how the hormonal change initiates unique microenvironment development. Later, we have elaborately discussed the precautionary contribution of lactic acid bacteria and their unexplored commercial utilization, which in turn would help in various ways to improve the standards of reproductive hygiene products.
Collapse
Affiliation(s)
- Shreaya Das
- Dept. of Molecular Biology and Biotechnology, Tezpur University (Central), Napaam, 784028, Assam, India.
| | - Bolin Kumar Konwar
- Dept. of Molecular Biology and Biotechnology, Tezpur University (Central), Napaam, 784028, Assam, India
| |
Collapse
|
4
|
Ma T, Li Y, Yang N, Wang H, Shi X, Liu Y, Jin H, Kwok LY, Sun Z, Zhang H. Efficacy of a postbiotic and its components in promoting colonic transit and alleviating chronic constipation in humans and mice. Cell Rep Med 2025; 6:102093. [PMID: 40286792 DOI: 10.1016/j.xcrm.2025.102093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/06/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
This study evaluates the efficacy of the postbiotic Probio-Eco in alleviating constipation in humans and mice. A randomized, double-blind, placebo-controlled crossover trial involving 110 adults with chronic constipation (Rome IV criteria) demonstrates that a 3-week Probio-Eco intervention significantly improves constipation symptoms, stool straining, and worry scores. Gut microbiota and metabolomic analyses reveal modulations in specific gut microbiota, succinate, tryptophan derivatives, deoxycholate, propionate, butyrate, and cortisol, correlating with symptom relief. A loperamide-induced mouse model confirms that Probio-Eco and its bioactive components (succinate, 3-indoleacrylic acid, and 5-hydroxytryptophan) alleviate constipation by stimulating mucin-2 secretion, regulating intestinal transport hormones, and promoting anti-inflammatory responses. Multi-omics integration identifies key pathways, including succinate-short-chain fatty acid, tryptophan-5-hydroxytryptophan-serotonin, and tryptophan-3-indoleacrylic acid, driving intestinal homeostasis and motility. These findings highlight the comprehensive efficacy of Probio-Eco and provide robust evidence for its clinical application in constipation management. This study was registered at Chinese Clinical Trial Registry (ChiCTR2100054376).
Collapse
Affiliation(s)
- Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yalin Li
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Ni Yang
- State Key Laboratory of Research and Development of Classical Prescription and Modern Chinese Medicine, 1899 Meiling Road, Nanchang 330103, China
| | - Huan Wang
- Inner Mongolia People's Hospital, Hohhot, China
| | - Xuan Shi
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanfang Liu
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China.
| |
Collapse
|
5
|
Wu J, Wang Z, Hu J, Liu J, Han X, Chen H, Zhu S, Deng J. Bacillus subtilis-Derived Postbiotics as a Multifunctional Bio-Catalyst for Enhancing Lactic Acid Bacteria Viability and Yogurt Quality. Foods 2025; 14:1806. [PMID: 40428584 DOI: 10.3390/foods14101806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/12/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
This study demonstrates that Bacillus subtilis GDAAS-A32-derived postbiotics (BSP) enhance yogurt production by optimizing lactic acid bacteria (LAB) viability and functionality. BSP enhanced the growth kinetics and biomass accumulation of Streptococcus thermophilus and Lactobacillus bulgaricus in both an anaerobic and aerobic pure system. The addition of BSP significantly increased the viable cell counts of S. thermophilus and L. bulgaricus, milk-clotting activity, sensory properties, and extracellular polysaccharide content and improved the rheological properties. Moreover, BSP elevated viable counts of S. thermophilus and L. bulgaricus to 6.18 × 108 CFU/g and 1.03 × 108 CFU/g, respectively, by day 7-representing 11.3-fold and 9.3-fold increases versus controls at 20% supplementation. Metabolomic signatures confirmed peptidoglycan reinforcement and flavor enhancement. Mechanistically, BSP supplementation might reduce urate and H2O2 toxicity through NH3-mediated proton neutralization and oxygen scavenging, while establishing a pyrimidine salvage network and redirecting one-carbon metabolism, resulting in enhanced stress tolerance and significant improvements in bacterial viability.
Collapse
Affiliation(s)
- Jing Wu
- Xinjiang Key Laboratory of Biological Resources and Ecology of Pamirs Plateau, College of Life and Geographic Sciences, Kashi University, Kashi 844008, China
| | - Zhilin Wang
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Jingyi Hu
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jing Liu
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Xueying Han
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Hongping Chen
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Siming Zhu
- Xinjiang Key Laboratory of Biological Resources and Ecology of Pamirs Plateau, College of Life and Geographic Sciences, Kashi University, Kashi 844008, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Junjin Deng
- Agro-Biological Gene Research Center, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- Guangdong Laboratory for Lingnan Modern Agriculture Heyuan Sub-Center, Heyuan 517500, China
| |
Collapse
|
6
|
Zhang X, Wang Y, E Q, Naveed M, Wang X, Liu Y, Li M. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication. J Nanobiotechnology 2025; 23:349. [PMID: 40380331 PMCID: PMC12082936 DOI: 10.1186/s12951-025-03435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium spp. have been shown to be critical for maintaining host homeostasis. In recent years, key compounds of postbiotics derived from probiotic metabolism and cellular secretion have been identified for their role in maintaining organ immunity and regulating intestinal inflammation. In particular, probiotic-derived extracellular vesicles (PEVs) can act as postbiotics, maintaining almost the same functional activity as probiotics. They also have strong biocompatibility and loading capacity to carry exogenous or parental active molecules to reach distal organs to play their roles. This provides a new direction for understanding the intrinsic microbiota-host communication mechanism. However, most current studies on PEVs are limited to their functional effects/benefits, and their specific physicochemical properties, composition, intrinsic mechanisms for maintaining host homeostasis, and possible threats remain to be explored. Here, we review and summarize the unique physicochemical properties of PEVs and their bioactivities and mechanisms in mediating microbiota-host communication, and elucidate the limitations of the current research on PEVs and their potential application as postbiotics.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ye Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiyu E
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Naveed
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
Morgado-Cáceres P, Huerta H, Bergman C, Figueroa R, Farias P, Quiroz G, Woehlbier U, Mella K, Díaz-Rivera O, Linsambarth S, Calderón-Romero P, Court FA, Sepulveda D, Sauma D, Luz-Crawford P, Vargas AA, Gonzalez-Seguel C, Cárdenas JC, Lovy A. Postbiotic Parabacteroides Distasonis Supplementation Enhances Intestinal and Skeletal Muscle Function in Aged Mice. Aging Dis 2025:AD.2025.0188. [PMID: 40423631 DOI: 10.14336/ad.2025.0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Parabacteroides distasonis (Pd), a core member of the human gut microbiota, is enriched in centenarians, suggesting a potential role in promoting organismal resilience. While Pd supplementation has been shown to alleviate cancer and inflammatory diseases, its ability to mitigate the decline associated with aging remains unexplored. Here, we demonstrate that postbiotic Pd supplementation induces multiple beneficial effects in 18- and 26-month-old mice following three months of treatment. Pd-treated mice exhibit lower blood glucose levels and increased ketone body production. In the gut, Pd reduces colon shortening observed in aged control mice and decreases the inflammatory mediator NFκB, in the colonic mucosa. Microbiome analysis further reveals enhanced gut microbiota diversity in Pd-supplemented mice. Additionally, FITC-dextran permeability assays indicate improved intestinal barrier function. Cell culture experiments in HCT116 colon cell line show that Pd reduces oxygen consumption and promotes mitochondrial networking, accompanied by upregulation of PGC1α and CHOP, suggesting a mitohormetic response. Beyond metabolic and gut-related benefits, Pd supplementation enhances skeletal muscle strength in both 18- and 26-month-old mice. Proteomic analysis of gastrocnemius muscle reveals that Pd increases the expression of mitochondrial proteins associated with mitochondrial fitness and survival. Notably, Pd-supplemented mice challenged with a high-fat diet gain weight at a slower rate, while maintaining better skeletal muscle coordination and strength. In summary, our findings suggest that postbiotic Pd supplementation enhances metabolic health, reduces inflammation, improves mitochondrial function, and preserves muscle strength in aged mice. These results position Pd as a promising therapeutic tool for promoting healthy aging and combating aging-related diseases.
Collapse
Affiliation(s)
- Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Hernán Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Cristian Bergman
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Reinaldo Figueroa
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Paula Farias
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Gabriel Quiroz
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Ute Woehlbier
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Karen Mella
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Osmán Díaz-Rivera
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Sergio Linsambarth
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | | | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Denisse Sepulveda
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- BNI, Biomedical Neuroscience Institute, Santiago, Chile
| | - Daniela Sauma
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Patricia Luz-Crawford
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Anibal A Vargas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Catalina Gonzalez-Seguel
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - J César Cárdenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, USA
| | - Alenka Lovy
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Center for Vision Research and the Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University Syracuse, NY 13210, USA
| |
Collapse
|
8
|
Gao YQ, Tan YJ, Fang JY. Roles of the gut microbiota in immune-related adverse events: mechanisms and therapeutic intervention. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01026-w. [PMID: 40369317 DOI: 10.1038/s41571-025-01026-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2025] [Indexed: 05/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) constitute a major breakthrough in the field of cancer therapy; their use has resulted in improved outcomes across various tumour types. However, ICIs can cause a diverse range of immune-related adverse events (irAEs) that present a considerable challenge to the efficacy and safety of these treatments. The gut microbiota has been demonstrated to have a crucial role in modulating the tumour immune microenvironment and thus influences the effectiveness of ICIs. Accumulating evidence indicates that alterations in the composition and function of the gut microbiota are also associated with an increased risk of irAEs, particularly ICI-induced colitis. Indeed, these changes in the gut microbiota can contribute to the pathogenesis of irAEs. In this Review, we first summarize the current clinical challenges posed by irAEs. We then focus on reported correlations between alterations in the gut microbiota and irAEs, especially ICI-induced colitis, and postulate mechanisms by which these microbial changes influence the occurrence of irAEs. Finally, we highlight the potential value of gut microbial changes as biomarkers for predicting irAEs and discuss gut microbial interventions that might serve as new strategies for the management of irAEs, including faecal microbiota transplantation, probiotic, prebiotic and/or postbiotic supplements, and dietary modulations.
Collapse
Affiliation(s)
- Ya-Qi Gao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Jie Tan
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Wang Q, Yang Y, Xiao Y, Liu Z, Li M, Yu Y, Wang N, Li J, Li X, Xu C, Liu D, Wu C. Bifidobacterium animalis subsp. lactis BL-11 promotes height growth in 3- to 7-year-Old children with below-average height: A self-controlled clinical trial. Clin Nutr 2025; 50:57-65. [PMID: 40367596 DOI: 10.1016/j.clnu.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Short stature of children has captured extensive attention since it reflects the health status of children and affects physical and psychological health throughout children's life. Compelling evidences have revealed that gut microbiota plays a critical role in growth regulation through nutrient metabolism, inflammation modulation, and endocrine signaling. Bifidobacterium animalis subsp. lactis BL-11 (BL-11) is a probiotic strain with potential benefits for gut health and metabolic function, but its effect on growth promotion in children with mild growth delay has not been fully explored. OBJECTIVES This study aimed to evaluate the effect of BL-11 supplementation on height velocity (HV) and annual growth rate (AGR) in 3- to 7-year-old children with below-average height using a self-controlled clinical trial design over a 3-month intervention period. METHODS A total of 124 children aged 3-7 years with height in the 3rd to 25th percentiles (P3-P25) range and an AGR of <5 cm/year were enrolled. Each child received a daily dose of BL-11 for 3 months. Baseline height was measured before the intervention, and post-intervention height was recorded at the end of the 3-month period. The primary outcome was the change in HV and AGR and secondary outcomes included changes in height percentile. RESULTS After 3 months of BL-11 supplementation, the children showed a significant increase in HV, with a mean improvement of 2.23 cm/3-month (8.92 cm/year) compared to the pre-intervention rate of <5 cm/year (P value = 0.004). The height percentiles were also improved and about 55.88 % (19/34) children with height in P3-P10 shifted to P10-P25. No serious adverse events were reported. CONCLUSIONS Supplementation with Bifidobacterium animalis subsp. lactis BL-11 for 3 months significantly improved height velocity in 3- to 7-year-old children with below-average height. The findings suggest that BL-11 may be a safe and effective probiotic intervention for promoting growth in children with mild growth delay. Further long-term studies are warranted to confirm the durability and consistency of these effects.
Collapse
Affiliation(s)
- Qian Wang
- The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yin Xiao
- Anhui Medical University, Hehui, Anhui, China
| | - Ziyi Liu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mingxi Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Yu
- The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nannan Wang
- The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Junyan Li
- West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohui Li
- New Century Women's and Children's Hospital, Beijing, China
| | - Chuanhui Xu
- Minerva Hospital for Women and Children, Chengdu, Sichuan, China
| | - Deyun Liu
- The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Chinese Medicine Modernization, China.
| |
Collapse
|
10
|
Girija S, Kolluri G, Tyagi JS, Kurva SK, Sahoo PR, Srivastava V, Singh T. Postbiotics: The Dynamic Biomolecules in Poultry Nutrition, Health, and Production. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10548-9. [PMID: 40358876 DOI: 10.1007/s12602-025-10548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
The interest in employing cell components as well as metabolites obtained from varied probiotic strains is increasing because of the growing concern regarding safety issues related to live microbial cells. Numerous benefits over conventional probiotics will facilitate the implementation of postbiotics as feed supplements in poultry nutrition. The beneficial characteristics, such as immunomodulation and antibacterial action, point to the possibility of postbiotics improving host health by altering the physiology of the host and ameliorating the disease condition in poultry. As we proceed forward with the ultimate aim of lowering antibiotic use in poultry, maximising performance and maintaining poultry production will be reliant on the best mixes of diverse alternatives as postbiotics, together with appropriate farm management practices. Trials with emphasis on validating the health claims made by these bioactive compounds are highly necessary. Furthermore, the employment of postbiotics under challenging conditions in poultry needs to be explored. This review focuses on highlighting the aspects about postbiotics and its various biological effects on meat and egg production in the poultry industry.
Collapse
Affiliation(s)
- Simi Girija
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Gautham Kolluri
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India.
| | - Jagbir Singh Tyagi
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Shiva Kumar Kurva
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Pravas Ranjan Sahoo
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Vivek Srivastava
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Twinkle Singh
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| |
Collapse
|
11
|
Schiavone N, Isoldi G, Calcagno S, Rovida E, Antiga E, De Almeida CV, Lulli M. Exploring the Gut Microbiota-Retina Axis: Implications for Health and Disease. Microorganisms 2025; 13:1101. [PMID: 40431274 DOI: 10.3390/microorganisms13051101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
The gut microbiota represents a rich and adaptive microbial network inhabiting the gastrointestinal tract, performing key functions in nutrient processing, immune response modulation, intestinal wall protection, and microbial defense. Its composition remains highly personalized and responsive to external influences, including lifestyle patterns, physical activity, body composition, and nutritional intake. The interactions of the gut microbiota with bodily systems are conventionally interpreted as broad systemic impacts on organ balance. Yet, emerging research-exemplified by the gut microbiota-brain axis-suggests the potential existence of more targeted and direct communication mechanisms. Dysbiosis, characterized by microbial ecosystem disturbance, generates multiple metabolic compounds capable of entering systemic circulation and reaching distant tissues, notably including ocular structures. This microbial imbalance has been associated with both systemic and localized conditions linked to eye disorders. Accumulating scientific evidence now supports the concept of a gut-retina axis, underscoring the significant role of microbiota disruption in generating various retinal pathologies. This review comprehensively investigates gut microbiota composition, functional dynamics, and dysbiosis-induced alterations, with specific focus on retinal interactions in age-related macular degeneration, diabetic retinopathy, glaucoma, and retinal artery occlusion. Moreover, the review explores microbiota-targeted therapeutic strategies, including precision nutritional interventions and microbial transplantation, as potential modulators of retinal disease progression.
Collapse
Affiliation(s)
- Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
| | - Giulia Isoldi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
| | - Sara Calcagno
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
| | - Emiliano Antiga
- Department of Health Sciences, Section of Dermatology, University of Florence, 50139 Florence, Italy
| | - Carolina Vieira De Almeida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
- Press Start SRL Società Benefit, 50134 Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50134 Florence, Italy
| |
Collapse
|
12
|
Zhao P, Li Y, Yang Y, Xiao Q, Zhang Z, Hong X, Ni H, Xia Z, Zhan K, Yang S, Zhang Y. Probiotic efficacy and mechanism of a pigeon derived Ligilactobacillus salivarius strain in promoting growth and intestinal development of pigeons. Front Microbiol 2025; 16:1584380. [PMID: 40415945 PMCID: PMC12098536 DOI: 10.3389/fmicb.2025.1584380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Background With the gradual rise of antibiotic-free farming practices, the exploration of novel, green, and low-pollution alternatives to antibiotics has become one of the key research focus in the field of agricultural science. In the development of antibiotic alternatives, probiotics, particularly host-associated probiotics, have been found to play a significant role in enhancing the production performance of livestock and poultry. However, research on and application of probiotics specifically for meat pigeons remain relatively underdeveloped. Objective To assess and investigate the probiotic efficacy and mechanisms during homologous lactic acid bacteria (LAB) transplant to host-pigeons, LAB strains with good probiotic properties were isolated from the intestinal contents of 28-day-old Mimas pigeons. And then measured the production indexes, intestinal flora, and intestinal transcriptomics of the hosts after instillation of LAB strains. Methods A total of 360 at 1-day-old pigeons were randomly divided into four groups and gavaged 0.4 mL Ligilactobacillus salivarius S10 with concentration of 0, 108, 109, and 1010 CFU/mL, designated as the control group (CG), the low concentration group (LG), the medium concentration group (MG), and the high concentration group (HG), respectively. Results The findings revealed that an optimal concentration of 109 CFU/mL L. salivarius S10, a dominant strain isolated and screened, enhanced the growth performance and intestinal development of young pigeons. 16S rRNA gene sequencing analysis demonstrated a significant increase in the abundance of Lactobacillus, Pantoea_A and Enterococcus_H and a significant reduction in the abundance of Clostridium_T in the pigeon ileum (p < 0.05) under selected concentration treatment. Transcriptomic profiling of the ileum revealed 1828 differentially expressed genes (DEGs) between CG and MG. Notably, DEGs involved in the MAPK signaling pathway, such as RAF1, PDGFRB, and ELK4, were significantly correlated with differential ileal bacteria, suggesting that modulation of intestinal flora can influence the expression of genes related to cell proliferation and differentiation in the ileum, which is potentially important in promoting the growth and development of pigeons. Conclusion Ligilactobacillus salivarius S10 possesses the potential to be used as a probiotic for pigeons, which can influence the expression of gut development-related DEGs by regulating the intestinal flora, and further improve the growth performance of pigeons. This research provides a scientific foundation for developing pigeon-specific probiotics and promotes healthy farming practices for meat pigeons. Furthermore, it opens new avenues for improving the economic efficiency of pigeon farming.
Collapse
Affiliation(s)
- Puze Zhao
- College of Animal Science, Jilin University, Changchun, China
| | - Yumei Li
- College of Animal Science, Jilin University, Changchun, China
| | - Yuwei Yang
- College of Animal Science, Jilin University, Changchun, China
| | - Qingxing Xiao
- College of Animal Science, Jilin University, Changchun, China
| | - Ziyi Zhang
- College of Animal Science, Jilin University, Changchun, China
| | - Xiaoqing Hong
- College of Animal Science, Jilin University, Changchun, China
| | - Hongyu Ni
- College of Animal Science, Jilin University, Changchun, China
| | - Zhuxuan Xia
- College of Animal Science, Jilin University, Changchun, China
| | - Kun Zhan
- College of Animal Science, Jilin University, Changchun, China
| | - Sibao Yang
- Department of Cardiovascular Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yonghong Zhang
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
13
|
Zhang J, Zhang R, Wang J, Abbas Z, Tong Y, Fang Y, Zhou Y, Zhang H, Li Z, Si D, Wei X. Efficient Production Strategy of a Novel Postbiotic Produced by Bacillus subtilis and Its Antioxidant and Anti-Inflammatory Effects. Molecules 2025; 30:2089. [PMID: 40430262 DOI: 10.3390/molecules30102089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Microbially synthesized postbiotics have unique properties and advantages; however, systematic studies on the efficient production and biological functions of postbiotics from Bacillus subtilis are limited, which greatly restricts their applications. In this study, we obtained a novel crude exopolysaccharide (EPS) postbiotic from Bacillus subtilis H4. We systematically optimized the EPS production strategy using single-factor analysis, Plackett-Burman design, the path of steepest ascent method, and response surface methodology. The optimized EPS yield was significantly improved, with a maximum yield of 15.01 g/L under the addition of 4.12% soy peptone, 8.99% sucrose, and 0.06% MnSO4. We found that EPS is a neutral, heterogeneous polysaccharide with a pyranose ring, with a molecular weight of 44,304.913 kDa and a melting point of 218 °C. It consists of glucose, galactose, arabinose, glucosamine, and mannose at a molar ratio of 58.85:19.81:14.75:10.89:6.58. EPS exhibits strong antioxidant capacities, scavenging ABTS and DPPH radicals with IC50 values of 1 and 6 mg/mL, respectively. Moreover, it shows notable anti-inflammatory properties, dramatically inhibiting the lipopolysaccharide (LPS)-induced elevation of nitric oxide (NO) levels and over-activation of the TLR4-NF-κB signaling pathway. These findings highlight the potential of EPS as a multifunctional bioactive compound, offering great promise for its application in the food, clinical, and feed industries.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Rijun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junyong Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zaheer Abbas
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yucui Tong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yong Fang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yichen Zhou
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Haosen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhenzhen Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dayong Si
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xubiao Wei
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
14
|
D'Urso F, Paladini F, Miraglia A, D'Amuri A, Chieppa M, Pollini M, Broccolo F. Translating Patent Innovation into Clinical Practice: Two Decades of Therapeutic Advancements in Dysbiosis Management. Microorganisms 2025; 13:1064. [PMID: 40431238 DOI: 10.3390/microorganisms13051064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Dysbiosis, characterized by a microbial imbalance, particularly within the gut microbiota, has emerged as a significant health concern linked to various diseases. This study analyzed 8097 patent documents from The Lens database (2005-2024) to examine global innovation trends in dysbiosis management. The patent filings showed exponential growth, peaking at 1222 documents in 2022, with the United States leading in publications (4361 documents). The analysis revealed three primary innovation clusters: bacterial-based therapeutics (44.8% of patents), specific therapeutic applications (27.6%), and diagnostic methods (15.9%). The disease associations predominantly included inflammatory conditions, infections, and cancer. The patent classifications highlighted a significant focus on probiotic development and microbiota modulation. The surge in patent activity since 2014 correlates with advances in DNA sequencing technology and the growing recognition of dysbiosis's role in human health. This analysis provides valuable insights into the evolving landscape of microbiome therapeutics and future directions for dysbiosis management.
Collapse
Affiliation(s)
- Fabiana D'Urso
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Federica Paladini
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Alessandro Miraglia
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Alessandro D'Amuri
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Marcello Chieppa
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Mauro Pollini
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| | - Francesco Broccolo
- Department of Experimental Medicine (DiMeS), University of Salento, 73100 Lecce, Italy
| |
Collapse
|
15
|
Caffrey EB, Perelman D, Ward CP, Sonnenburg ED, Gardner CD, Sonnenburg JL. Unpacking Food Fermentation: Clinically Relevant Tools for Fermented Food Identification and Consumption. Adv Nutr 2025; 16:100412. [PMID: 40120687 PMCID: PMC12056253 DOI: 10.1016/j.advnut.2025.100412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Fermented foods have been consumed for millennia, valued for their extended shelf life, distinctive sensory properties, and potential health benefits. Emerging research suggests that fermented food consumption may contribute to gut microbiome diversity, immune modulation, and metabolic regulation; however, mechanistic insights and clinical validation remain limited. This review synthesizes current scientific evidence on the microbial and metabolite composition of fermented foods, their proposed health effects, and safety considerations for vulnerable populations. Additionally, we highlight the need for standardized definitions, serving sizes, and regulatory frameworks to enhance consumer transparency and research reproducibility. By providing a structured overview of existing data and knowledge gaps, this review establishes a foundation for integrating fermented foods into dietary recommendations and guiding future research directions.
Collapse
Affiliation(s)
- Elisa B Caffrey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Dalia Perelman
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, California, United States
| | - Catherine P Ward
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, California, United States
| | - Erica D Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Christopher D Gardner
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, California, United States.
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States; Chan Zuckerberg Biohub, San Francisco, CA, United States; Center for Human Microbiome Studies, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
16
|
Dissanayake IH, Tabassum W, Alsherbiny M, Chang D, Li CG, Bhuyan DJ. Lactic acid bacterial fermentation as a biotransformation strategy to enhance the bioavailability of phenolic antioxidants in fruits and vegetables: A comprehensive review. Food Res Int 2025; 209:116283. [PMID: 40253191 DOI: 10.1016/j.foodres.2025.116283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 04/21/2025]
Abstract
Fruits and vegetables (FVs) are rich sources of macro and micro-nutrients crucial for a healthy diet. In addition to these nutrients, FVs also contain fibre and phytochemicals known for their antioxidant properties. Despite the growing evidence of the disease-preventive role of antioxidants in FVs, their bioavailability and bioaccessibility vary significantly and have not been adequately explored. Lactic acid bacterial (LAB) fermentation is considered the most appropriate and accessible biotechnological approach to maintain and enhance the safety, nutritional, sensory and shelf-life properties of perishable foods such as FVs. This review critically assesses how LAB fermentation could be utilised as a promising biotransformation strategy to enhance the bioavailability of antioxidants in FVs. Furthermore, it discusses the potential use of uniquely nutritious Australian native fruits as suitable candidates for LAB fermentation. Further research is essential to identify the beneficial properties of bioactive compounds and effective LAB-based biotransformation strategies to improve the bioavailability and bioaccessibility of antioxidants in FVs.
Collapse
Affiliation(s)
| | - Wahida Tabassum
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
| | - Muhammad Alsherbiny
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt; Freedman Foundation Metabolomics Facility, Innovation Centre, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
| | - Chung Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia
| | - Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia; School of Science, Western Sydney University, Penrith, NSW 2751, Australia.
| |
Collapse
|
17
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Metris A, Walker AW, Showering A, Doolan A, McBain AJ, Ampatzoglou A, Murphy B, O'Neill C, Shortt C, Darby EM, Aldis G, Hillebrand GG, Brown HL, Browne HP, Tiesman JP, Leng J, Lahti L, Jakubovics NS, Hasselwander O, Finn RD, Klamert S, Korcsmaros T, Hall LJ. Assessing the safety of microbiome perturbations. Microb Genom 2025; 11. [PMID: 40371892 DOI: 10.1099/mgen.0.001405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
Everyday actions such as eating, tooth brushing or applying cosmetics inherently modulate our microbiome. Advances in sequencing technologies now facilitate detailed microbial profiling, driving intentional microbiome-targeted product development. Inspired by an academic-industry workshop held in January 2024, this review explores the oral, skin and gut microbiomes, focussing on the potential long-term implications of perturbations. Key challenges in microbiome safety assessment include confounding factors (ecological variability, host influences and external conditions like geography and diet) and biases from experimental measurements and bioinformatics analyses. The taxonomic composition of the microbiome has been associated with both health and disease, and perturbations like regular disruption of the dental biofilm are essential for preventing caries and inflammatory gum disease. However, further research is required to understand the potential long-term impacts of microbiome disturbances, particularly in vulnerable populations including infants. We propose that emerging technologies, such as omics technologies to characterize microbiome functions rather than taxa, leveraging artificial intelligence to interpret clinical study data and in vitro models to characterize and measure host-microbiome interaction endpoints, could all enhance the risk assessments. The workshop emphasized the importance of detailed documentation, transparency and openness in computational models to reduce uncertainties. Harmonisation of methods could help bridge regulatory gaps and streamline safety assessments but should remain flexible enough to allow innovation and technological advancements. Continued scientific collaboration and public engagement are critical for long-term microbiome monitoring, which is essential to advancing safety assessments of microbiome perturbations.
Collapse
Affiliation(s)
- Aline Metris
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Alan W Walker
- Microbiome, Food Innovation and Food Security Theme, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | | | | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Antonis Ampatzoglou
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Barry Murphy
- Unilever R&D Port Sunlight, Bebington, Wirral, UK
| | - Catherine O'Neill
- Division of Dermatology and Musculoskeletal Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | | | - Elizabeth M Darby
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | | | - Greg G Hillebrand
- University of Cincinnati, James L. Winkle College of Pharmacy, Cincinnati, OH, USA
| | - Helen L Brown
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Hilary P Browne
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College, Cork, Ireland
| | | | - Joy Leng
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Leo Lahti
- Department of Computing, University of Turku, Turku FI-20014, Finland
| | - Nicholas S Jakubovics
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Robert D Finn
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Silvia Klamert
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Tamas Korcsmaros
- Food, Microbiomes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Division of Digestive Diseases, Imperial College London, London, UK
- NIHR Imperial BRC Organoid Facility, Imperial College London, London, UK
| | - Lindsay J Hall
- Food, Microbiomes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
19
|
Costa RJS, Gaskell SK, Henningsen K, Jeacocke NA, Martinez IG, Mika A, Scheer V, Scrivin R, Snipe RMJ, Wallett AM, Young P. Sports Dietitians Australia and Ultra Sports Science Foundation Joint Position Statement: A Practitioner Guide to the Prevention and Management of Exercise-Associated Gastrointestinal Perturbations and Symptoms. Sports Med 2025; 55:1097-1134. [PMID: 40195264 DOI: 10.1007/s40279-025-02186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 04/09/2025]
Abstract
It is now well-established that exercise can disturb various aspects of gastrointestinal integrity and function. The pathophysiology of these perturbations, termed "exercise-induced gastrointestinal syndrome (EIGS)," can lead to exercise-associated gastrointestinal symptom (Ex-GIS) inconveniences. EIGS outcomes can impact physical performance and may lead to clinical manifestation warranting medical intervention, as well as systemic responses leading to fatality. Athlete support practitioners seek prevention and management strategies for EIGS and Ex-GIS. This current position statement aimed to critically appraise the role of EIGS and Ex-GIS prevention and management strategies to inform effective evidence-based practice and establish translational application. Intervention strategies with mostly consistent beneficial outcomes include macronutrient (i.e., carbohydrate and protein) intake and euhydration before and during exercise, dietary manipulation of fermentable oligo-, di-, and mono-saccharides and polyols (FODMAP), and gut training or feeding tolerance adjustments for the specific management of Ex-GIS from gastrointestinal functional issues. Strategies that may provide benefit and/or promising outcomes, but warrant further explorations include heat mitigating strategies and certain nutritional supplementation (i.e., prebiotics and phenols). Interventions that have reported negative outcomes included low-carbohydrate high-fat diets, probiotic supplementation, pharmaceutical administration, and feeding intolerances. Owing to individual variability in EIGS and Ex-GIS outcomes, athletes suffering from EIGS and/or support practitioners that guide athletes through managing EIGS, are encouraged to undertake gastrointestinal assessment during exercise to identify underlying causal and exacerbation factor/s, and adopt evidence-based strategies that provide individualized beneficial outcomes. In addition, abstaining from prevention and management strategies that present unclear and/or adverse outcomes is recommended.
Collapse
Affiliation(s)
- Ricardo J S Costa
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia.
| | - Stephanie K Gaskell
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | - Kayla Henningsen
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | | | - Isabel G Martinez
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | - Alice Mika
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| | - Volker Scheer
- Ultra Sports Science Foundation, Pierre-Benite, France
| | - Rachel Scrivin
- University of the Sunshine Coast, Sippy Downs, QLD, Australia
- Toi Ohomai Institute of Technology, Tauranga, New Zealand
| | - Rhiannon M J Snipe
- School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | | | - Pascale Young
- Department of Nutrition Dietetics and Food, Monash University, Level 1, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia
| |
Collapse
|
20
|
Nobels A, van Marcke C, Jordan BF, Van Hul M, Cani PD. The gut microbiome and cancer: from tumorigenesis to therapy. Nat Metab 2025; 7:895-917. [PMID: 40329009 DOI: 10.1038/s42255-025-01287-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 03/20/2025] [Indexed: 05/08/2025]
Abstract
The gut microbiome has a crucial role in cancer development and therapy through its interactions with the immune system and tumour microenvironment. Although evidence links gut microbiota composition to cancer progression, its precise role in modulating treatment responses remains unclear. In this Review, we summarize current knowledge on the gut microbiome's involvement in cancer, covering its role in tumour initiation and progression, interactions with chemotherapy, radiotherapy and targeted therapies, and its influence on cancer immunotherapy. We discuss the impact of microbial metabolites on immune responses, the relationship between specific bacterial species and treatment outcomes, and potential microbiota-based therapeutic strategies, including dietary interventions, probiotics and faecal microbiota transplantation. Understanding these complex microbiota-immune interactions is critical for optimizing cancer therapies. Future research should focus on defining microbial signatures associated with treatment success and developing targeted microbiome modulation strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Amandine Nobels
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
| | - Cédric van Marcke
- UCLouvain, Université catholique de Louvain, Institut de Recherche Expérimentale et Clinique (IREC), Pole of Medical Imaging, Radiotherapy and Oncology (MIRO), Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Breast Clinic, King Albert II Cancer Institute, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bénédicte F Jordan
- UCLouvain, Université catholique de Louvain, Biomedical Magnetic Resonance group (REMA), Louvain Drug Research Institute (LDRI), Brussels, Belgium
| | - Matthias Van Hul
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium.
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium.
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium.
| |
Collapse
|
21
|
BharathwajChetty B, Kumar A, Deevi P, Abbas M, Alqahtani A, Liang L, Sethi G, Liu L, Kunnumakkara AB. Gut microbiota and their influence in brain cancer milieu. J Neuroinflammation 2025; 22:129. [PMID: 40312370 PMCID: PMC12046817 DOI: 10.1186/s12974-025-03434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
Microbial communities are not simply remnants of the past but dynamic entities that continuously evolve under the selective pressures of nature, reflecting the intricate and adaptive processes of evolution. The microbiota residing in the various regions of the human body has numerous roles in different physiological processes such as nutrition, metabolism, immune regulation, etc. In the zeal of achieving empirical insights into the ambit of the gut microbiome, the research over the years led to the revelation of reciprocal interaction between the gut microbiome and the cognitive functioning of the human body. Dysbiosis in the gut microbial composition disturbs the homeostatic cognitive functioning of the human body. This dysbiosis has been associated with various chronic diseases, including brain cancer, such as glioma, glioblastoma, etc. This review explores the mechanistic role of dysbiosis-mediated progression of brain cancers and their subtypes. Moreover, it demonstrates the regulatory role of microbial metabolites produced by the gut microbiota, such as short-chain fatty acids, amino acids, lipids, etc., in the tumour progression. Further, we also provide valuable insights into the microbiota mediating the efficiency of therapeutic regimens, thereby leveraging gut microbiota as potential biomarkers and targets for improved treatment outcomes.
Collapse
Affiliation(s)
- Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Pranav Deevi
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia
| | - Athba Alqahtani
- Research Centre, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin Scool of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Le Liu
- Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China.
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
- International Joint M. Tech Degree in Food Science and Technology, Department of Chemical Engineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
22
|
Li S, Niu XX, Liu JL, Su M, Li QQ, Wang CY, Wang JJ, Chen HY, Ji D. Leveraging the gut microbiome to understand the risk factor of cognitive impairment in patients with liver cirrhosis. Eur J Gastroenterol Hepatol 2025; 37:627-637. [PMID: 39976005 DOI: 10.1097/meg.0000000000002934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
OBJECTIVES The role of the gut-liver axis in liver cirrhosis is becoming increasingly recognized. We investigated the fecal microbiome in patients with liver cirrhosis and its potential function as a predictive biomarker of hepatic encephalopathy. METHODS Patients were divided into either a high plasma ammonia (HPA) group or a low plasma ammonia (LPA) group according to the upper limit of normal of plasma ammonia concentration. 16S rRNA sequencing of fecal samples was performed to study how the microbiota affects the clinical symptoms of liver cirrhosis. The Stroop test was used to assess the ability of the brain to inhibit habitual behaviors. RESULTS Totally, 21 subjects were enrolled. Among the 18 patients with liver cirrhosis, 14 were male, the age range was 42-56 years, and the plasma ammonia level range was 20-125.9 μmol/l. The Stroop test showed more severe cognitive impairment in HPA than in LPA individuals. At the same time, there were significant differences in fecal microbiome characteristics between the two groups, characterized by a further increase in the abundance of the Proteobacteria phylum in the gut (especially aerobic Enterobacteriaceae ). Function predictions of Phylogenetic Investigation of Communities by Reconstruction of Unobserved States in the microbiome further explained the increase in the Enterobacteriaceae -dominated polyamine synthesis pathway in the gut microbiome of HPA groups. CONCLUSION Cirrhotic patients with hyperammonemia have a specific fecal bacterial composition (characterized via expansion of Enterobacteriaceae ). The ability to bio-synthesize polyamines that Enterobacteriaceae possesses is likely to be a key factor in the elevation of plasma ammonia.
Collapse
Affiliation(s)
- Shuyao Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Xiao-Xia Niu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Jia-Liang Liu
- Department of General Internal Medicine, Hospital of North China Electric Power University, Beijing, China
| | - Min Su
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Qian-Qian Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Chun-Yan Wang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Jian-Jun Wang
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| | - Hong-Yan Chen
- Department of General Internal Medicine, Hospital of North China Electric Power University, Beijing, China
| | - Dong Ji
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital
| |
Collapse
|
23
|
Chen Z, Xiao C, Zhang J, Jian S, Li P, Lin J, He C, Chen Z, Qi Y, Shi J, Chen Q, Chen J, Bo H. The Impact of Diet on the Colonization of Beneficial Microbes from an Ecological Perspective. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10069-10092. [PMID: 40234746 DOI: 10.1021/acs.jafc.5c02086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
With growing recognition of the pivotal role of gut microbiota in human health, probiotics have gained widespread attention for their potential to restore microbial homeostasis. However, a critical challenge persists: limited colonization efficiency among most probiotic strains compromises their therapeutic efficacy. This overview synthesizes ecological principles with cutting-edge microbiome research to elucidate the dynamic interplay between dietary components and probiotic colonization within the intestinal niche. This overview systematically analyzes: (1) stage-specific colonization mechanisms spanning microbial introduction, establishment, and proliferation; (2) nutrient-driven modulation of gut microbiota composition and function; and (3) the dual role of common dietary patterns as both facilitators and disruptors of probiotic persistence. Notably, this overview identifies key dietary strategies, including precision delivery of prebiotic fibers and polyphenol-microbiota crosstalk, that enhance niche adaptation through pH optimization, adhesion potentiation, and competitive exclusion of pathogens. Furthermore, this overview critically evaluates current limitations in probiotic research, particularly strain-specific variability and methodological constraints in simulating host-microbe-diet tripartite interactions. To bridge these gaps, this overview proposes an interdisciplinary framework integrating omics-driven strain selection, engineered delivery systems, and personalized nutrition models. Collectively, this work advances a mechanistic understanding of diet-microbiota interactions while providing actionable insights for developing targeted probiotic therapies and evidence-based dietary interventions to optimize gut ecosystem resilience.
Collapse
Affiliation(s)
- Zelin Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Chuntao Xiao
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jiantang Zhang
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Shiqi Jian
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Pinyue Li
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jiayi Lin
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Cai He
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Zixia Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Yutong Qi
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jingwen Shi
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Qizhu Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jun Chen
- College of Pharmacy, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Huaben Bo
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| |
Collapse
|
24
|
Zhou L, Chen H, He J, Ma H, Wang P, Wu X, Wang G, Li Y, Li H, Xu B, Wang R. The effect of heat-inactivated BBMN68 on immune regulation and gut microbiota in immunosuppressed mice and exploration of its immunomodulation postbiotic functional components. Int J Biol Macromol 2025; 311:143758. [PMID: 40316116 DOI: 10.1016/j.ijbiomac.2025.143758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/04/2025]
Abstract
Previous research shown that live probiotics have immunomodulatory effects, however the effectiveness of heat-inactivated bacteria and their immune-related components require further study. This study evaluated the immunomodulatory effects of heat-inactivated BBMN68 in BALB/c mice and explored immunomodulatory postbiotic components in vitro. Cyclophosphamide-induced immunosuppressed mice were orally administered heat-inactivated BBMN68 for 15 days. Postbiotic components from heat-inactivated BBMN68 were utilized to stimulate RAW264.7 cells for 24 h. In vivo, heat-inactivated BBMN68 significantly mitigated body weight decline, improved immune organ index and enhanced hematopoietic function in immunosuppressed mice. Additionally, it promoted delayed-type hypersensitivity responses and increased serum hemolysin and immunoglobulin (IgA, IgG, and IgM) levels. Oral administration of heat-inactivated BBMN68 increased serum cytokine (IL-2, TNF-α, IFN-γ, IL-4 and IL-6), while reducing IL-1β and IL-10. It also boosted beneficial microbiota (Lachnospiraceae_NK4A136_group and Dubosiella) and reduced harmful microbiota (Clostridium_sensu_stricto_1, Faecalibaculum and Romboutsia) by altering the Firmicutes/Bacteroidetes ratio. Unlike live probiotics, heat-inactivated BBMN68 is safer and has stronger immunomodulatory effects than Levamisole. It uniquely modulated gut microbiota compared to other heat-inactivated probiotics. In vitro, peptidoglycan from heat-inactivated BBMN68 cell wall suppressed TNF-α and IL-1β mRNA in RAW264.7 cells. These results suggest BBMN68 and its postbiotics can enhance immune responses, offering potential as safe immunomodulatory agents.
Collapse
Affiliation(s)
- Limian Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Huiyu Chen
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Jingjing He
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Hairan Ma
- Key Laboratory of Dairy Quality Digital Intelligent Monitoring Technology, State Administration for Market Regulation, Inner Mongolia Mengniu Dairy Group Co., Ltd., Beijing 101100, China
| | - Pengjie Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xiuying Wu
- Key Laboratory of Dairy Quality Digital Intelligent Monitoring Technology, State Administration for Market Regulation, Inner Mongolia Mengniu Dairy Group Co., Ltd., Beijing 101100, China
| | - Guohong Wang
- Key Laboratory of Dairy Quality Digital Intelligent Monitoring Technology, State Administration for Market Regulation, Inner Mongolia Mengniu Dairy Group Co., Ltd., Beijing 101100, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Hongliang Li
- Key Laboratory of Dairy Quality Digital Intelligent Monitoring Technology, State Administration for Market Regulation, Inner Mongolia Mengniu Dairy Group Co., Ltd., Beijing 101100, China.
| | - Baocai Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China.
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China.
| |
Collapse
|
25
|
Liu L, Qi W, Zhang N, Zhang J, Liu S, Wang H, Jiang L, Sun Y. Nutraceuticals for Gut-Brain Axis Health: A Novel Approach to Combat Malnutrition and Future Personalised Nutraceutical Interventions. Nutrients 2025; 17:1551. [PMID: 40362863 PMCID: PMC12073618 DOI: 10.3390/nu17091551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
The gut-brain axis (GBA) is a bidirectional communication network between the gastrointestinal tract and the brain, modulated by gut microbiota and related biomarkers. Malnutrition disrupts GBA homeostasis, exacerbating GBA dysfunction through gut dysbiosis, impaired neuroactive metabolite production, and systemic inflammation. Nutraceuticals, including probiotics, prebiotics, synbiotics, postbiotics, and paraprobiotics, offer a promising approach to improving GBA homeostasis by modulating the gut microbiota composition and related neuroactive metabolites. This review aims to elucidate the interplay between gut microbiota-derived biomarkers and GBA dysfunction in malnutrition and evaluate the potential of nutraceuticals in combating malnutrition. Furthermore, it explores the future of personalised nutraceutical interventions tailored to individual genetic and microbiome profiles, providing a targeted approach to optimise health outcomes. The integration of nutraceuticals into GBA health management could transform malnutrition treatment and improve cognitive and metabolic health.
Collapse
Affiliation(s)
- Litai Liu
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
- Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6UR, UK
| | - Wen Qi
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Na Zhang
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Jinhao Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Shen Liu
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| | - Huan Wang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Lianzhou Jiang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.Z.); (H.W.); (L.J.)
| | - Ying Sun
- Tourism & Cuisine College, Harbin University of Commerce, Harbin 150028, China; (L.L.); (W.Q.); (N.Z.); (S.L.)
| |
Collapse
|
26
|
Liu X, Alharbi A, Gibson R, Rodriguez-Mateos A. (Poly)phenol-gut microbiota interactions and their impact on human health. Curr Opin Clin Nutr Metab Care 2025:00075197-990000000-00218. [PMID: 40293967 DOI: 10.1097/mco.0000000000001132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
PURPOSE OF REVIEW (Poly)phenols are bioactive compounds in plant-based foods with well documented health benefits. Their metabolism in dependent on gut microbiota composition, diversity, and function, which modulate their bioavailability and physiological effects. This review examines recent insights into (poly)phenol-gut microbiota interactions, emphasizing their relevance in mediating health benefits and mechanisms of action. RECENT FINDINGS Recent studies have identified a high interindividual variability in (poly)phenol metabolism, explored microbial-derived metabolites with potential health benefits, and revealed bidirectional influences between (poly)phenols and gut microbiota composition. Advances in analytical techniques and large-scale studies have refined our understanding of these interactions, but inconsistencies remain in linking specific microbial signatures to health outcomes. SUMMARY Standardized methodologies and long-term studies are needed to clarify the impact of (poly)phenol-derived metabolites on human health. Future research should focus on personalized and targeted nutrition approaches to optimize (poly)phenol benefits.
Collapse
Affiliation(s)
- Xiaohe Liu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Amnah Alharbi
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Rachel Gibson
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ana Rodriguez-Mateos
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
27
|
Lee SH, Han C, Shin C. IUPHAR Review: Microbiota-Gut-Brain Axis and its role in Neuropsychiatric Disorders. Pharmacol Res 2025; 216:107749. [PMID: 40306604 DOI: 10.1016/j.phrs.2025.107749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
The human gut microbiome, composed of a vast array of microorganisms that have co-evolved with humans, is crucial for the development and function of brain systems. Research has consistently shown bidirectional communication between the gut and the brain through neuronal, endocrine, and immunological, and chemical pathways. Recent neuroscience studies have linked changes in the microbiome and microbial metabolites to various neuropsychiatric disorders such as autism, depression, anxiety, schizophrenia, eating disorders, and neurocognitive disorders. Novel metagenome-wide association studies have confirmed these microbiome variations in large samples and expanded our understanding of the interactions between human genes and the gut microbiome. The causal relationship between gut microbiota and neuropsychiatric disorders is being elucidated through the establishment of large cohort studies incorporating microbiome data and advanced statistical techniques. Ongoing animal and human studies focused on the microbiota-gut-brain axis are promising for developing new prevention and treatment strategies for neuropsychiatric conditions. The scope of these studies has broadened from microbiome-modulating therapies including prebiotics, probiotics, synbiotics and postbiotics to more extensive approaches such as fecal microbiota transplantation. Recent systematic reviews and meta-analyses have strengthened the evidence base for these innovative treatments. Despite extensive research over the past decade, many intriguing aspects still need to be elucidated regarding the role and therapeutic interventions of the microbiota-gut-brain axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Fan D, Hu J, Lin N. Effects of probiotics, prebiotics, synbiotics and postbiotics on pediatric asthma: a systematic review. Front Nutr 2025; 12:1586129. [PMID: 40352259 PMCID: PMC12061971 DOI: 10.3389/fnut.2025.1586129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Background Pediatric asthma, a prevalent chronic disease with rising global incidence, imposing substantial healthcare and socioeconomic burdens. Emerging evidence highlights the gut-lung axis as a pivotal therapeutic target, with microbiota dysbiosis implicated in immune dysregulation and airway hyperresponsiveness. This systematic review evaluated the efficacy and safety of probiotics, prebiotics, synbiotics, and postbiotics in pediatric asthma management. Methods A comprehensive search of PubMed, Cochrane library, Web of Science, and Embase was conducted up to 2nd January 2025. Inclusion criteria encompassed randomized controlled trials (RCTs) evaluating the therapeutic use of probiotics, prebiotics, synbiotics, or postbiotics in children and/or adolescents (<18 years) with asthma. Results Eighteen studies (13 RCTs, n = 2,419 participants) were analyzed, focusing on children aged < 18 years. Probiotic interventions, predominantly Lactobacillus (5 studies) and Bifidobacterium (5 studies), demonstrated significant reductions in asthma exacerbations and improved pulmonary function, with strain-specific effects linked to Th2 cytokine suppression and gut-lung axis modulation. Postbiotics, including bacterial lysates (OM-85 BV, PMBL®), attenuated airway hyperresponsiveness and systemic inflammation. Synbiotics reduced viral respiratory infections and healthcare utilization. However, there is still a lack of direct RCTs to explore the therapeutic effects of prebiotics on pediatric asthma. Key limitations include methodological heterogeneity (dosing: 108-1010 CFU/day; duration: 8 weeks-12 months) and risk of bias (3 low-risk, 12 with concerns). Conclusion Our findings underscored the potential of microbiota-targeted therapies but highlight the need for standardized protocols, strain-specific trials, and pediatric prebiotic research. Future studies should integrate multi-omics to elucidate mechanisms and optimize personalized interventions. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42025641318, identifier: CRD42025641318.
Collapse
Affiliation(s)
| | | | - Ning Lin
- Clinical Nutrition, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
29
|
Hemalatha M, Dev A, Mahajan R, De D, Handa S. Non-pharmacological therapies and their efficacy in atopic dermatitis: A narrative review. Indian J Dermatol Venereol Leprol 2025; 0:1-9. [PMID: 40357939 DOI: 10.25259/ijdvl_1076_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/27/2025] [Indexed: 05/15/2025]
Abstract
Atopic dermatitis (AD) is a complex immune-mediated disease characterised by recurrent eczematous lesions and pruritus, which adversely affects the quality of life (QoL). Genetic factors, environmental factors, immune dysregulation, and skin barrier dysfunction contribute to its pathophysiology. Non-pharmacological management strategies aim to preserve the skin barrier, address immune dysregulation, and minimise triggers. In this review, wediscuss various non-pharmacological interventions, including allergen (aeroallergens, food allergens, and contact allergens) avoidance, bathing-related measures, moisturisers, clothing choices, therapies targeting the skin microbiome, and allergen-specific immunotherapy, in addition to education and psychotherapy. Non-pharmacological therapies are essential for the holistic management of AD, but their effectiveness varies, highlighting the need for further research and tailored approaches to individual patient needs.
Collapse
Affiliation(s)
- Mude Hemalatha
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anubha Dev
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rahul Mahajan
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Dipankar De
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjeev Handa
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
30
|
Häsler R, Mikš MH, Bajic D, Soyyilmaz B, Bendik I, van Buul VJ, Steinert RE, Rehman A. Human Milk Oligosaccharides Modulating Inflammation in Infants, Adults, and Older Individuals-From Concepts to Applications. Adv Nutr 2025; 16:100433. [PMID: 40287068 DOI: 10.1016/j.advnut.2025.100433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
The increasing global prevalence of inflammatory diseases, such as ulcerative colitis and irritable bowel syndrome, represents a challenging task for healthcare systems. Several approaches to disease management target the intestinal microbiome, which plays a key role in health and disease. One promising approach is modulating the microbiome using human milk oligosaccharides (HMOs). Originating from human milk, HMOs are indigestible carbohydrates that act in a host-optimized prebiotic fashion by providing an energy source for health-promoting intestinal bacteria and exhibiting systemic effects. Commercial products supporting infant health and development have been the primary fields of HMO application. Advancements in the large-scale production of HMOs through bioengineering and precision fermentation have led to evaluation of their potential for managing inflammatory diseases. Several in vitro studies and observations on model systems have been clinically validated in infants, resulting in a large body of evidence supporting the safety and efficacy of HMOs in inflammatory disorders. Although novel approaches seek to explore interventions in adults, the primary goal for the future is to provide cost-efficient, safe, and reliable healthcare compounds across all age groups.
Collapse
Affiliation(s)
- Robert Häsler
- Department of Dermatology and Allergology, University Kiel, Kiel, Germany.
| | - Marta Hanna Mikš
- Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland; dsm-firmenich, Glycom A/S, Hørsholm, Denmark
| | - Danica Bajic
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| | | | - Igor Bendik
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| | | | | | - Ateequr Rehman
- dsm-firmenich, Health, Nutrition & Care, Kaiseraugst, Switzerland
| |
Collapse
|
31
|
Demirhan HK, Omer Oglou E, Aksoy ZB, Kiran F. Evaluation of the anti-inflammatory, antioxidant and regenerative effects of microbiota-derived postbiotics in human periodontal ligament mesenchymal stromal cells. Clin Oral Investig 2025; 29:262. [PMID: 40263129 PMCID: PMC12014813 DOI: 10.1007/s00784-025-06341-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
OBJECTIVE This study investigates the regenerative and protective effects of postbiotics (cell-free supernatant) derived from the Lactiplantibacillus plantarum EIR/IF-1 strain on human periodontal ligament mesenchymal stromal cells (hPDL-MSCs). MATERIALS AND METHODS hPDL-MSCs were isolated from periodontal ligament tissues (PDL) of wisdom teeth using enzymatic digestion and subsequently characterized through immunophenotyping. The effect of postbiotics on the viability of hPDL-MSCs was assessed using the MTT assay and flow cytometry, while their impact on cell migration was evaluated via the scratch assay. Anti-inflammatory effects of postbiotics were investigated on lipopolysaccharide (LPS, derived from Porphyromonas gingivalis)-stimulated hPDL-MSCs through Enzyme-Linked Immunosorbent Assay (ELISA). Additionally, the antioxidant effects of postbiotics were analyzed in hydrogen peroxide (H₂O₂)-induced hPDL-MSCs by measuring reactive oxygen species (ROS) levels using flow cytometry. The expression of collagen type I (COL1A1) gene was further assessed by quantitative reverse transcription PCR and immunofluorescence staining. RESULTS Treatment with postbiotics (250 µg/mL) significantly increased the viability and migration capability of hPDL-MSCs, while enhancing collagen production for PDL repair. Treatment with postbiotics for 24 h resulted in a 54.53 ± 2.01% reduction in intracellular ROS levels compared to untreated H2O2-induced hPDL-MSCs. Furthermore, postbiotics significantly decreased the production of pro-inflammatory cytokines (IL-8, IL-6, and IL-1β), and increased the anti-inflammatory cytokine IL-10 (2.67-fold) compared to untreated LPS-stimulated hPDL-MSCs. CONCLUSION Our findings indicate that postbiotics exhibit biological activity throughout all stages of the healing process, beginning with the modulation of the inflammatory response to LPS stimulation, followed by the promotion of cell migration, proliferation, and collagen synthesis. Given the unmet need for safe and adjuvant therapeutic approaches that promote comprehensive periodontal regeneration in periodontal diseases, this study presents postbiotics as a promising candidate. CLINICAL RELEVANCE Postbiotics could be integrated into regenerative therapies as a novel bioactive material to improve the healing and regenerative outcomes in periodontal defects by both controlling inflammation and stimulating tissue repair processes.
Collapse
Affiliation(s)
- Hazal Kibar Demirhan
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey
- Graduate School of Natural and Applied Sciences, Ankara University, Ankara, 06110, Turkey
| | - Emine Omer Oglou
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey
- Graduate School of Natural and Applied Sciences, Ankara University, Ankara, 06110, Turkey
| | | | - Fadime Kiran
- Pharmabiotic Technologies Research Laboratory, Department of Biology, Faculty of Science, Ankara University, Ankara, 06100, Turkey.
| |
Collapse
|
32
|
Jiao J, Zhao Y, Tan Z. Leveraging Gastrointestinal Microbiota‒Host Co-oscillation Patterns for Improving Nitrogen Utilization Efficiency in Ruminants: A Review. J Nutr 2025:S0022-3166(25)00183-X. [PMID: 40204020 DOI: 10.1016/j.tjnut.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/10/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Improving nitrogen utilization efficiency in ruminant livestock is vital for feeding a growing global population and lowering environmental pollution. The rumen and intestine harbor distinct epithelial structures and biogeographically stratified microbiota, and their co-oscillation assemblage patterns fulfill the pivotal role of metabolizing dietary nitrogen into bioavailable nutrients in ruminants. There is cursory evidence to suggest that an increased understanding of the spatial gastrointestinal microbiota‒host interactions will aid in the development of nutritional strategies to improve nitrogen utilization efficiency. In this review, we first explore the current knowledge on the processes of protein degradation, microbial protein synthesis, and urea nitrogen salvage in ruminal microorganisms and the epithelium. Second, we summarize the mechanisms of microbiota‒host interplay with regard to the amino acid utilization process in the intestine. Finally, we discuss the most pertinent and promising manipulation strategies that have emerged to balance food security and environmental impacts. In this review, we highlight the significance of leveraging gastrointestinal microbiota‒host co-oscillation patterns to improve nitrogen utilization efficiency, and put forward perspectives for future research opportunities that precisely target this coordinated interplay in the nitrogen metabolic network.
Collapse
Affiliation(s)
- Jinzhen Jiao
- State Key Laboratory of Forage Breeding-by-Design and Utilization, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; Yuelushan Laboratory, Changsha, Hunan, China
| | - Yechan Zhao
- State Key Laboratory of Forage Breeding-by-Design and Utilization, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Zhiliang Tan
- State Key Laboratory of Forage Breeding-by-Design and Utilization, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China; Yuelushan Laboratory, Changsha, Hunan, China.
| |
Collapse
|
33
|
Avcı E, Avcı GA. Effect of new bioactive substances obtained from possible probiotic bacteria on miRNA expression by in vitro cocultivation in colorectal cancer cells. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2025:1-10. [PMID: 40260513 DOI: 10.1080/09603123.2025.2495195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
In our study, were evaluated effect on colorectal cancer cells of possible probiotic bacteria and new bioactive substances (paraprobiotics/postbiotics), miRNA-expression. Lactobacillus rhamnosus, Lactobacillus reuteri strains, HT-29, and Caco-2 were used. The cytotoxicity of the biotics was determined by MTT and miRNA expression. In line with the data obtained, it was determined that probiotics had a proliferative effect on the fibroblast cell line and a cytotoxic effect on cancer cells. It was observed that paraprobiotics had a minimal effect compared to probiotics, postbiotics had a greater effect. In this work, the activities of new pharmabiotics are compared with living cells, eliminating the limitations of probiotics in terms of shelf life and viability, and thus allowing the creation of new commercial domestic and national products, which increases the unique value of our study. In addition, there will be safer alternatives for suppressed immune systems, and various disadvantages of probiotics can be eliminated.
Collapse
Affiliation(s)
- Emre Avcı
- Department of Biochemistry, Faculty of Gulhane Pharmacy, University of Health Sciences, Ankara, Turkiye
| | - Gulcin Alp Avcı
- Department of Basic Medical Sciences, Faculty of Gulhane Dentistry, University of Health Sciences, Ankara, Turkiye
| |
Collapse
|
34
|
Urbani G, Rondini E, Distrutti E, Marchianò S, Biagioli M, Fiorucci S. Phenotyping the Chemical Communications of the Intestinal Microbiota and the Host: Secondary Bile Acids as Postbiotics. Cells 2025; 14:595. [PMID: 40277921 PMCID: PMC12025480 DOI: 10.3390/cells14080595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025] Open
Abstract
The current definition of a postbiotic is a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics can be mainly classified as metabolites, derived from intestinal bacterial fermentation, or structural components, as intrinsic constituents of the microbial cell. Secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA) are bacterial metabolites generated by the enzymatic modifications of primary bile acids by microbial enzymes. Secondary bile acids function as receptor ligands modulating the activity of a family of bile-acid-regulated receptors (BARRs), including GPBAR1, Vitamin D (VDR) receptor and RORγT expressed by various cell types within the entire human body. Secondary bile acids integrate the definition of postbiotics, exerting potential beneficial effects on human health given their ability to regulate multiple biological processes such as glucose metabolism, energy expenditure and inflammation/immunity. Although there is evidence that bile acids might be harmful to the intestine, most of this evidence does not account for intestinal dysbiosis. This review examines this novel conceptual framework of secondary bile acids as postbiotics and how these mediators participate in maintaining host health.
Collapse
Affiliation(s)
- Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Elena Rondini
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, 06123 Perugia, Italy; (E.R.); (E.D.)
| | - Silvia Marchianò
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| | - Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06123 Perugia, Italy; (G.U.); (S.M.); (M.B.)
| |
Collapse
|
35
|
Abdul Manan M. Progress in Probiotic Science: Prospects of Functional Probiotic-Based Foods and Beverages. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2025; 2025:5567567. [PMID: 40259922 PMCID: PMC12011469 DOI: 10.1155/ijfo/5567567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
This comprehensive review explores the evolving role of probiotic-based foods and beverages, highlighting their potential as functional and "future foods" that could significantly enhance nutrition, health, and overall well-being. These products are gaining prominence for their benefits in gut health, immune support, and holistic wellness. However, their future success depends on addressing critical safety concerns and navigating administrative complexities. Ensuring that these products "do more good than harm" involves rigorous evaluations of probiotic strains, particularly those sourced from the human gastrointestinal tract. Lactic acid bacteria (LABs) serve as versatile and effective functional starter cultures for the development of probiotic foods and beverages. The review emphasizes the role of LABs as functional starter cultures and the development of precision probiotics in advancing these products. Establishing standardized guidelines and transparent practices is essential, requiring collaboration among regulatory bodies, industry stakeholders, and the scientific community. The review underscores the importance of innovation in developing "friendly bacteria," "super probiotics," precision fermentation, and effective safety assessments. The prospects of functional probiotic-based foods and beverages rely on refining these elements and adapting to emerging scientific advancements. Ultimately, empowering consumers with accurate information, fostering innovation, and maintaining stringent safety standards will shape the future of these products as trusted and beneficial components of a health-conscious society. Probiotic-based foods and beverages, often infused with LABs, a "friendly bacteria," are emerging as "super probiotics" and "future foods" designed to "do more good than harm" for overall health.
Collapse
Affiliation(s)
- Musaalbakri Abdul Manan
- Food Science and Technology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), MARDI Headquarters, Persiaran MARDI-UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
36
|
Kang A, Eor JY, Lee J, Kwak MJ, Lee DJ, Seo E, Lee WJ, Son SH, Song M, Kim JM, Kim HW, Yang J, Oh S, Kim Y. Lacticaseibacillus casei IDCC 3451 alleviates cognitive and behavioral functions by reshaping the gut microbiome and regulating intestinal barrier integrity in chronic stress animal models. Curr Res Food Sci 2025; 10:101051. [PMID: 40290371 PMCID: PMC12023876 DOI: 10.1016/j.crfs.2025.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Lacticaseibacillus casei IDCC 3451 (3451) was evaluated for its effects on the gut-brain axis using Caenorhabditis elegans (C. elegans) and mouse models of stress and inflammation. In C. elegans, 3451 extended lifespans by 25 %, improved motility, and chemotaxis, enhanced survival under pathogen challenge, and reduced amyloid beta accumulation by 42 %. Transcriptomic profiling revealed upregulation of genes involved in neurotransmitter signaling and serine/threonine pathways. In the unpredictable chronic mild stress (UCMS) mouse model, 3451 administration increased the time spent in the center of the open field by 65 % and reduced immobility in the forced swim test by 32 %, indicating anxiolytic and antidepressant effects. Serum levels of aspartate aminotransferase (AST) and gamma-glutamyl transferase (GGT) were decreased by 18 % and 24 %, respectively. Additionally, 3451 restored the expressions of 5HT1AR, GABAR, and tight junction proteins, including ZO-1 and Claudin1. Metabolomic analysis showed increased glycine and decreased palmitic acid levels, associated with an increased abundance of Ruminococcus and Akkermansia. In the dextran sulfate sodium (DSS)-induced colitis model, 3451 reduced the disease activity index by 36 %, improved colon histology, increased goblet cell preservation, and upregulated ZO-1 and IL-10 expression. Threonine levels were also increased and correlated with a higher abundance of Coprococcus. These findings demonstrate that 3451 improved behavioral and intestinal outcomes through coordinated modulation of host signaling, metabolite production, and gut microbial composition, highlighting its therapeutic potential for managing IBD and neurobehavioral disorders.
Collapse
Affiliation(s)
- Anna Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Junbeom Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Eunsol Seo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Woong Ji Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Seon-hui Son
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, 34134, South Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Gyeonggi-do, South Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul, 05006, South Korea
| | - Jungwoo Yang
- Department of Microbiology, College of Medicine, Dongguk University, Gyeongju, 38066, South Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| |
Collapse
|
37
|
Wang N, Wu P, Chen XD. New Insights into a Conceptual Bionic Colonic Bioreactor: A Model, 'Probiotics in Human Colon', Showing How Probiotics Alleviate Constipation from a Bioprocess Engineering Perspective. Foods 2025; 14:1335. [PMID: 40282737 PMCID: PMC12027397 DOI: 10.3390/foods14081335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Constipation is a common problem which often causes negative impacts on the patient's quality of life. Apart from the pharmacologic and diet approaches, the use of probiotics has gradually shown promising efficacy to alleviate constipation. However, an exact understanding of the underlying mechanisms of probiotic actions on alleviating constipation is still unclear and need to be explored. In this review, we propose a model, 'probiotics in human colon', from a bioprocess engineering perspective. This model can be interpreted as a new concept of bionic colonic bioreactor design of a human colon in vitro, in which the transport phenomena during the fermentation of chyme by probiotics can be detected. By reviewing the anatomy structure and peristalsis mode of the human colon, we have focused on the influence by probiotics on the physical properties of colonic contents during the fermentation process. We relate physical properties such as shape, water content, density, hardness, viscosity, and elasticity to constipation symptoms directly. The influences on the physical properties of colon contents triggered by probiotics can be a potential key to understand the mechanisms for alleviating constipation.
Collapse
Affiliation(s)
- Ni Wang
- Life Quality Engineering Interest Group, School of Chemical and Environmental Engineering, College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou 215123, China;
| | | | - Xiao Dong Chen
- Life Quality Engineering Interest Group, School of Chemical and Environmental Engineering, College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou 215123, China;
| |
Collapse
|
38
|
Mishra S, Jain S, Agadzi B, Yadav H. A Cascade of Microbiota-Leaky Gut-Inflammation- Is it a Key Player in Metabolic Disorders? Curr Obes Rep 2025; 14:32. [PMID: 40208464 DOI: 10.1007/s13679-025-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE OF REVIEW This review addresses critical gaps in knowledge and provides a literature overview of the molecular pathways connecting gut microbiota dysbiosis to increased intestinal permeability (commonly referred to as "leaky gut") and its contribution to metabolic disorders. Restoring a healthy gut microbiota holds significant potential for enhancing intestinal barrier function and metabolic health. These interventions offer promising therapeutic avenues for addressing leaky gut and its associated pathologies in metabolic syndrome. RECENT FINDINGS In metabolic disorders such as obesity and type 2 diabetes (T2D), beneficial microbes such as those producing short-chain fatty acids (SCFAs) and other key metabolites like taurine, spermidine, glutamine, and indole derivatives are reduced. Concurrently, microbes that degrade toxic metabolites such as ethanolamine also decline, while proinflammatory, lipopolysaccharide (LPS)-enriched microbes increase. These microbial shifts place a higher burden on intestinal epithelial cells, which are in closest proximity to the gut lumen, inducing detrimental changes that compromise the structural and functional integrity of the intestinal barrier. Such changes include exacerbation of tight junction protein (TJP)s dysfunction, particularly through mechanisms such as destabilization of zona occludens (Zo)-1 mRNA or post-translational modifications. Emerging therapeutic strategies including ketogenic and Mediterranean diets, as well as probiotics, prebiotics, synbiotics, and postbiotics have demonstrated efficacy in restoring beneficial microbial populations, enhancing TJP expression and function, supporting gut barrier integrity, reducing leaky gut and inflammation, and ultimately improving metabolic disorders. This review summarizes the mechanisms by which gut microbiota contribute to the development of leaky gut and inflammation associated with metabolic syndrome. It also explores strategies for restoring gut microbiota balance and functionality by promoting beneficial microbes, increasing the production of beneficial metabolites, clearing toxic metabolites, and reducing the proportion of proinflammatory microbes. These approaches can alleviate the burden on intestinal epithelial cells, reduce leaky gut and inflammation, and improve metabolic health.
Collapse
Affiliation(s)
- Sidharth Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bryan Agadzi
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Director of USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, MDC78, Tampa, FL, 33612, USA.
| |
Collapse
|
39
|
Zhu X, Zhang X, Zhang Y, Li F. Supplemental Probiotics, Postbiotics, and Their Combination on the Growth, Slaughter Variables, Organ Development, Intestinal Morphology, and Cecal Microbiota of Broilers. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10494-6. [PMID: 40205163 DOI: 10.1007/s12602-025-10494-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2025] [Indexed: 04/11/2025]
Abstract
This study was performed to evaluate the effects of dietary supplementation with viable Bacillus subtilis, inactivated Lactobacillus plantarum, and their combination on growth and slaughter performance, immune organ and small intestine development, and cecal microbiota of broilers. A total of 480 one-day-old broilers were used across a 42-day feeding experiment and were fed a basal diet, a basal diet + 300 mg/kg viable B. subtilis (probiotic), 320 mg/kg heat-killed L. plantarum (postbiotic), or their mixtures (combination). Each diet had six replicates, and each replicate had 20 broilers. Compared to the control group, the final body weight and average daily gain increased, and the feed conversion ratio decreased in the probiotic and postbiotic groups, while the combination group showed parameters comparable to those of the postbiotic group. Feeding broilers with probiotic, postbiotic, and their combination increased the dressing-out percentage, indices of the spleen and bursa of Fabricius, and lengths and weights of the small intestine. In addition, the villus height/crypt depth ratio of both the duodenum and jejunum was increased in the probiotic and postbiotic groups. Broilers in the combination group had a lower abundance of Proteobacteria, while those in the probiotic and postbiotic groups had a higher abundance of Desulfovibrio in the cecum. Overall, we concluded that dietary supplementation with viable B. subtilis and inactivated L. plantarum could improve growth and slaughter performance, organ development, intestinal morphology, and cecal microbiota of broilers. However, their combination had not yielded a synergistic and additive effect on broilers.
Collapse
Affiliation(s)
- Xin Zhu
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Xinjie Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Yong Zhang
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Fangfang Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| |
Collapse
|
40
|
Guo J, Zhao Y, Guo W, Sun Y, Zhang W, Zhao Q, Zhang Y, Jiang Y. Effects of Lactobacillus paracei JY062 Postbiotic on Intestinal Barrier, Immunity, and Gut Microbiota. Nutrients 2025; 17:1272. [PMID: 40219029 PMCID: PMC11990213 DOI: 10.3390/nu17071272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Research on postbiotics derived from probiotic fermented milk bases require further expansion, and the mechanisms through which they exert their effects have yet to be fully elucidated. This study utilized in vitro cell co-culture, digestion, and fermentation experiments, combined with targeted T500 technology, to elucidate the mechanism by which postbiotic Pa JY062 safeguards intestinal health. Compared to the LPS group, Pa JY062 boosted phagocytic ability in RAW264.7 macrophages, decreased NO levels, and alleviated LPS-induced excessive inflammation. Pa JY062 suppressed pro-inflammatory cytokines (IL-6, IL-17α, and TNF-α) while elevating anti-inflammatory IL-10. It prevented LPS-induced TEER reduction in Caco-2 monolayers, decreased FITC-dextran permeability, restored intestinal microvilli integrity, and upregulated tight junction genes (ZO-1, occludin, claudin-1, and E-cadherin). The hydrolysis rate of Pa JY062 progressively rose in gastrointestinal fluids in 0-120 min. At 5 mg/mL, it enriched gut microbiota diversity and elevated proportions of Limosilactobacillus, Lactobacillus, Pediococcus, and Lacticaseibacillus while augmenting the microbial production of acetic acid (120.2 ± 8.08 μg/mL), propionic acid (9.9 ± 0.35 μg/mL), and butyric acid (10.55 ± 0.13 μg/mL). Pa JY062 incorporated αs-casein/β-lactoglobulin hydrolysate (L-glutamic acid, alanine, lysine, tyrosine, phenylalanine, histidine, and arginine) to mitigate protein allergenic potential while harboring bioactive components, including tryptophan metabolites, vitamin B6 (VB6), and γ-aminobutyric acid (GABA). Pa JY062 represented a novel postbiotic with demonstrated intestinal health-promoting properties. These findings advance the current knowledge on postbiotic-mediated gut homeostasis regulation and expedite the translational development of dairy-derived postbiotic formulations.
Collapse
Affiliation(s)
- Jinfeng Guo
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Ying Zhao
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Wenqian Guo
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yilin Sun
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Wei Zhang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Qianyu Zhao
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yu Zhang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
| | - Yujun Jiang
- Key Lab of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; (J.G.); (Y.Z.); (W.G.); (Y.S.); (W.Z.); (Q.Z.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| |
Collapse
|
41
|
Kumar M, Devi WM, Choudhury TG, Kamilya D, Monsang SJ, Irungbam S, Saha RK. Unraveling the Bioactivities and Immunomodulatory Potential of Postbiotics Derived from Bacillus subtilis and B. amyloliquefaciens for Aquaculture. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10528-z. [PMID: 40186049 DOI: 10.1007/s12602-025-10528-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/07/2025]
Abstract
Postbiotics are molecules or soluble factors released as a result of a probiotic's metabolic activity. Their use in enhancing the growth, health, and disease resistance of aquatic animals has gained considerable attention in aquaculture. The present investigation was designed to assess the beneficial effects of postbiotic products derived from two probiotic strains, Bacillus subtilis and B. amyloliquefaciens. Postbiotics from B. subtilis exhibited significantly greater (p < 0.05) antibacterial activity against various pathogenic bacterial strains, more robust antagonistic growth kinetics, stronger anti-virulence potential, enhanced inhibition of biofilm formation, and increased antioxidant activity compared to those from B. amyloliquefaciens. Additionally, B. subtilis postbiotics triggered a significant (p < 0.05) cellular immune response, including higher myeloperoxidase activity, leucocyte proliferation, and production of nitric oxide and superoxide anions, along with a notable upregulation of immune-related gene expression (IL-1β, IL-10, IFN-γ, and TNF-α) in the head kidney leucocytes of Labeo rohita. A challenge test on L. rohita fingerlings confirmed the safety of B. subtilis postbiotics. These findings highlight the anti-pathogenic, immunostimulatory, and disease-resistant properties of B. subtilis postbiotics, suggesting their promising application in aquaculture.
Collapse
Affiliation(s)
- Monalisha Kumar
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India
| | - Wangkheimayum Malemnganbi Devi
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India
| | - Tanmoy Gon Choudhury
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India.
| | - Dibyendu Kamilya
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur, 721302, West Bengal, India.
| | - Shongsir Joy Monsang
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India
| | - Surajkumar Irungbam
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India
| | - Ratan Kumar Saha
- Department of Aquatic Health and Environment, College of Fisheries, Central Agricultural University (Imphal), Lembucherra, Tripura, 799210, India
- Techno India University, Tripura, Agartala, 799004, Tripura, India
| |
Collapse
|
42
|
Mosiej W, Długosz E, Kruk M, Zielińska D. Immunomodulatory Properties of Live and Thermally-Inactivated Food-Origin Lactic Acid Bacteria-In Vitro Studies. Mol Nutr Food Res 2025:e70047. [PMID: 40166824 DOI: 10.1002/mnfr.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
The study investigates the strain-specific immunomodulatory properties of live and thermally-inactivated (TI) lactic acid bacteria (LAB) derived from traditional Polish fermented foods, focusing on their potential as probiotics and postbiotics. LAB strains, known for their role in food fermentation, were assessed for their ability to influence cytokine production in THP-1 macrophages, maintain intestinal epithelial barrier integrity in Caco-2 monolayers, exhibit antioxidant activity, and produce specific organic acids and sugars. The research demonstrated that live LAB strains significantly upregulated the anti-inflammatory cytokine IL-10, particularly under inflammatory conditions, while TI strains exhibited notable antioxidant and anti-inflammatory properties. TI strains showed a greater ability to protect epithelial barrier function and reduce pro-inflammatory cytokine secretion than live strains, suggesting a promising role for postbiotics. The findings underscore the potential of LAB from fermented foods, demonstrating that postbiotic derivatives can differently influence inflammation compared to live bacteria, highlighting their potential as immune-enhancing agents, capable of modulating immune responses and offering therapeutic benefits against inflammation-related disorders. However, the limitations of in vitro models highlight the need for further in vivo and clinical studies to validate these effects and fully uncover the health benefits of these LAB strains for humans.
Collapse
Affiliation(s)
- Wioletta Mosiej
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Ewa Długosz
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Marcin Kruk
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| |
Collapse
|
43
|
López Martí Á, Montero Palma C, López Martí H, Ranchal Sánchez A. Efficacy of probiotic, prebiotic, synbiotic and postbiotic supplementation on gastrointestinal health in cats: systematic review and meta-analysis. J Small Anim Pract 2025; 66:219-235. [PMID: 39800337 PMCID: PMC12000713 DOI: 10.1111/jsap.13822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/25/2024] [Accepted: 12/09/2024] [Indexed: 04/17/2025]
Abstract
OBJECTIVES The clinical efficacy of the use of probiotics, prebiotics, synbiotics and postbiotics (biotics) in cats is unknown, despite their use in daily practice. The objectives of the study is to evaluate the effectiveness of biotic supplementation in treating and preventing gastroenteropathies, and in reducing gastrointestinal signs associated with antibiotics in cats. MATERIALS AND METHODS A systematic review was conducted by searching four databases for publications before August 2, 2024, following a pre-registered protocol. Eligible publications were trials involving healthy cats or those with gastroenteropathies, supplemented with biotics (and an inactive control), studying outcomes such as faecal consistency, faecal microbiota or vomiting. Risk of bias and quality of reports were assessed. Effects were synthesised by meta-analyses and vote counting based on direction of effect. Certainty of evidence was rated using GRADE approach. RESULTS Twenty reports were included, presenting unclear or low risk of bias. The evidence did not permit a high-confidence evaluation of the effectiveness of biotics, although five of the seven probiotic trials showed beneficial effects on faecal consistency. Synbiotics presented no clinically relevant effect in reducing antibiotics-associated vomiting, with very low certainty, in a meta-analysis including 32 adult cats. Probiotics significantly reduce the Bacillota/Actinomycetota ratio, with low certainty, in a meta-analysis involving 34 healthy young-adult cats. Following vote counting, probiotics improved immune profile in young cats, and increased butyric acid concentration in healthy cats. CLINICAL SIGNIFICANCE Current data highlight the need for further research, especially focused on at-risk groups and sick cats, before advocating the use of biotic supplementation.
Collapse
Affiliation(s)
- Á. López Martí
- Institute of Postgraduate StudiesUniversity of CordobaCordobaSpain
| | | | - H. López Martí
- Faculty of Medicine and NursingUniversity of CordobaCordobaSpain
| | - A. Ranchal Sánchez
- Department of Nursing, Pharmacology and Physiotherapy, Faculty of Medicine and NursingUniversity of CordobaCordobaSpain
| |
Collapse
|
44
|
Salvatore MM, Maione A, Buonanno A, Guida M, Andolfi A, Salvatore F, Galdiero E. Biological activities, biosynthetic capacity and metabolic interactions of lactic acid bacteria and yeast strains from traditional home-made kefir. Food Chem 2025; 470:142657. [PMID: 39756085 DOI: 10.1016/j.foodchem.2024.142657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Given the widespread industrial and domestic use of probiotic blends based on combinations of lactic acid bacteria (LAB) and yeasts to produce fermented foods or beverages that are supposed to provide health benefits, this study aimed to generate knowledge and concepts on biologically relevant activities, metabolism and metabolic interactions in yeast/LAB communities. For this, the postbiotic capabilities of three probiotic candidates, including two lactic acid bacteria (i.e., Lactococcus lactis subsp. hordniae and Lactococcus lactis subsp. lactis) and the yeast Pichia kudriavzevii, isolated from a traditional home-made kefir, were explored combining an assortment of bioassays with a GC-MS footprint metabolomic strategy. Cell-free supernatants from cultures showed antimicrobial/antioxidant activity and inhibited biofilm formation by Salmonella sp. Several bioactive secondary metabolites (including tyrosol, phenylethyl alcohol, 2,3-butanediol, erythritol, tryptophol, putrescine, cadaverine, 3-phenyllactate, 2-hydroxyisocaproate) were detected which may contribute to the odor and flavour of the fermented products and their effects on human body.
Collapse
Affiliation(s)
- Maria Michela Salvatore
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy.
| | - Angela Maione
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Annalisa Buonanno
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Marco Guida
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, (NA), Italy
| | - Anna Andolfi
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy; BAT Center-Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80055 Portici, (NA), Italy
| | - Francesco Salvatore
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Emilia Galdiero
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
45
|
Aran KR, Porel P, Hunjan G, Singh S, Gupta GD, Rohit. Postbiotics as a therapeutic tool in Alzheimer's disease: Insights into molecular pathways and neuroprotective effects. Ageing Res Rev 2025; 106:102685. [PMID: 39922231 DOI: 10.1016/j.arr.2025.102685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, characterized by oxidative stress, neuroinflammation, mitochondrial dysfunction, neurotransmitter imbalance, tau hyperphosphorylation, and amyloid beta (Aβ) accumulation in brain regions. The gut microbiota (GM) has a major impact on brain function due to its bidirectional interaction with the gut through the gut-brain axis. The gut dysbiosis has been associated with neurological disorders, emphasizing the importance of gut homeostasis in maintaining appropriate brain function. The changes in the composition of microbiomes influence neuroinflammation and Aβ accumulation by releasing pro-inflammatory cytokines, decreasing gut and blood-brain barrier (BBB) integrity, and microglial activation in the brain. Postbiotics, are bioactive compounds produced after fermentation, have been shown to provide several health benefits, particularly in terms of neuroinflammation and cognitive alterations associated with AD. Several research studies on animal models and human have successfully proven the effects of postbiotics on enhancing cognition and memory in experimental animals. This article explores the protective effects of postbiotics on cellular mechanisms responsible for AD pathogenesis and studies highlighting the influence of postbiotics as a total combination and specific compounds, including short-chain fatty acids (SCFAs). In addition, postbiotics act as a promising option for future research to deal with AD's progressive nature and improve an individual's life quality using microbiota modulation.
Collapse
Affiliation(s)
- Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India.
| | - Pratyush Porel
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Garry Hunjan
- Research Scholar, Department of Pharmacy Practice, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Rohit
- Research Scholar, Department of Pharmacy Practice, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
46
|
Chen L, Wang X, Wang S, Liu W, Song Z, Liao H. The impact of gut microbiota on the occurrence, treatment, and prognosis of ischemic stroke. Neurobiol Dis 2025; 207:106836. [PMID: 39952411 DOI: 10.1016/j.nbd.2025.106836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
Ischemic stroke (IS) is a cerebrovascular disease that predominantly affects middle-aged and elderly populations, exhibiting high mortality and disability rates. At present, the incidence of IS is increasing annually, with a notable trend towards younger affected individuals. Recent discoveries concerning the "gut-brain axis" have established a connection between the gut and the brain. Numerous studies have revealed that intestinal microbes play a crucial role in the onset, progression, and outcomes of IS. They are involved in the entire pathophysiological process of IS through mechanisms such as chronic inflammation, neural regulation, and metabolic processes. Although numerous studies have explored the relationship between IS and intestinal microbiota, comprehensive analyses of specific microbiota is relatively scarce. Therefore, this paper provides an overview of the typical changes in gut microbiota following IS and investigates the role of specific microorganisms in this context. Additionally, it presents a comprehensive analysis of post-stroke microbiological therapy and the relationship between IS and diet. The aim is to identify potential microbial targets for therapeutic intervention, as well as to highlight the benefits of microbiological therapies and the significance of dietary management. Overall, this paper seeks to provide key strategies for the treatment and management of IS, advocating for healthy diets and health programs for individuals. Meanwhile, it may offer a new perspective on the future interdisciplinary development of neurology, microbiology and nutrition.
Collapse
Affiliation(s)
- Liying Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shiqi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Weili Liu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Huiling Liao
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
47
|
Chen Y, Zhu F, Yu G, Peng N, Li X, Ge M, Yang L, Dong W. Bifidobacterium bifidum postbiotics prevent Salmonella Pullorum infection in chickens by modulating pyroptosis and enhancing gut health. Poult Sci 2025; 104:104968. [PMID: 40043668 PMCID: PMC11927735 DOI: 10.1016/j.psj.2025.104968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/24/2025] Open
Abstract
The overuse of antibiotics in poultry farming has led to the emergence of multidrug-resistant pathogens, posing severe threats to animal health and public safety. Salmonella Pullorum (S. Pullorum), a host-specific pathogen targeting poultry, causes high mortality in chicks and disrupts intestinal health. This study evaluated the protective effects of Bifidobacterium bifidum postbiotics (BbP) against S. Pullorum infection, focusing on their mechanisms in regulating pyroptosis, restoring intestinal barrier function, and modulating gut microbiota. Both in vivo (chickens challenged with S. Pullorum) and in vitro (chicken small intestinal epithelial cells, CSIEC) models were used to assess the effects of BbP and its components (bacterial lysates or metabolites). Results showed that BbP significantly improved growth performance in infected chickens, reducing mortality from 66.66 % to 8.33 %. BbP effectively suppressed the expression of pyroptosis-related proteins, including apoptosis-associated speck-like protein containing a CARD (ASC), Caspase-1 (cysteine-aspartic acid protease-1), and Gasdermin D N-terminal (GSDMD-N), and reduced inflammatory cytokines, including interleukin-1β (IL-1β) and interleukin-8 (IL-8), while increasing anti-inflammatory cytokines, such as interleukin-10 (IL-10) and interleukin-4 (IL-4), thereby mitigating inflammation. Furthermore, BbP restored intestinal barrier function by upregulating the expression of tight junction proteins, including zonula occludens-1 (ZO-1), Occludin, and Claudin-1. The cecal microbiota diversity was also improved by BbP, with a decrease in the abundance of harmful bacteria (e.g., Escherichia-Shigella) and an enrichment of beneficial bacteria (e.g., Lactobacillus and Ruminococcus). These findings demonstrate that BbP provides significant protection against S. Pullorum infection by modulating pyroptosis, protecting the intestinal barrier, and restoring microbial balance. As an effective antibiotic alternative, BbP shows promise for the prevention and control of S. Pullorum infections in poultry farming.
Collapse
Affiliation(s)
- Yuhao Chen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Fuqiang Zhu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Guobi Yu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Nana Peng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Xinying Li
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Meng Ge
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Lei Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China
| | - Wei Dong
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410125, China.
| |
Collapse
|
48
|
Zhang Y, Jiang Y, Zhao J, Mo Q, Wang C, Wang D, Li M. Weizmannia coagulans Extracellular Proteins Reduce Skin Acne by Inhibiting Pathogenic Bacteria and Regulating TLR2/TRAF6-Mediated NF-κB and MAPKs Signaling Pathways. Probiotics Antimicrob Proteins 2025; 17:705-720. [PMID: 37870674 DOI: 10.1007/s12602-023-10175-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
As a probiotic, Weizmannia coagulans (W. coagulans) is often used in food and medicine to regulate intestinal flora and exert anti-inflammatory effects. In this study, the anti-acne efficacy and mechanism of extracellular proteins (YTCY-EPs) from W. coagulans YTCY strain are analyzed. The main components of YTCY-EPs, extracted and separated from the fermentation broth, are peptides ranging from 1.51 to 11.44 kDa, accounting for about 80%. Among the peptides identified by LC/MS-MS, YTCY_A-F possess the properties of antimicrobial peptides, while YTCY_1-4 possess antioxidative properties. These peptides have a strong effect on Cutibacterium acnes (C. acnes) and significantly inhibit Staphylococcus aureus. The inhibition rate of biofilm adhesion of YT-EPs to C. acnes reached 50% under the MIC. It was found that YTCY-EPs possess strong antioxidant and anti-inflammatory properties. It can effectively reduce active oxygen nearly 3 times and can reduce the downstream TLR2/NF-κB and MAPKs/AP-1 pathways by regulating the nuclear translocation of NF-κB and AP-1 in vitro. The transcriptional expression of inflammatory cytokines, inflammatory chemokines, and matrix metalloproteinase genes is also regulated, thereby slowing the recruitment of inflammatory cells and the development of inflammation, and increasing keratinocyte mobility. In addition, the expression levels of inflammatory factors and matrix metalloproteinases in the rabbit ears with acne problems that were tested with YTCY-EPs were significantly reduced, and it was obviously observed that the rabbit ear inflammation, acne, and keratinization problems were repaired. The results of this study prove that YTCY-EPs can be used as a potential anti-acne raw material in cosmetics.
Collapse
Affiliation(s)
- Yongtao Zhang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Yanbing Jiang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Jingsha Zhao
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Qiuting Mo
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Changtao Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Dongdong Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China
| | - Meng Li
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, 100048, China.
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, China.
| |
Collapse
|
49
|
Li J, Wang Y, Wu S, Zhou Z, Jia W, Shen X, Li Y, He F, Cheng R. Postbiotics Made From Selected Lactic Acid Bacteria Improves Chronic Restraint Stress-Induced Anhedonia and Sleep Disorders. Mol Nutr Food Res 2025; 69:e70005. [PMID: 40045653 DOI: 10.1002/mnfr.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/23/2024] [Accepted: 02/05/2025] [Indexed: 04/25/2025]
Abstract
Sleep disorders have become one of the most prevalent neuropsychiatric disorders in recent years. This study aimed to investigate the effects of postbiotics derived from selected lactic acid bacteria on anhedonia and sleep disorders in chronic restraint stress (CRS)-induced mice, as well as their potential mechanisms. Mice were orally administered normal saline, low, medium, or high doses of postbiotics for 30 days, with CRS applied from days 1 to 21. The medium dose of postbiotics significantly increased the sucrose preference index, and the high dose of postbiotics significantly increased sleep duration. Postbiotic treatment effectively restored the diversity and composition of the gut microbiota to levels comparable to those observed in the vehicle (Veh) group. Furthermore, low and medium doses of postbiotics significantly reduced serum corticosterone levels, and medium and high doses significantly reduced serum IL-1β levels. Additionally, postbiotics administration significantly increased glutamate and GABA levels in both the prefrontal cortex and hypothalamus, as well as GABA levels in the feces. These results indicate that postbiotics alleviate CRS-induced anhedonia and sleep disorders in a dose-dependent manner. This effect may be mediated through the restoration of homeostasis in the MGB axis, HPA axis, inflammation pathways, and neurotransmitter balance.
Collapse
Affiliation(s)
- Jinxing Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yimei Wang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Simou Wu
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhimo Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Wen Jia
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Xi Shen
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yun Li
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Fang He
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Ruyue Cheng
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
50
|
Kim S, Ndwandwe C, Devotta H, Kareem L, Yao L, O'Mahony L. Role of the microbiome in regulation of the immune system. Allergol Int 2025; 74:187-196. [PMID: 39955207 DOI: 10.1016/j.alit.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 02/17/2025] Open
Abstract
Immune health and metabolic functions are intimately connected via diet and the microbiota. Immune cells are continuously exposed to a wide range of microbes and microbial-derived compounds, with important mucosal and systemic ramifications. Microbial fermentation of dietary components in vivo generates thousands of molecules, some of which are integral components of the molecular circuitry that regulates immune and metabolic functions. These in turn protect against aberrant inflammatory or hyper-reactive processes and promote effector immune responses that quickly eliminate pathogens, such as SARS-CoV-2. Potent tolerance mechanisms should ensure that these immune cells do not over-react to non-pathogenic factors (e.g. food proteins), while maintaining the ability to respond to infectious challenges in a robust, effective and well controlled manner. In this review we examine the factors and mechanisms that shape microbiota composition and interactions with the host immune system, their associations with immune mediated disorders and strategies for intervention.
Collapse
Affiliation(s)
- Songhui Kim
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Cebile Ndwandwe
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah Devotta
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Lamiah Kareem
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Lu Yao
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Liam O'Mahony
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|