1
|
Hamza M, Wang S, Liu Y, Li K, Zhu M, Chen L. Unraveling the potential of bioengineered microbiome-based strategies to enhance cancer immunotherapy. Microbiol Res 2025; 296:128156. [PMID: 40158322 DOI: 10.1016/j.micres.2025.128156] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
The human microbiome plays a pivotal role in the field of cancer immunotherapy. The microbial communities that inhabit the gastrointestinal tract, as well as the bacterial populations within tumors, have been identified as key modulators of therapeutic outcomes, affecting immune responses and reprogramming the tumor microenvironment. Advances in synthetic biology have made it possible to reprogram and engineer these microorganisms to improve antitumor activity, enhance T-cell function, and enable targeted delivery of therapies to neoplasms. This review discusses the role of the microbiome in modulating both innate and adaptive immune mechanisms-ranging from the initiation of cytokine production and antigen presentation to the regulation of immune checkpoints-and discusses how these mechanisms improve the efficacy of immune checkpoint inhibitors. We highlight significant advances with bioengineered strains like Escherichia coli Nissle 1917, Lactococcus lactis, Bifidobacterium, and Bacteroides, which have shown promising antitumor efficacy in preclinical models. These engineered microorganisms not only efficiently colonize tumor tissues but also help overcome resistance to standard therapies by reprogramming the local immune environment. Nevertheless, several challenges remain, such as the requirement for genetic stability, effective tumor colonization, and the control of potential safety issues. In the future, the ongoing development of genetic engineering tools and the optimization of bacterial delivery systems are crucial for the translation of microbiome-based therapies into the clinic. This review highlights the potential of bioengineered microbiota as an innovative, personalized approach in cancer immunotherapy, bringing hope for more effective and personalized treatment options for patients with advanced malignancies.
Collapse
Affiliation(s)
- Muhammad Hamza
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Yike Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Kun Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Motao Zhu
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Lin Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
2
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [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: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
3
|
Jeyaraman N, Jeyaraman M, Dhanpal P, Ramasubramanian S, Nallakumarasamy A, Muthu S, Santos GS, da Fonseca LF, Lana JF. Integrative review of the gut microbiome’s role in pain management for orthopaedic conditions. World J Exp Med 2025; 15:102969. [DOI: 10.5493/wjem.v15.i2.102969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/03/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
The gut microbiome, a complex ecosystem of microorganisms, has a significant role in modulating pain, particularly within orthopaedic conditions. Its impact on immune and neurological functions is underscored by the gut-brain axis, which influences inflammation, pain perception, and systemic immune responses. This integrative review examines current research on how gut dysbiosis is associated with various pain pathways, notably nociceptive and neuroinflammatory mechanisms linked to central sensitization. We highlight advancements in meta-omics technologies, such as metagenomics and metaproteomics, which deepen our understanding of microbiome-host interactions and their implications in pain. Recent studies emphasize that gut-derived short-chain fatty acids and microbial metabolites play roles in modulating neuroinflammation and nociception, contributing to pain management. Probiotics, prebiotics, synbiotics, and faecal microbiome transplants are explored as potential therapeutic strategies to alleviate pain through gut microbiome modulation, offering an adjunct or alternative to opioids. However, variability in individual microbiomes poses challenges to standardizing these treatments, necessitating further rigorous clinical trials. A multidisciplinary approach combining microbiology, immunology, neurology, and orthopaedics is essential to develop innovative, personalized pain management strategies rooted in gut health, with potential to transform orthopaedic pain care.
Collapse
Affiliation(s)
- Naveen Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
| | - Madhan Jeyaraman
- Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Priya Dhanpal
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Swaminathan Ramasubramanian
- Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
| | - Arulkumar Nallakumarasamy
- Department of Orthopaedics, Jawaharlal Institute of Postgraduate Medical Education and Research–Karaikal, Puducherry 609602, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Meenakshi Academy of Higher Education and Research, Chennai 631552, Tamil Nadu, India
| | - Gabriel Silva Santos
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - Lucas Furtado da Fonseca
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| | - José Fábio Lana
- Department of Orthopaedics, Brazilian Institute of Regenerative Medicine, Indaiatuba 13334-170, São Paulo, Brazil
| |
Collapse
|
4
|
Liu Y, Huang X, Jia X, Huang J, Cao R, Yu F, Xue K, Hui H, Lu J. Biosynthesis of Melanin with Engineered Probiotics for Oral Treatment of Ulcerative Colitis. ACS NANO 2025; 19:21333-21347. [PMID: 40464444 DOI: 10.1021/acsnano.4c17942] [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: 06/18/2025]
Abstract
Ulcerative colitis (UC) is a chronic intestinal inflammation characterized by immune overactivity and gut microbiota imbalance, leading to oxidative stress and inflammation. New therapeutics are required because existing ones are frequently unsuccessful and have long-term adverse effects. The research aims to manage oxidative stress and restore gut microbiota balance. The benefits of probiotics for UC can be compromised by gastrointestinal conditions that interfere with their adhesion and activity. Coating methods enhance bacterial survival in the gastrointestinal environment but face challenges like instability at low pH, short-lived effects, complexity, layer interactions, and biosafety issues. Melanin-like nanozymes are stable in the gastrointestinal environment and effectively scavenge reactive oxygen species, specifically targeting colitis lesions. We developed biosynthetic melanin-producing engineered bacteria (EcN-Mel) derived from genetically modified Nissle 1917 Escherichia coli expressing tyrosinase genes. This study evaluated the feasibility and effectiveness of administering EcN-Mel orally in UC mouse models. Results showed that EcN-Mel produced and secreted melanin, exhibiting targeted intestinal adhesion, a free radical scavenging ability, and gastrointestinal stability. In vivo imaging revealed increased colonization efficiency and retention time of EcN-Mel in inflamed intestinal segments. EcN-Mel enhances beneficial bacteria of the Lactobacillus genus while decreasing harmful members of the Proteobacteria genus, promoting gut microbiota homeostasis, and alleviating colitis. EcN-Mel alleviated intestinal mucosal damage through combined actions, including gut microbiota modulation, oxidative stress reversal, cytokine regulation, and barrier restoration. Our findings confirm the safety, feasibility, and effectiveness of EcN-Mel for UC treatment.
Collapse
Affiliation(s)
- Yu Liu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Xiazi Huang
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaohua Jia
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Department of Ultrasound, Shuozhou Grand Hospital of Shanxi Medical University, Shuozhou 036000, China
| | - Jing Huang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Ruoyao Cao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Fan Yu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Kaizhong Xue
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| | - Hui Hui
- Key Laboratory of Molecular Imaging of Chinese Academy of Sciences, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- National Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing 100053, China
| |
Collapse
|
5
|
Hijová E, Bertková I, Štofilová J. Incorporating Postbiotics into Intervention for Managing Obesity. Int J Mol Sci 2025; 26:5362. [PMID: 40508171 PMCID: PMC12154039 DOI: 10.3390/ijms26115362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/20/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
Obesity is reaching global epidemic proportions worldwide, posing a significant burden on individual health and society. Altered gut microbiota is considered a key factor in the pathogenesis of many diseases, producing metabolites that contribute to the health-beneficial properties of postbiotics. Postbiotics, bioactive microbial components derived from probiotics, are emerging as a valuable strategy in modern medicine and a promising alternative for managing obesity without the need for live bacteria. This work provides a comprehensive overview of the potential health benefits of postbiotics, particularly in relation to obesity, which represents an important health challenge. Despite the encouraging insights into the health benefits of postbiotics, we highlight the need for further research to clarify the mechanisms and the specific roles of different postbiotic components. Integrating postbiotics into health interventions has the potential to enhance preventive care and significantly improve health outcomes in at-risk populations.
Collapse
Affiliation(s)
- Emília Hijová
- Center of Clinical and Preclinical Research-MediPark, Faculty of Medicine, Pavol Jozef Šafárik University, SNP 1, 040 11 Košice, Slovakia; (I.B.); (J.Š.)
| | | | | |
Collapse
|
6
|
Gao Y, Wang P, Huang S, Zheng Q, Wu S, Huang J, Chen Z, Gao B. Antibacterial reuterin as a multifunctional crosslinker for constructing chitosan-based hydrogels to promote infected wound healing. Int J Biol Macromol 2025; 316:144494. [PMID: 40419054 DOI: 10.1016/j.ijbiomac.2025.144494] [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: 02/11/2025] [Revised: 05/10/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025]
Abstract
Compared to traditional antibiotics, which often contribute to antibiotic resistance, probiotics and their metabolites have garnered increasing attention for their potential to improve the wound microenvironment. Among these, reuterin, a metabolite produced by Limosilactobacillus reuteri, exhibits notable antioxidant and antimicrobial activities, making it particularly advantageous for medical applications. Inspired by the structural similarity of reuterin to conventional chemical crosslinkers, we explored its dual role for the first time-as both a bioactive component and a crosslinking agent-by combining it with chitosan to develop a novel therapeutic hydrogel, eliminating the need for additional drug loading. At an optimal reuterin-to-chitosan molar ratio of 1:2, the hydrogel demonstrated outstanding performance. Cytotoxicity assays confirmed its safety, with a hemolysis rate below 2 %, indicating excellent biocompatibility. Remarkably, the hydrogel achieved an impressive 99 % antibacterial efficacy and over 60 % DPPH radical scavenging capacity in vitro. In a rat model of infected wounds, the hydrogel exhibited acid-responsive release of reuterin, effectively suppressing bacterial proliferation. These combined effects significantly accelerated wound healing, demonstrating an approximately 10 % improvement in healing rates compared to commercial controls. In conclusion, the reuterin-crosslinked chitosan hydrogel represents a promising, safer and more effective alternative to traditional chemical crosslinked hydrogels for clinical wound management.
Collapse
Affiliation(s)
- Yuwei Gao
- Department of Prosthodontics, Hospital of Stomatology Hebei Medical University, Hebei Key Laboratory of Stomatology, Hebei Clinical Medical Research Centre for Oral Diseases, Hebei Medical University, Shijiazhuang, China; National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Panpan Wang
- Department of periodontology, Guanghua School and Hospital of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shunfen Huang
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China
| | - Qizhe Zheng
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shipeng Wu
- Department of Stomatology, Shanghai Eighth People's Hospital, Shanghai 200235, China
| | - Jun Huang
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.
| | - Zhiyu Chen
- Department of Prosthodontics, Hospital of Stomatology Hebei Medical University, Hebei Key Laboratory of Stomatology, Hebei Clinical Medical Research Centre for Oral Diseases, Hebei Medical University, Shijiazhuang, China.
| | - Botao Gao
- National Engineering Research Center for Healthcare Devices, Guangdong Key Lab of Medical Electronic Instruments and Materials, Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.
| |
Collapse
|
7
|
Chen Y, Lei Z, Gu D, Zhao H, Yu R, He Q, Xu M, Du H. Gut microbiota: A potential enhancing factor for the therapeutic efficacy of bioactive compounds in herbal medicines. Fitoterapia 2025; 183:106570. [PMID: 40288589 DOI: 10.1016/j.fitote.2025.106570] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
The therapeutic efficacy of herbal medicines (HMs) is often compromised by the low bioavailability of their bioactive compounds. However, emerging evidence highlights the crucial role of gut microbiota in enhancing their effectiveness. This review summarizes gut microbiota-mediated metabolism of key herbal components, including terpenoids, flavonoids, alkaloids, and quinones. Particular emphasis is placed on the diverse gut microbiota, enzymes, and metabolites that participate in the biotransformation pathways of these active components of HMs. Exploring the metabolism between the gut microbiota and bioactive compounds gives a better understanding of HMs with multiple components against multiple targets, complex mechanisms of action, and diverse physiological activities. This review underscores the critical importance of gut microbiota in modulating and potentially enhancing the pharmacological effects of HMs, which offers new insights into gut microbiota-mediated transformation pathways and molecular mechanisms of bioactive compounds and deepens the understanding of the therapeutic effects of HMs. Moreover, it suggests new research directions for studying HMs based on gut microbiota-mediated biotransformation.
Collapse
Affiliation(s)
- Yu Chen
- Department of Otolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ziqin Lei
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu 610014, Sichuan, China
| | - Deying Gu
- Department of Otolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Huiling Zhao
- Department of Otolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rong Yu
- Department of Otolaryngology Head and Neck Surgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qin He
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu 610014, Sichuan, China
| | - Min Xu
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu 610014, Sichuan, China
| | - Huan Du
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu 610014, Sichuan, China.
| |
Collapse
|
8
|
Huerga López C, Sánchez Martín MJ, Herráez Moreta A, Calvo Urrutia M, Cristóbal García I, Díaz Morillo C, Blanco-Rojo R, Sáez ME, Olivares M, Arroyo R, Herranz C, Alba C, Rodríguez JM, Fernández L. Ligilactobacillus salivarius CECT5713 Increases Term Pregnancies in Women with Infertility of Unknown Origin: A Randomized, Triple-Blind, Placebo-Controlled Trial. Nutrients 2025; 17:1860. [PMID: 40507130 PMCID: PMC12158033 DOI: 10.3390/nu17111860] [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: 04/29/2025] [Revised: 05/21/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Background/Objectives: Unexplained infertility is a worldwide problem affecting a significant proportion of couples of reproductive age. Recent studies suggest that alterations in the vaginal microbiota are related to female infertility, while supplementation with some probiotic strains has been shown to improve pregnancy rates in couples experiencing this problem. This study aimed to evaluate the impact of oral administration of Ligilactobacillus salivarius CECT5713 on pregnancy success rates in couples with unexplained infertility prior to in vitro fertilization (IVF). Methods: Seventy couples were randomized to receive either a placebo or a probiotic intervention (one capsule per day containing an excipient only or 3 × 109 viable cells of L. salivarius CECT5713 plus an excipient, respectively); 57 couples completed the study. Baseline data on demographics, health status (including gynecological and reproductive history), and lifestyle habits were collected. Vaginal swabs and semen samples were obtained from each couple before the intervention and immediately prior to IVF or upon confirmed pregnancy and were analyzed for microbiological (using both culture-dependent and -independent methods) and immunological profiles. Results: Oral administration of L. salivarius CECT5713 in couples with unexplained infertility scheduled for IVF resulted in a significantly higher pregnancy success rate (48.1%) compared to the placebo group (20.0%) (one-tailed Chi-square test; p < 0.024). The probiotic intervention improved both vaginal and semen immunological profiles, with no substantial changes observed in their microbial composition. Conclusions: These preliminary findings support the potential of L. salivarius CECT5713 supplementation to enhance fertility outcomes in couples with unexplained infertility.
Collapse
Affiliation(s)
- Cristina Huerga López
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (C.H.L.); (M.J.S.M.); (A.H.M.); (M.C.U.); (I.C.G.)
| | - María J. Sánchez Martín
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (C.H.L.); (M.J.S.M.); (A.H.M.); (M.C.U.); (I.C.G.)
| | - Aránzazu Herráez Moreta
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (C.H.L.); (M.J.S.M.); (A.H.M.); (M.C.U.); (I.C.G.)
| | - Marta Calvo Urrutia
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (C.H.L.); (M.J.S.M.); (A.H.M.); (M.C.U.); (I.C.G.)
| | - Ignacio Cristóbal García
- Hospital Clínico San Carlos, 28040 Madrid, Spain; (C.H.L.); (M.J.S.M.); (A.H.M.); (M.C.U.); (I.C.G.)
| | - Cristina Díaz Morillo
- Kerry Group, Camino de Purchil 66, 18004 Granada, Spain; (C.D.M.); (R.B.-R.); (M.E.S.); (M.O.)
| | - Ruth Blanco-Rojo
- Kerry Group, Camino de Purchil 66, 18004 Granada, Spain; (C.D.M.); (R.B.-R.); (M.E.S.); (M.O.)
| | - María E. Sáez
- Kerry Group, Camino de Purchil 66, 18004 Granada, Spain; (C.D.M.); (R.B.-R.); (M.E.S.); (M.O.)
| | - Mónica Olivares
- Kerry Group, Camino de Purchil 66, 18004 Granada, Spain; (C.D.M.); (R.B.-R.); (M.E.S.); (M.O.)
| | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (R.A.); (C.H.); (C.A.); (J.M.R.)
| | - Carmen Herranz
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (R.A.); (C.H.); (C.A.); (J.M.R.)
| | - Claudio Alba
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (R.A.); (C.H.); (C.A.); (J.M.R.)
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain; (R.A.); (C.H.); (C.A.); (J.M.R.)
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain
| | - Leonides Fernández
- Instituto Pluridisciplinar, Complutense University of Madrid, 28040 Madrid, Spain
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
9
|
Gupta MK, Srivastava R. Gut Microbiome Interventions: From Dysbiosis to Next-Generation Probiotics (NGPs) for Disease Management. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10582-7. [PMID: 40434505 DOI: 10.1007/s12602-025-10582-7] [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: 05/06/2025] [Indexed: 05/29/2025]
Abstract
The gut microbiome, sometimes referred to as the "second brain," the "lost organ," the "identification card of the individual," and the "fingerprint of the host," possesses diverse traits and functions that influence health. The impact of gut commensal bacteria on health, as opposed to environmental pathogenic factors, has generated increasing interest in recent years, culminating in a substantial body of study. Research indicates that dysbiosis of the intestinal microbiota is commonly observed in chronic inflammatory diseases, including colitis, obesity/metabolic syndrome, diabetes mellitus, liver infections, allergic conditions, cardiovascular diseases, COVID-19, cancers, and neurodegenerative disorders. The International Scientific Association for Probiotics and Prebiotics has recently refined the theory of complementary and synergistic synbiotics. In recent years, the field of microbiome research has been significantly advanced by technological developments such as massive culturomics, gnotobiotics, metabolomics, parallel DNA sequencing, and RNA sequencing. This review article examined the potential next generation probiotics (NGPs) and explored some of them, Faecalibacterium prausnitzii, Bacteroides thetaiotaomicron, Akkermansia muciniphila, Parabacteroides goldsteinii, Bacteroides fragilis, Eubacterium hallii, Roseburia intestinalis, Christensenella minuta, Prevotella copri, and Oscillospira guilliermondii. In addition to these useful probiotic strains, psychobiotics, members of the families of Lactobacilli, Streptococci, Bifidobacteria, Escherichia, and Enterococci, have extended applicability in the use for neurodevelopmental and neurodegenerative disorders. The article also reviewed current trends and limitations in NGPs to enhance our comprehensive understanding of key concepts associated with the consumption of probiotics and proposed necessary initiatives for researchers to engage in collaborative translational research as future therapeutic solutions.
Collapse
Affiliation(s)
- Mandeep Kumar Gupta
- Moradabad Educational Trust Group of Institutions Faculty of Pharmacy, Moradabad, 244001, Uttar Pradesh, India.
| | - Rajnish Srivastava
- Chitkara University School of Pharmacy, Chitkara University, Baddi, 174103, Himachal Pradesh, India
| |
Collapse
|
10
|
Iraporda C, Rubel IA, Bengoa AA, Manrique GD, Garrote GL, Abraham AG. Lactic Acid Bacteria Strains Isolated from Jerusalem Artichoke (Helianthus tuberosus L.) Tubers as Potential Probiotic Candidates. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10594-3. [PMID: 40411633 DOI: 10.1007/s12602-025-10594-3] [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] [Accepted: 05/12/2025] [Indexed: 05/26/2025]
Abstract
The search for probiotic candidates is an area that accompanies the world trend of development of novel probiotic strains and new products. In recent years, unconventional sources of potential probiotic bacteria have been studied. Furthermore, there has been an increasing interest in non-dairy and plant-based probiotic foods, currently being considered as a priority for the food industry. The aim of this study was to evaluate the probiotic potential of lactic acid bacteria (LAB) isolated from Jerusalem artichoke tubers. These strains were characterized by in vitro tests for their biochemical and probiotic properties and safety aspects. The results demonstrated that the LAB strains isolated exhibited a survival rate after acid exposure exceeding 90%, maintained viability above 88% under simulated gastrointestinal conditions, and the autoaggregation capacity ranged from 61 to 81%. Additionally, the strains showed no hemolytic activity and were sensitive to antibiotics. The isolates also downregulated the proinflammatory responses and showed antimicrobial activity against E. coli and B. cereus. The bacilli strains showed a high similarity with Lentilactobacillus kosonis and Lentilactobacillus curieae. Hence, these strains revealed potential probiotic in vitro characteristics that position them to be used in plant-based functional food. This strategic exploration of probiotic bacteria sourced from vegetables not only enhances the diversity of available strains-both taxonomically and functionally-but also fosters the development of sustainable, plant-based probiotic applications.
Collapse
Affiliation(s)
- Carolina Iraporda
- Facultad de Ingeniería, Núcleo Tecnología de Semillas y Alimentos, Universidad Nacional del Centro de La Provincia de Buenos Aires, Av. Del Valle 5737, Olavarría, Provincia de Buenos Aires, 7400, Argentina.
| | - Irene A Rubel
- Facultad de Ingeniería, Núcleo Tecnología de Semillas y Alimentos, Universidad Nacional del Centro de La Provincia de Buenos Aires, Av. Del Valle 5737, Olavarría, Provincia de Buenos Aires, 7400, Argentina
| | - Ana A Bengoa
- Centro de Investigación y Desarrollo en Ciencia y Tecnología de Alimentos (CIDCA, UNLP-CIC-CONICET), Calle 47 y 116, La Plata, Provincia de Buenos Aires, 1900, Argentina
| | - Guillermo D Manrique
- Facultad de Ingeniería, Núcleo Tecnología de Semillas y Alimentos, Universidad Nacional del Centro de La Provincia de Buenos Aires, Av. Del Valle 5737, Olavarría, Provincia de Buenos Aires, 7400, Argentina
| | - Graciela L Garrote
- Centro de Investigación y Desarrollo en Ciencia y Tecnología de Alimentos (CIDCA, UNLP-CIC-CONICET), Calle 47 y 116, La Plata, Provincia de Buenos Aires, 1900, Argentina
| | - Analía G Abraham
- Centro de Investigación y Desarrollo en Ciencia y Tecnología de Alimentos (CIDCA, UNLP-CIC-CONICET), Calle 47 y 116, La Plata, Provincia de Buenos Aires, 1900, Argentina
- Facultad de Ciencias Exactas, Área Bioquímica y Control de los Alimentos, Universidad Nacional de La Plata, Calle 47 y 115, La Plata, Provincia de Buenos Aires, 1900, Argentina
| |
Collapse
|
11
|
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
|
12
|
Hanna A, Abbas H, Yassine F, AlBush A, Bilen M. Systematic review of gut microbiota composition, metabolic alterations, and the effects of treatments on PCOS and gut microbiota across human and animal studies. Front Microbiol 2025; 16:1549499. [PMID: 40438215 PMCID: PMC12116390 DOI: 10.3389/fmicb.2025.1549499] [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: 12/21/2024] [Accepted: 04/15/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Polycystic ovary syndrome (PCOS) is an endocrine disorder affecting around 12% of women globally, associated with infertility and various comorbidities. Emerging evidence suggests a crucial role of gut microbiota in PCOS pathophysiology, prompting research to investigate alterations in gut microbial composition in patients with PCOS. Methods This systematic review aims to analyze human and animal studies that compare gut microbiota composition, gut-derived metabolites, and treatment interventions in PCOS patients versus healthy controls. A comprehensive literature search was conducted using PubMed, Scopus, and Web of Science, yielding studies examining gut microbiota, metabolomic shifts, and treatment responses in PCOS models and human populations. Results Our analysis revealed decreases in alpha diversity in PCOS patients, with more pronounced changes in beta diversity in animal models. Specific bacterial taxa, such as Bacteroides vulgatus, Escherichia-Shigella and Lactobacillus, showed implication in PCOS pathogenesis, suggesting potential microbial markers. Furthermore, discrepancies between human and animal studies show the need for humanized mouse models to bridge this gap. Interventions like probiotics and fecal microbiota transplantation (FMT) showed varying levels of efficacy, with FMT emerging as a more promising but invasive option, offering live bacteriotherapy as a potential therapeutic alternative. Alterations in gut-derived metabolites, including short-chain fatty acids and bile acids, highlighted the multifaceted nature of PCOS, with implications extending to metabolic, hormonal, and gut-brain axis disruptions. Discussion In conclusion, PCOS exhibits complex interactions between gut microbiota and metabolic pathways, necessitating further research with standardized methods and larger sample sizes to elucidate the microbiome's role in PCOS.
Collapse
Affiliation(s)
- Aya Hanna
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Abbas
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fayez Yassine
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Alia AlBush
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Melhem Bilen
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Centre for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
- World Health Organization (WHO) Collaborating Centre for Reference and Research on Bacterial Pathogens, Beirut, Lebanon
| |
Collapse
|
13
|
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
|
14
|
Wang Y, Tan W, Li X, Yang G, Wang Y, Liao J, Lu A, Zhang G, Chen K, Yang L, Li W. A pharmacovigilance study on probiotic preparations based on the FDA Adverse Event Reporting System from 2005 to 2023. Front Cell Infect Microbiol 2025; 15:1455735. [PMID: 40433664 PMCID: PMC12106448 DOI: 10.3389/fcimb.2025.1455735] [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: 06/27/2024] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background Probiotics are recognized as beneficial foods, but adverse reactions reported by individuals still exist. This study aims to analysis adverse events (AE) related to probiotics from the FAERS database from the first quarter (Q1) of 2005 to the fourth quarter (Q4) of 2023. Methods The AE data related to probiotic from the 2005 Q1 to the 2023 Q4 were collected. R language was applied to analyze the standardized AE data and three algorithms including the reporting odds ratio (ROR), the proportional reporting ratio (PRR) and the empirical Bayes geometric mean (EBGM) were used to identify AE signals. Results In this study, 10,698,312 reports were collected from the FAERS database, of which 74 probiotic-related adverse events were reported. About one third of the reported cases were older than 60 years.36.36% of the reported cases required Hospitalization. A total of 285 preference terms (PTS) and 15 system organ classes (SOC) were identified. In the overall analysis, only 9 PTs and 2 SOCs met significant disproportionality for all three algorithms simultaneously. SOCs included Gastrointestinal disorders (N=97, ROR=5.3, PRR=3.84, EBGM=3.84) and Hepatobiliary disorders (N=9, ROR =3.39, PRR=3.32, EBGM=3.32). PTs included Gastrointestinal pain (ROR=77.76, PRR=76.69, EBGM=76.63), Hypophagia (ROR=24.13, PRR=23.88, EBGM=28.88), and Hepatobiliary disorders (N=97, ROR=5.3, PRR=3.84, EBGM=3.84) and Flatulence (ROR=23.75, PRR=23.28, EBGM=23.27) were the top four highest. Meanwhile, s found new unique adverse signals such as Agitation (ROR=12.48, PRR=12.32, EBGM=12.32) and Anxiety (ROR=4.10, PRR=4.04, EBGM=4.04). Additionally, subgroup analyses were performed to identify AE signals based on gender and age. Metabolism and nutrition disorders (N=6, ROR=3.21, PRR=3.04, EBGM=3.04) and Asthenia (N=3, ROR=5.9, PRR=5.71, EBGM=5.71) were unique AE signal for the male group. Conclusion Although, the risk of adverse reactions arising from the application of probiotics cannot be ignored. However, However, the results of this FAERS-based study continue to support the overall safety of probiotic preparations. It is necessary to pay attention to the potential influence of factors such as gender and age on the effects and adverse reactions of probiotic application in basic research and clinical application.
Collapse
Affiliation(s)
- Yitong Wang
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Weifu Tan
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Xuyang Li
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Department of Pediatric, The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guangli Yang
- Department of Central Laboratory, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Dongguan Key Laboratory of Prevention and Treatment of Critical Illness, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Yunxiao Wang
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
- Department of Pediatric, The First Clinical Medical College of Jinan University, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing Liao
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Aner Lu
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Guoqing Zhang
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Kuidai Chen
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Liling Yang
- Dongguan Key Laboratory of Prevention and Treatment of Critical Illness, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Wei Li
- Department of Neonatology, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| |
Collapse
|
15
|
Jin X, Zheng Q, Nguyen TTM, Yi GS, Yang SJ, Yi TH. Assessment of Antimicrobial Activity and Safety of Pediococcus pentosaceus Isolated from Ginseng as a Functional Cosmetic Ingredient. Microorganisms 2025; 13:1093. [PMID: 40431265 PMCID: PMC12114239 DOI: 10.3390/microorganisms13051093] [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: 04/15/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Lactic acid bacteria (LAB) are gaining increasing attention as functional ingredients in the cosmetic industry, particularly those derived from natural plant sources. Although various LAB strains have been widely applied in cosmetic formulations, studies investigating the effects of naturally derived LAB on the skin remain limited. In this study, we isolated an LAB strain from ginseng and evaluated its potential as a functional cosmetic ingredient. The antimicrobial activity of the strain was assessed against skin-associated pathogens Staphylococcus aureus and Staphylococcus epidermidis, while cytotoxicity was evaluated using HaCaT and Caco-2 cells. Considering the limitations of vertebrate animal testing, infection and survival assays were conducted using Galleria mellonella larvae as an alternative in vivo model. The ginseng-derived strain exhibited 99.93% similarity to Pediococcus pentosaceus and was designated P. pentosaceus THG-219. It exhibited an MIC of 0.625 mg/mL and 1.25 mg/mL against S. aureus KCTC 3881 and S. epidermidis KCTC 1917, respectively. Its antimicrobial activity was further enhanced following ethyl acetate fractionation. P. pentosaceus THG-219 showed no toxicity in G. mellonella larvae and exerted antibacterial effects in this model. No cytotoxicity was observed in HaCaT and Caco-2 cells. Furthermore, P. pentosaceus THG-219 promoted host cell adhesion while inhibiting pathogen adhesion. It also exhibited excellent acid, bile, and heat tolerance, suggesting strong survivability under harsh conditions. Collectively, these results indicate that P. pentosaceus THG-219, isolated from ginseng, is a promising, safe, and stable candidate for development as a functional cosmetic ingredient.
Collapse
Affiliation(s)
- Xiangji Jin
- Department of Dermatology, Graduate School, Kyung Hee University, 26 Kyungheedae-ro, Dong-daemun, Seoul 02447, Republic of Korea;
| | - Qiwen Zheng
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (S.-J.Y.)
| | - Trang Thi Minh Nguyen
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (S.-J.Y.)
| | - Gyeong-Seon Yi
- Department of Biopharmaceutical Biotechnology, Graduate School, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea;
| | - Su-Jin Yang
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (S.-J.Y.)
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (S.-J.Y.)
| |
Collapse
|
16
|
Jost S, Herzig C, Birringer M. A Balancing Act-20 Years of Nutrition and Health Claims Regulation in Europe: A Historical Perspective and Reflection. Foods 2025; 14:1651. [PMID: 40361733 PMCID: PMC12071930 DOI: 10.3390/foods14091651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
The Nutrition and Health Claims Regulation (NHCR) has introduced a new regulatory perspective in food manufacturing, along with influencing consumers' perception of health-related food claims. Since 2006, a new standard of science-based claims has significantly impacted the European health food market. Over the years, numerous additional decisions have been made, and the ongoing process remains challenging for policymakers striving to harmonize consumer protection and trade within and outside the European Union (EU). This paper presents the current state of the NHCR's implementation, along with key events aimed at enhancing understanding among consumer organizations and food industry stakeholders, while also offering an insider perspective on relevant policy issues. Additionally, we address two pertinent policy issues to elucidate the associated challenges and opportunities, providing insights to support informed decision-making by policymakers. We use the nutrient profiles framework as a case study to illustrate considerations underpinning the objective of "consumer protection", while the "probiotics" market serves as an example for exploring the goal of "facilitation of trade". This historical perspective and reflection lead us to propose possible solutions for future food regulation.
Collapse
Affiliation(s)
- Sonja Jost
- Center for Sustainable Food Systems, Justus Liebig University Giessen, Senckenbergstrasse 3, 35390 Giessen, Germany;
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, 36037 Fulda, Germany;
- Relationship Management, University of Freiburg, Loewenstrasse 16, 79098 Freiburg, Germany
| | - Christian Herzig
- Center for Sustainable Food Systems, Justus Liebig University Giessen, Senckenbergstrasse 3, 35390 Giessen, Germany;
| | - Marc Birringer
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, 36037 Fulda, Germany;
| |
Collapse
|
17
|
Mafe AN, Nkene IH, Ali ABM, Edo GI, Akpoghelie PO, Yousif E, Isoje EF, Igbuku UA, Ismael SA, Essaghah AEA, Ahmed DS, Umar H, Alamiery AA. Smart Probiotic Solutions for Mycotoxin Mitigation: Innovations in Food Safety and Sustainable Agriculture. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10569-4. [PMID: 40312537 DOI: 10.1007/s12602-025-10569-4] [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: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Mycotoxin contamination poses severe risks to food safety and agricultural sustainability. Probiotic-based interventions offer a promising strategy for mitigating these toxic compounds through adsorption, biodegradation, and gut microbiota modulation. This review examines the mechanisms by which specific probiotic strains inhibit mycotoxin biosynthesis, degrade existing toxins, and enhance host detoxification pathways. Emphasis is placed on strain-specific interactions, genetic and metabolic adaptations, and advancements in formulation technologies that improve probiotic efficacy in food matrices. Also, the review explores smart delivery systems, such as encapsulation techniques and biofilm applications, to enhance probiotic stability and functionality. Issues related to regulatory approval, strain viability, and large-scale implementation are also discussed. By integrating molecular insights, applied case studies, and innovative probiotic-based solutions, this review provides a roadmap for advancing safe and sustainable strategies to combat mycotoxin contamination in food and agricultural systems.
Collapse
Affiliation(s)
- Alice Njolke Mafe
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Jalingo, Nigeria
| | - Istifanus Haruna Nkene
- Department of Microbiology, Faculty of Natural and Applied Sciences, Nasarawa State University, Keffi, Nigeria
| | - Ali B M Ali
- Department of Air Conditioning Engineering, College of Engineering, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Great Iruoghene Edo
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria.
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq.
| | - Patrick Othuke Akpoghelie
- Department of Food Science and Technology, Faculty of Science, Delta State University of Science and Technology, Ozoro, Delta State, Nigeria
| | - Emad Yousif
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Endurance Fegor Isoje
- Department of Biochemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Ufuoma Augustina Igbuku
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Shams A Ismael
- Department of Medical Physics, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Arthur Efeoghene Athan Essaghah
- Department of Urban and Regional Planning, Faculty of Environmental Sciences, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Dina S Ahmed
- Department of Chemical Industries, Institute of Technology-Baghdad, Middle Technical University, Baghdad, Iraq
| | - Huzaifa Umar
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus
| | - Ahmed A Alamiery
- AUIQ, Al-Ayen Scientific Research Center, Al-Ayen Iraqi University, Thi Qar, P.O. Box: 64004, An Nasiriyah, Iraq
| |
Collapse
|
18
|
Veyrat-Durebex C, Osman S, Al Ojaimi Y, Gosset P, Dupuy C, Lefevre A, Emond P, Vourc'h P, Corcia P, Mereghetti L, Kempf F, Raoul C, Blasco H. Gut metabolomic and microbiota analyses in ALS mice reveal specific metabolites despite the absence of significant gut dysbiosis. Amyotroph Lateral Scler Frontotemporal Degener 2025; 26:368-374. [PMID: 39611550 DOI: 10.1080/21678421.2024.2433578] [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: 09/11/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE Over the past years, interest in the role of gut microbiota in neurodegenerative diseases has emerged. Despite numerous publications over the past decade, both in human and pre-clinical studies, there is no clear consensus on the microbiota's role or involvement in ALS. Few studies on mouse models of ALS highlighted a correlation between specific bacteria species and the prognostic or severity of the disease. Still these results lack reproducibility and remain controverted. In this article we present a study of fecal microbiota in the SOD1G93A mouse model associated with a metabolomic analysis of cecum content, compared to controls. METHODS Intestinal metabolomic profile and fecal microbiota were assessed in two cohorts of SODG93A mice compared to wildtype controls at the terminal stage of the ALS disease. RESULTS Results showed a significant difference in metabolomic profile in SOD1G93A mice compared to controls but without a marked change in composition and diversity of fecal microbiota. Nevertheless, we observed an increase of Lachnospiraceae family, which are butyrate-producer bacteria, in SOD1G93A mice. Moreover, some metabolites with significantly different intestinal concentrations are partially produced and linked with intestinal bacteria, such as riboflavin, hippurate, and N-acetylputrescine, leaving us convinced of the interest in looking further into the role of the microbiota in ALS. CONCLUSIONS Despite an alteration of the intestinal metabolome in SOD1G93A mice, microbiota data did not show significant changes, underlying the need for further research.
Collapse
Affiliation(s)
- Charlotte Veyrat-Durebex
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Tours, Tours, France
| | - Samira Osman
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
| | - Yara Al Ojaimi
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
| | | | - Camille Dupuy
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
| | - Antoine Lefevre
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
| | - Patrick Emond
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
| | - Patrick Vourc'h
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Tours, Tours, France
| | - Philippe Corcia
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
- CRMR SLA et autres maladies du neurone moteur, CHRU Tours, Tours, France
| | - Laurent Mereghetti
- UMR INRA 1282 Infectiologie et Santé Publique (ISP), Tours, France
- Laboratoire de Bactériologie, CHRU Tours, Tours, France, and
| | - Florent Kempf
- UMR INRA 1282 Infectiologie et Santé Publique (ISP), Tours, France
| | - Cédric Raoul
- INM, Univ Montpellier, INSERM, Montpellier, France
- ALS Reference Center, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Hélène Blasco
- INSERM Imaging Brain & Neuropsychiatry iBraiN U1253, Team Neurogenomic and Neuronal Pathophysiology, Université de Tours, Tours, France
- Laboratoire de Biochimie et Biologie Moléculaire, CHRU Tours, Tours, France
| |
Collapse
|
19
|
Klews CC, Bryant JL, McCabe J, Atchley AN, Cousins TW, Barnard-Davidson M, Ackermann MR, Netherland M, Hasan NA, Jordan PA, Forsythe ES, Ball PN, Seal BS. Reference Whole Genome Sequence Analyses and Characterization of a Novel Carnobacterium maltaromaticum Distinct Sequence Type Isolated from a North American Gray Wolf ( Canis lupus) Gastrointestinal Tract. Vet Sci 2025; 12:410. [PMID: 40431503 PMCID: PMC12115997 DOI: 10.3390/vetsci12050410] [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: 04/03/2025] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
We hypothesize that bacteria isolated from free-ranging animals could potentially be useful for practical applications. To meet this objective a Gram-positive bacterium was isolated from the gastrointestinal (GI) tract of a Gray Wolf (Canis lupus) using Brucella broth with hemin and vitamin K (BBHK). By small ribosomal RNA (16S) gene sequencing the bacterium was initially identified as a novel Carnobacterium maltaromaticum strain. The bacterium could be propagated both anaerobically and aerobically and was both catalase/oxidase negative and negative by the starch hydrolysis as well as negative using lipase assays. The reference whole genome sequence (WGS) was obtained using both Illumina and Nanopore sequencing. The genome assembly was 3,512,202 bp in length, encoding core bacterial genes with a GC% content of 34.48. No lysogenic bacteriophage genes were detected, although the genome harbors genes for the expression of bacteriocin and other secondary metabolites with potential antimicrobial properties. Multilocus sequence typing (MLST), WGS phylogenetics, average nucleotide identity (ANI), and single nucleotide polymorphism (SNP) analyses of the isolate's genome indicate this bacterium is a newly identified Carnobacterium maltaromaticum sequence type (ST). Members of the Carnobacteria have anti-listeria activities, highlighting their potential functional properties. Consequently, the isolate could be a potential probiotic for canids and this is the first report on an axenic C. maltaromaticum culture from the genus Canis.
Collapse
Affiliation(s)
- C. Cristoph Klews
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Jessika L. Bryant
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Jennifer McCabe
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Ariel N. Atchley
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Thomas W. Cousins
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Maya Barnard-Davidson
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Mark R. Ackermann
- Oregon Veterinary Diagnostic Laboratory, OSU Carlson College of Veterinary Medicine, 134 Magruder Hall, 700 SW 30th, Corvallis, OR 97331, USA;
| | - Michael Netherland
- EzBiome Inc., 704 Quince Orchard Rd Suite 250, Gaithersburg, MD 20878, USA; (M.N.J.); (N.A.H.)
| | - Nur A. Hasan
- EzBiome Inc., 704 Quince Orchard Rd Suite 250, Gaithersburg, MD 20878, USA; (M.N.J.); (N.A.H.)
| | - Peter A. Jordan
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Evan S. Forsythe
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
- Integrative Biology Department, Oregon State University, 2403 Cordley Hall, Corvallis, OR 97331, USA
| | - Patrick N. Ball
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| | - Bruce S. Seal
- Biology Program, Oregon State University-Cascades, 1500 SW Chandler Avenue, Bend, OR 97702, USA; (C.C.K.); (J.L.B.); (J.M.); (A.N.A.); (T.W.C.); (M.B.-D.); (P.A.J.); (E.S.F.)
| |
Collapse
|
20
|
Fan F, Wang Z, Luo Q, Liu Z, Xiao Y, Ren Y. Medical Potential of Insect Symbionts. INSECTS 2025; 16:457. [PMID: 40429170 PMCID: PMC12111880 DOI: 10.3390/insects16050457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/18/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025]
Abstract
Insect symbionts and their metabolites are complex and diverse and are gradually becoming an important source of new medical materials. Some culturable symbionts from insects produce a variety of active compounds with medical potential. Among them, fatty acids, antibacterial peptides, polyene macrolides, alkaloids, and roseoflavin can inhibit the growth of human pathogenic bacteria and fungi; lipases, yeast killer toxins, reactive oxygen species, pyridines, polyethers, macrotetrolide nactins, and macrolides can kill human parasites; and peptides and polyketides can inhibit human tumors. However, due to difficulty in the culture of symbionts in vitro, difficulty in targeting bacteria to specific sites in the human body, the limited capability of symbionts to produce active metabolites in vitro, inconsistent clinical research results, adverse reactions on humans, and the development of antibiotic resistance, the application of insect symbionts and their metabolites in the medical field remains in its infancy. This paper summarizes the medical potential of insect symbionts and their metabolites and analyzes the status quo and existing problems with their medical application. Possible solutions to these problems are also proposed, with the aim of hastening the utilization of insect symbionts and their metabolites in the medical field.
Collapse
Affiliation(s)
- Fanglei Fan
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China; (F.F.); (Q.L.); (Z.L.)
| | - Zhengyan Wang
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China; (F.F.); (Q.L.); (Z.L.)
| | - Qiong Luo
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China; (F.F.); (Q.L.); (Z.L.)
| | - Zhiyuan Liu
- School of Food and Strategic Reserves, Henan University of Technology, Zhengzhou 450001, China; (F.F.); (Q.L.); (Z.L.)
| | - Yu Xiao
- College of Environmental and Life Sciences, Murdoch University, Perth, WA 6150, Australia;
| | - Yonglin Ren
- College of Environmental and Life Sciences, Murdoch University, Perth, WA 6150, Australia;
| |
Collapse
|
21
|
Di Pierro F, Sagheddu V, Galletti S, Casaroli A, Labrini E, Soldi S, Cazzaniga M, Bertuccioli A, Matera M, Cavecchia I, Palazzi CM, Tanda ML, Zerbinati N. Selection, Comparative Genomics, and Potential Probiotic Features of Escherichia coli 5C, a pks-Negative Strain Isolated from Healthy Infant Donor Feces. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10522-5. [PMID: 40238037 DOI: 10.1007/s12602-025-10522-5] [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: 03/10/2025] [Indexed: 04/18/2025]
Abstract
Among the emerging issues in probiotic safety, the possible presence of pks, a gene cluster synthetizing a genotoxin known as colibactin, is one of the most alarming. Indeed, indigenous E. coli strain pks-positive are found in 60% of patients with colorectal cancer, and the most widely used E. coli-based probiotic, known as E. coli Nissle 1917 (DSM 6601), is pks-positive. Starting from 25 potential candidates selected by screening 25 infant stool samples, we have selected an E. coli strain (named 5C, deposited as LMG S-33222) belonging to the phylotype A and having the serovar O173:H1. Having been previously completely sequenced by our group, we have further characterized this strain, demonstrating that it is (i) devoid of the most known potential pathogenic-related genes, (ii) devoid of possible plasmids, (iii) antibiotic-sensitive according to the EFSA panel, (iv) resistant in gastric and enteric juice, (v) significantly producing acetate, (vi) poorly producing histamine, (vii) endowed with a significant in vitro antipathogenic profile, (viii) promoting a significant in vitro immunological response based on IL-10 and IL-12, and (ix) devoid of the pks genes. A comparative genomics versus E. coli Nissle 1917 is also provided. Considering that the other two most commonly used E. coli-based probiotics (E. coli DSM 17252 and E. coli A0 34/86) are respectively pks-positive and alpha-hemolysin-(hly) and cytotoxic necrotizing factor-1-(cnf1) positive, this novel strain (E. coli 5C) is likely the probiotic E. coli strain with the best safety profile available to date for human use.
Collapse
Affiliation(s)
- Francesco Di Pierro
- Microbiota International Clinical Society, 10123, Turin, Italy
- Scientific & Research Department, Velleja Research, 20125, Milan, Italy
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Valeria Sagheddu
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Serena Galletti
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Alice Casaroli
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Edoardo Labrini
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | - Sara Soldi
- AAT-Advanced Analytical Technologies, Fiorenzuola d'Arda, 29017, Piacenza, Italy
| | | | - Alexander Bertuccioli
- Microbiota International Clinical Society, 10123, Turin, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61122, Urbino, Italy
| | - Mariarosaria Matera
- Microbiota International Clinical Society, 10123, Turin, Italy
- Department of Pediatric Emergencies, Misericordia Hospital, 58100, Grosseto, Italy
| | - Ilaria Cavecchia
- Microbiota International Clinical Society, 10123, Turin, Italy
- Microbiomic Department, Koelliker Hospital, 10134, Turin, Italy
| | | | - Maria Laura Tanda
- Endocrine Unit, Department of Medicine and Surgery, University of Insubria, 21100, Varese, Italy
| | - Nicola Zerbinati
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| |
Collapse
|
22
|
Ren M, Jin T, Tong J, Song D, Xie Q, Li X, Li Y, Liu K, Gao J, Liu M, Cheng J. Anti-Inflammatory Effects of Weissella cibaria SDS2.1 Against Klebsiella pneumoniae-Induced Mammary Gland Inflammation. Animals (Basel) 2025; 15:1139. [PMID: 40281973 PMCID: PMC12024108 DOI: 10.3390/ani15081139] [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: 03/04/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
Dairy cows are highly susceptible to mastitis caused by Klebsiella pneumoniae, and treating these infections poses a challenge due to the resistance of the bacterium to common antibiotics. This study aimed to evaluate the safety of W. cibaria SDS2.1 and investigate its protective effects against K. pneumoniae-induced mastitis. The safety of W. cibaria SDS2.1 was assessed through comprehensive analyses, including antibiotic resistance profiling, hemolysis assays, cell cytotoxicity tests, and whole-genome sequencing. Furthermore, its ability to protect against cellular and tissue damage caused by K. pneumoniae-induced mastitis was evaluated using both in vitro and in vivo models. Our results revealed that W. cibaria SDS2.1 was non-hemolytic, non-cytotoxic, and significantly inhibited the growth of K. pneumoniae (p < 0.05). Additionally, W. cibaria SDS2.1 effectively reduced the adhesion and invasion of K. pneumoniae. In the K. pneumoniae-induced mouse mastitis model, W. cibaria SDS2.1 significantly reduced myeloperoxidase (MPO) activity, mammary tissue damage, and the expression of inflammatory cytokines (IL-6, IL-1β, and TNF-α) (p < 0.05). In K. pneumoniae-infected bovine mammary epithelial cells (bMECs), W. cibaria SDS2.1 significantly decreased lactate dehydrogenase (LDH) release, indicating reduced cellular damage. These findings demonstrate that W. cibaria SDS2.1 exhibits anti-inflammatory properties in experimental models, suggesting its potential role in mitigating K. pneumoniae-induced mastitis.
Collapse
Affiliation(s)
- Meiyi Ren
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Tianxiong Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Jingdi Tong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Deyuan Song
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Qinna Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Xiaohan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Yan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| | - Kangping Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
| | - Jian Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China;
| | - Mingchao Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| | - Jia Cheng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; (M.R.); (T.J.); (J.T.); (D.S.); (Q.X.); (X.L.); (Y.L.); (K.L.)
- Key Laboratory of Healthy Breeding in Dairy Cattle (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
23
|
Zhao H, Abbas S, Ren J, Huang H, Song Y, Su X, Wu Q, Ma Y, Tang H, Gao YZ, Li Y, Gu X, Feng J, Hou J, Cheng Y, Li Z, Ma W. Dextran from human feces-derived Weissella cibaria facilitates intestinal mucosal barrier function by modulating gut bacteria and propionate levels. Carbohydr Polym 2025; 354:123300. [PMID: 39978893 DOI: 10.1016/j.carbpol.2025.123300] [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: 09/30/2024] [Revised: 01/03/2025] [Accepted: 01/19/2025] [Indexed: 02/03/2025]
Abstract
The disruption of the intestinal mucosal barrier is strongly associated with the onset of various diseases, including inflammatory bowel disease. Exopolysaccharides (EPS) support the functionality of the intestinal barrier. Weissella Cibaria (W. cibaria), belonging to the lactic acid bacteria, exhibits a significant capacity for EPS production. However, the specific mechanisms by which the EPS produced by W. cibaria confers intestinal barrier protection remain unexplored. Here, we characterized the polysaccharide, EPS-2, produced by W. cibaria isolated from the feces of healthy infants. EPS-2 was a novel dextran composed of α-(1 → 6) and α-(1 → 3,6) glycosidic linkages with a molecular weight of 845 kDa. EPS-2 alleviates intestinal mucosal barrier dysfunction in a mouse model of colitis, via a mechanism specifically reliant on the gut microbiota and their metabolic products, which is different from the well-known direct protective effects of other EPS on the intestinal barrier. EPS-2 reversed colitis-induced reductions in Muribaculaceae and propionate levels, thereby enhancing colonic goblet cell function and mucin content. Additionally, EPS-2 decreased the number of LPS-producing bacteria, such as Escherichia_Shigella. EPS-2 alleviated dextran sulfate sodium-induced intestinal inflammation and barrier damage. Therefore, EPS-2 shows promise as a postbiotic treatment for diseases associated with intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Huan Zhao
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Sakandar Abbas
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Jing Ren
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Haibin Huang
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Ying Song
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Xiaoning Su
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Qiuyang Wu
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Yane Ma
- Department of Gynecological Oncology Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Yi-Zhou Gao
- The Center for Microbes, Development, and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanzhe Li
- Department of Pediatrics, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaoming Gu
- Department of Colon and Rectal Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Jianguo Feng
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province 646000, China
| | - Jingjing Hou
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China
| | - Yan Cheng
- Department of Gynecological Oncology Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China.
| | - Wang Ma
- Oncology department, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China.
| |
Collapse
|
24
|
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
|
25
|
El Boukhari R, Matin M, Bouissane L, Ławiński M, Lushchak O, Singla RK, Mickael M, Mayneris‐Perxachs J, Grafakou ME, Xu S, Liu B, Guan J, Półtorak A, Szpicer A, Wierzbicka A, Tzvetkov NT, Banach M, Horbańczuk JO, Jóźwik A, Cascella M, Shen B, Pirgozliev VR, Wang D, Litvinova O, Adamska O, Kamińska A, Łapiński M, Stolarczyk A, Berindan‐Neagoe I, Milella L, Yeung AWK, Suravajhala P, Bishayee A, Lordan R, Iantovics LB, Lagoa R, Michalczuk M, Stoyanov J, Kinghorn AD, Jalil B, Weckwerth W, Goh BH, Li M, Chaubey G, Russo GL, Frazzini S, Rossi L, Battino M, Jia W, Su Q, Ma X, Rollinger JM, Rittmann SKR, Sheridan H, Walsh JJ, Lizard G, Karpiński TM, Silva AS, Piwowarski J, Xie L, Fan T, Giampieri F, El Midaoui A, Wong K, Gan R, Fatimi A, Atanasov AG. Enhancing human gut health: Global innovations in dysbiosis management. IMETA 2025. [DOI: 10.1002/imt2.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Affiliation(s)
- Reda El Boukhari
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Maima Matin
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Latifa Bouissane
- Molecular Chemistry, Materials and Catalysis Laboratory, Faculty of Sciences and Technologies Sultan Moulay Slimane University Beni Mellal Morocco
| | - Michał Ławiński
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of General, Gastroenterologic and Oncologic Surgery Medical University of Warsaw Warsaw Poland
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology Vasyl Stefanyk Precarpathian National University Ivano‐Frankivsk Ukraine
- Research and Development University Ivano‐Frankivsk Ukraine
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease‐related Molecular Network, West China Hospital Sichuan University Chengdu Sichuan China
- School of Pharmaceutical Sciences Lovely Professional University Phagwara Punjab India
| | - Michel‐Edwar Mickael
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Jordi Mayneris‐Perxachs
- Department of Diabetes, Endocrinology and Nutrition Dr. Josep Trueta University Hospital Girona Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) Madrid Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI‐CERCA) Parc Hospitalari Martí i Julià Salt Spain
| | - Maria Eleni Grafakou
- Chair of Pharmaceutical Biology, Faculty of Pharmacy and Chemistry University of Regensburg Germany
| | - Shuhua Xu
- Center for Evolutionary Biology, School of Life Sciences Fudan University Shanghai China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center Fudan University Shanghai China
| | - Bowen Liu
- School of Agriculture Yunnan University Kunming China
| | - Jiayi Guan
- Henan Institute of Medical and Pharmaceutical Sciences Zhengzhou University Zhengzhou China
| | - Andrzej Półtorak
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Arkadiusz Szpicer
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Agnieszka Wierzbicka
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Department of Technique and Food Development, Institute of Human Nutrition Sciences Warsaw University of Life Sciences Warsaw Poland
| | - Nikolay T. Tzvetkov
- Department of Biochemical Pharmacology and Drug Design, Institute of Molecular Biology “Roumen Tsanev” Bulgarian Academy of Sciences Sofia Bulgaria
| | - Maciej Banach
- Faculty of Medicine The John Paul II Catholic University of Lublin (KUL) Lublin Poland
- Department of Cardiology and Adult Congenital Heart Diseases Polish Mother's Memorial Hospital Research Institute (PMMHRI) Lodz Poland
- Department of Preventive Cardiology and Lipidology Medical University of Lodz (MUL) Lodz Poland
- Ciccarone Center for the Prevention of Cardiovascular Disease Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Jarosław Olav Horbańczuk
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Artur Jóźwik
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
| | - Marco Cascella
- Anesthesia and Pain Medicine, Department of Medicine, Surgery and Dentistry “Scuola MedicaSalernitana” University of Salerno Baronissi Italy
| | - Bairong Shen
- Department of Critical Care Medicine and Institutes for Systems Genetics Frontiers Science Center for Disease‐Related Molecular Network, West China Hospital,Sichuan University Chengdu Sichuan China
- Center for High Altitude Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | | | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research McMaster University Hamilton Ontario Canada
- Division of Endocrinology and Metabolism, Department of Medicine McMaster University Hamilton Ontario Canada
| | - Olena Litvinova
- National University of Pharmacy of the Ministry of Health of Ukraine Kharkiv Ukraine
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
| | - Olga Adamska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Agnieszka Kamińska
- Faculty of Medicine Collegium Medicum Cardinal Stefan Wyszyński University in Warsaw Warsaw Poland
| | - Marcin Łapiński
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Artur Stolarczyk
- Orthopaedic and Rehabilitation Department Medical University of Warsaw Warsaw Poland
| | - Ioana Berindan‐Neagoe
- Department of Genomics MEDFUTURE ‐ Institute for Biomedical Research“Iuliu Hațieganu” University of Medicine and Pharmacy No. 23 Cluj‐Napoca Romania
| | - Luigi Milella
- Department of Health Sciences University of Basilicata Potenza Italy
| | - Andy Wai Kan Yeung
- Oral and Maxillofacial Radiology, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry The University of Hong Kong Pokfulam Hong Kong SAR
| | - Prashanth Suravajhala
- Amrita School of Biotechnology Amrita Viswa Vidyapeetham Clappana Kerala India
- Department of Biosciences Manipal University Jaipur, Dehmi Kala Jaipur Rajasthan India
| | - Anupam Bishayee
- Department of Pharmacology College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine Bradenton Florida USA
| | - Ronan Lordan
- The Institute for Translational Medicine and Therapeutics, Perelman School of Medicine University of Pennsylvania Philadelphia Pennsylvania USA
| | - Laszlo Barna Iantovics
- Department of Electrical Engineering and Information Technology George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures Targu Mures Romania
| | - Ricardo Lagoa
- ESTG‐Polytechnic Institute of Leiria Morro do Lena‐Alto do Vieiro Leiria Portugal
- LSRE‐LCM‐Associate Laboratory in Chemical Engineering University of Porto Porto Portugal
| | - Monika Michalczuk
- Department of Animal Breeding, Institute of Animal Sciences Warsaw University of Life Sciences ‐ SGGW Warsaw Poland
| | - Jivko Stoyanov
- Swiss Paraplegic Research Nottwil Switzerland
- Institute of Social and Preventive Medicine (ISPM) University of Bern Bern Switzerland
| | | | - Banaz Jalil
- Pharmacognosy and Phytotherapy UCL School of Pharmacy London UK
| | - Wolfram Weckwerth
- Molecular Systems Biology Lab (MOSYS), Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
- Vienna Metabolomics Center (VIME) University of Vienna Vienna Austria
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre (SBMDC) School of Medical and Life Sciences Subang Jaya Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy Monash University Malaysia Subang Jaya Malaysia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine University of Technology Sydney Ultimo New South Wales Australia
| | - Meng‐Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Biliary‐Pancreatic Surgery, Renji Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology Banaras Hindu University Varanasi Uttar Pradesh India
| | - Gian Luigi Russo
- National Research Council Institute of Food Sciences Avellino Italy
| | - Sara Frazzini
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Science (DIVAS) University of Milan Lodi Italy
| | - Maurizio Battino
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- International Joint Research, Laboratory of Intelligent Agriculture and Agri‐Products Processing Jiangsu University Zhenjiang China
| | - Wei Jia
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong SAR
| | - Qi Su
- Microbiota I‐Center Shatin Hong Kong SAR
- Department of Medicine and Therapeutics The Chinese University of Hong Kong Shatin Hong Kong SAR
| | - Xiaoqiang Ma
- Department of Food Science and Technology, School of Agriculture and Biology Shanghai Jiao Tong University Shanghai China
| | - Judith M. Rollinger
- Division of Pharmacognosy, Department of Pharmaceutical Sciences, Faculty of Life Sciences University of Vienna Vienna Austria
| | - Simon K.‐M. R. Rittmann
- Archaea Physiology & Biotechnology Group, Department of Functional and Evolutionary Ecology University of Vienna Vienna Austria
| | - Helen Sheridan
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - John J. Walsh
- The NatPro Centre & School of Pharmacy and Pharmaceutical Sciences Trinity College Dublin Dublin Ireland
| | - Gérard Lizard
- Université Bourgogne Europe/INSERM, 21000 Dijon and PHYNOHA Consulting Fontaine‐lès‐Dijon France
| | - Tomasz M. Karpiński
- Department of Medical Microbiology Poznań University of Medical Sciences Poznań Poland
| | - Ana Sanches Silva
- University of Coimbra, Faculty of Pharmacy, Polo III, Azinhaga de Santa Comba Coimbra Portugal
- Centre for Animal Science Studies (CECA), ICETA University of Porto Porto Portugal
| | - Jakub Piwowarski
- Microbiota Lab, Department of Pharmaceutical Microbiology and Bioanalysis Medical University of Warsaw Warsaw Poland
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology Institute of Microbiology, Guangdong Academy of Sciences Guangzhou China
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Tai‐Ping Fan
- School of Life & Health Sciences Fuyao University of Science & Technology Fuzhou Fujian China
| | - Francesca Giampieri
- Department of Clinical Sciences Polytechnic University of Marche Ancona Italy
- Joint Laboratory on Food Science, Nutrition, and Intelligent Processing of Foods Polytechnic University of Marche (Italy), Universidad Europea del Atlántico (Spain), and Jiangsu University (China) Ancona Italy
- Research Group on Food, Nutritional Biochemistry and Health Universidad Europea del Atlántico Santander Spain
- International Research Center for Food Nutrition and Safety Jiangsu University Zhenjiang China
| | - Adil El Midaoui
- Faculty of Sciences and Techniques Errachidia, Moulay Ismail University of Meknes Meknes Morocco
- Department of Pharmacology and Physiology, Faculty of Medicine University of Montreal Montreal Quebec Canada
| | - Ka‐Hing Wong
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ren‐You Gan
- Research Institute for Future Food The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
- Department of Food Science and Nutrition The Hong Kong Polytechnic University Hung Hom Hong Kong SAR
| | - Ahmed Fatimi
- Chemical Science and Engineering Research Team (ERSIC), Department of Chemistry, Polydisciplinary Faculty of Beni Mellal (FPBM) Sultan Moulay Slimane University (USMS) Beni Mellal Morocco
| | - Atanas G. Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences Jastrzębiec Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety Medical University of Vienna Vienna Austria
- Laboratory of Natural Products and Medicinal Chemistry (LNPMC), Center for Global Health Research, Saveetha Medical College and Hospital Saveetha Institute of Medical and Technical Sciences (SIMATS) Thandalam Chennai India
| |
Collapse
|
26
|
Lu S, Yue K, He S, Huang Y, Ren Z, Xu J. Safety Assessment of Lactiplantibacillus plantarum GUANKE Based on Whole-Genome Sequencing, Phenotypic, and Anti-Inflammatory Capacity Analysis. Microorganisms 2025; 13:873. [PMID: 40284709 PMCID: PMC12029457 DOI: 10.3390/microorganisms13040873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Lactiplantibacillus plantarum GUANKE (L. plantarum GUANKE) is a Gram-positive bacterium isolated from the feces of healthy volunteers. Whole-genome sequencing analysis (WGS) revealed that the genome of L. plantarum GUANKE consists of one chromosome and two plasmids, with the chromosome harbors 2955 CDS, 66 tRNAs, and 5 rRNAs. The genome is devoid of virulence factors and Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) systems. It contains three intact prophage regions and bacteriocin biosynthesis genes (plantaricins K, F, and E), as well as seventeen genomic islands lacking antibiotic resistance or pathogenicity determinants. Functional prediction outcomes identified that the genome of L. plantarum GUANKE is closely related to transcription, carbohydrate transport and metabolism, and amino acid transport and metabolism. Carbohydrate-active enzymes (CAZymes) analysis and GutSMASH analysis revealed that the genome of L. plantarum GUANKE contained 100 carbohydrate-active enzyme genes and two specialized metabolic gene clusters. Safety assessments confirmed that L. plantarum GUANKE neither exhibited β-hemolytic activity nor harbored detectable transferable drug resistance genes. The strain exhibited remarkable acid tolerance and bile salt resistance. Cellular adhesion assays demonstrated moderate binding capacity to Caco-2 intestinal epithelium (4.3 ± 0.007)%. In vitro analyses using lipopolysaccharide (LPS)-stimulated macrophage models demonstrated that L. plantarum GUANKE significantly suppressed the secretion of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β), exhibiting dose-dependent anti-inflammatory activity. In vivo experiments showed that L. plantarum GUANKE was involved in the regulation of the apical junction pathway and interferon pathway in colon tissue of normal mice.
Collapse
Affiliation(s)
| | | | | | | | - Zhihong Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102200, China
| | - Jianguo Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102200, China
| |
Collapse
|
27
|
Nista EC, Parello S, Brigida M, Amadei G, Saviano A, De Lucia SS, Petruzziello C, Migneco A, Ojetti V. Exploring the Role of Gut Microbiota and Probiotics in Acute Pancreatitis: A Comprehensive Review. Int J Mol Sci 2025; 26:3433. [PMID: 40244415 PMCID: PMC11989318 DOI: 10.3390/ijms26073433] [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: 03/09/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Acute pancreatitis (AP) is a common and potentially severe gastrointestinal condition characterized by acute inflammation of the pancreas. The pathophysiology of AP is multifactorial and intricate, involving a cascade of events that lead to pancreatic injury and systemic inflammation. The progression of AP is influenced by many factors, including genetic predispositions, environmental triggers, and immune dysregulation. Recent studies showed a critical involvement of the gut microbiota in shaping the immune response and modulating inflammatory processes during AP. This review aims to provide a comprehensive overview of the emerging role of gut microbiota and probiotics in AP. We analyzed the implication of gut microbiota in pathogenesis of AP and the modification during an acute attack. The primary goals of microbiome-based therapies, which include probiotics, prebiotics, antibiotics, fecal microbiota transplantation, and enteral nutrition, are to alter the composition of the gut microbial community and the amount of metabolites derived from the microbiota. By resetting the entire flora or supplementing it with certain beneficial organisms and their byproducts, these therapeutic approaches aim to eradicate harmful microorganisms, reducing inflammation and avoiding bacterial translocation and the potential microbiota-based therapeutic target for AP from nutrition to pre- and probiotic supplementation to fecal transplantation.
Collapse
Affiliation(s)
- Enrico Celestino Nista
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Simone Parello
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Mattia Brigida
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
| | - Giulio Amadei
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Angela Saviano
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Sara Sofia De Lucia
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | | | - Alessio Migneco
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Veronica Ojetti
- Ospedale San Carlo di Nancy, GVM Research, 00165 Rome, Italy
- Department of Internal Medicine, UniCamillus International Medical University of Rome, 00131 Rome, Italy
| |
Collapse
|
28
|
Li J, Xu J, Guo X, Xu H, Huang C, Nie Y, Zhou Y. Odoribacter splanchnicus-A Next-Generation Probiotic Candidate. Microorganisms 2025; 13:815. [PMID: 40284651 PMCID: PMC12029356 DOI: 10.3390/microorganisms13040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
As an important intestinal microorganism, Odoribacter splanchnicus frequently appears in high-throughput sequencing analyses, although pure culture research on this microorganism is not as advanced. It is widely present in the mammalian gut and is closely associated with the health status of the host and the incidence of various diseases. In recent years, changes in the abundance of O. splanchnicus have been found to be positively or negatively correlated with health issues, such as obesity, metabolic syndrome, diabetes, and intestinal inflammation. It may exhibit a dual protective or promotional role in specific diseases. Thus, it may play an important role in regulating host metabolism, immune response, and intestinal homeostasis. Additional research has revealed that O. splanchnicus can synthesize various metabolites, especially short-chain fatty acids (SCFAs), which play a key role in promoting intestinal health, enhancing energy metabolism, improving insulin resistance, and regulating immune responses in the host. Therefore, O. splanchnicus is a strong candidate for "next-generation probiotics", and its potential probiotic function provides novel ideas for the development of functional foods and the prevention and treatment of metabolic and intestinal inflammatory diseases. These findings can help develop new biological treatment strategies and optimize health management plans.
Collapse
Affiliation(s)
- Jianhong Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; (J.L.); (J.X.); (X.G.); (H.X.); (C.H.)
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou 510180, China
| |
Collapse
|
29
|
Horill S, Zhou XK, Jin W. Probiotics as a possible novel therapeutic option to mitigate perioperative neurocognitive disorders: A review exploring the latest research findings. J Clin Anesth 2025; 103:111801. [PMID: 40043583 DOI: 10.1016/j.jclinane.2025.111801] [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: 10/19/2024] [Revised: 01/27/2025] [Accepted: 02/26/2025] [Indexed: 05/16/2025]
Abstract
Perioperative neurocognitive disorders (PND) refer to a constellation of symptoms that primarily affect the elderly and typically manifest as common complications after exposure to surgery and anesthesia. PND is associated with high morbidity, mortality, and progression to neurodegenerative diseases, thus exerting significant financial strains on families as well as the healthcare system. Given that an ageing global population is an inevitable trend and, with the latest advances in the healthcare system, an ever-growing number of elderly people present for surgery and anesthesia, PND is of prominent concern. The two-way communication between the intestinal flora and the brain, also known as the microbiota-gut-brain axis, plays an important role in central nervous system development, and multiple studies have highlighted the influence exerted by gut microbiome in both health and disease. Pertinent studies have corroborated the fact that anesthesia and surgery disrupt the harmony of the gut ecology, which sets off a cascade of events that initiate neuroinflammation, eventually leading to PND. Probiotics, which are live microorganisms that promote the host's health, have been shown as a viable option to restore or minimise the disruption of gut flora. Evidence exists that probiotics exhibit immunomodulatory and anti-inflammatory benefits. Given the effectiveness of probiotics in reducing neuroinflammation, research has also focused on their impact on the development of PND. This review aims to compile the data from relevant clinical trials focusing on the influence of probiotics on PND to determine whether the derived findings might be applied for the prevention and treatment of PND.
Collapse
Affiliation(s)
- Smita Horill
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Xiao-Kai Zhou
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Wenjie Jin
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
30
|
Tufail MA, Schmitz RA. Exploring the Probiotic Potential of Bacteroides spp. Within One Health Paradigm. Probiotics Antimicrob Proteins 2025; 17:681-704. [PMID: 39377977 PMCID: PMC11925995 DOI: 10.1007/s12602-024-10370-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 03/21/2025]
Abstract
Probiotics are pivotal in maintaining or restoring the balance of human intestinal microbiota, a crucial factor in mitigating diseases and preserving the host's health. Exploration into Bacteroides spp. reveals substantial promise in their development as next-generation probiotics due to their profound interaction with host immune cells and capability to regulate the microbiome's metabolism by significantly impacting metabolite production. These beneficial bacteria exhibit potential in ameliorating various health issues such as intestinal disorders, cardiovascular diseases, behavioral disorders, and even cancer. Though it's important to note that a high percentage of them are as well opportunistic pathogens, posing risks under certain conditions. Studies highlight their role in modifying immune responses and improving health conditions by regulating lymphocytes, controlling metabolism, and preventing inflammation and cancer. The safety and efficacy of Bacteroides strains are currently under scrutiny by the European Commission for authorization in food processing, marking a significant step towards their commercialization. The recent advancements in bacterial isolation and sequencing methodologies, coupled with the integration of Metagenome-Assembled Genomes (MAGs) binning from metagenomics data, continue to unveil the potential of Bacteroides spp., aiding in the broader understanding and application of these novel probiotics in health and disease management.
Collapse
Affiliation(s)
- Muhammad Aammar Tufail
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| | - Ruth A Schmitz
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| |
Collapse
|
31
|
Xu R, Yu Y, Chen T. Exploring the dark side of probiotics to pursue light: Intrinsic and extrinsic risks to be opportunistic pathogens. Curr Res Food Sci 2025; 10:101044. [PMID: 40235735 PMCID: PMC11999689 DOI: 10.1016/j.crfs.2025.101044] [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: 01/26/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Probiotics, live microorganisms with multiple health benefits, have gained popularity for their roles in maintaining daily health and treating a variety of diseases. However, they have the potential to be opportunistic pathogens in some conditions. This review delves into the intrinsic and extrinsic risks associated with probiotics. Intrinsic risks involve the production of harmful substances, such as toxins and invasive factors, biofilm formation, bacteria emboli, antibiotic resistance with relevant genetic materials, genetic plasticity, and metabolic issues, while extrinsic risks include problems in regulatory oversight and public awareness, host health status and appropriately administration. It emphasizes the need for a balanced view of their therapeutic benefits and potential hazards, advocating for further research to understand the complex interactions between probiotics and the human microbiome, to optimize the safety and efficacy of probiotics.
Collapse
Affiliation(s)
- Ruiyan Xu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yifeng Yu
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tingtao Chen
- Ophthalmologic Centre, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- National Engineering Research Centre for Bioengineering Drugs and the Technologies, Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
32
|
Xie Q, Liu J, Yu P, Qiu T, Jiang S, Yu R. Unlocking the power of probiotics, postbiotics: targeting apoptosis for the treatment and prevention of digestive diseases. Front Nutr 2025; 12:1570268. [PMID: 40230717 PMCID: PMC11994438 DOI: 10.3389/fnut.2025.1570268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Digestive diseases are becoming an increasingly serious health burden, creating an urgent need to develop more effective treatment strategies. Probiotics and postbiotics have been extensively studied for their potential to prevent and treat digestive diseases. Growing evidence suggests that programmed cell death, especially apoptosis, is a critical mechanism influencing the molecular and biological aspects of digestive diseases, contributing to disease progression. Understanding the mechanisms and signaling pathways by which probiotics and postbiotics regulate apoptosis could reveal new therapeutic targets for treating digestive diseases. This review focuses on the beneficial effects of probiotics and postbiotics in regulating apoptosis across a range of liver diseases, including non-alcoholic fatty liver disease, liver injury, cirrhosis, and liver cancer. It also explores their effects on gastrointestinal diseases, such as colorectal cancer, colitis, gastrointestinal injury, and infectious diarrhea. Furthermore, some probiotics help balance the gut microbiota, enhance intestinal barrier function, and regulate the immune system, all of which are closely associated with apoptosis. Moreover, emerging technologies, such as encapsulation methods, have been developed to stabilize probiotics, primarily based on experimental findings from rodent and human studies.
Collapse
Affiliation(s)
- Qiuyan Xie
- Department of Neonatology, Affiliated Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Ji Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ping Yu
- Reproductive Medicine Centre, Affiliated Women’s Hospital of Jiangnan University, Wuxi, China
| | - Ting Qiu
- Department of Child Health Care, Affiliated Women’s Hospital of Jiangnan University, Wuxi, China
| | - Shanyu Jiang
- Department of Neonatology, Affiliated Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, Affiliated Women’s Hospital of Jiangnan University, Wuxi Maternity and Child Health Care Hospital, Wuxi, China
| |
Collapse
|
33
|
Huang F, Yang N, Zhang Q, Luo C, Wang J, Yang Y, Yue B, Chen P, Zhang X. Marine-Derived Enterococcus faecalis HY0110 as a Next-Generation Functional Food Probiotic: Comprehensive In Vitro and In Vivo Bioactivity Evaluation and Synergistic Fermentation of Periplaneta americana Extract Powder. Foods 2025; 14:1181. [PMID: 40238337 PMCID: PMC11988638 DOI: 10.3390/foods14071181] [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: 03/11/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Addressing the escalating global burdens of inflammatory bowel disease and antimicrobial resistance demanded innovative food-based approaches to fortify gut health and suppress pathogens. We introduced a novel edible probiotic, Enterococcus faecalis HY0110, isolated from marine Thunnus thynnus. Through comprehensive in vitro, in vivo, and metabolomic analyses, we demonstrated its superior antibacterial effects compared to Lactobacillus rhamnosus GG, along with significantly enhanced antioxidant and free-radical scavenging capacities. Notably, elevated acetic acid production strongly correlated with its antimicrobial efficacy (R ≥ 0.999). HY0110 also exerted antiproliferative effects on HT-29 colorectal cancer cells by attenuating β-catenin and BCL-2 expression while upregulating pro-apoptotic markers P62 and c-PARP. In a DSS-induced colitis model, HY0110 alleviated inflammation, restored gut microbial homeostasis, and enhanced deterministic processes in community assembly dynamics. Furthermore, fermenting Periplaneta americana powder with HY0110 triggered extensive metabolic remodeling, notably a 668.73-fold rise in astragaloside A, plus increases in L-Leucyl-L-Alanine, S-lactoylglutathione, and 16,16-dimethyl prostaglandin A1. These shifts diminished harmful components and amplified essential amino acids and peptides to bolster immune modulation, redox balance, and anti-inflammatory responses. This work established a transformative paradigm for utilizing marine probiotics and novel entomological substrates in functional foods, presenting strategic pathways for precision nutrition and inflammatory disease management.
Collapse
Affiliation(s)
- Feiyun Huang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Nan Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, China;
| | - Qingqing Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Cuiling Luo
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Jingheng Wang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Yu Yang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Bisong Yue
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Peng Chen
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu 610065, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| |
Collapse
|
34
|
Zhang F, Kamm MA, Wu X, Kao D, Borody TJ, Chen LA, He X, Fischer M, Wong SH, Ng SC, Cui B, Chan FKL, Nie Y, Sood A, Li J, Sun Y, Dai I, Chen Q, Lv M, Zhang Z, Ianiro G, Yang Y, Kelly CR. Preferred Reporting Items for Microbiotherapy (PRIM) Guidelines Across Medical Disciplines: An International Delphi Consensus. J Gastroenterol Hepatol 2025. [PMID: 40143713 DOI: 10.1111/jgh.16947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/06/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
Microbiotherapy has opened new avenues for managing dysbiosis-related diseases. However, many studies did not cover all the necessary reporting items for microbiotherapy making the interpretation of results, safety assessment, technology extension, and even the transparency of legitimacy difficult. This project consisted of 2 phases. First, we proposed an initial preferred reporting items for microbiotherapy (PRIM) checklist and applied it to oncology studies from 2011 to 2023 according to Meta-Analyses guideline. Only 39.3% (n = 64) of these studies (n = 163) met all PRIM checklist items. The culture-based microbiotherapy (CMT) studies had higher score than non-culture-based (NMT) ones (p = 0.018). In the second phase, the expert panel consisting of 22 specialists from eight countries across Asia, Australia, Europe, and North America refined and finalized the PRIM guidelines (named as PRIM 2024) through Delphi consensus. The PRIM 2024 guidelines conclude 10 statements and 18 points on diagnosis, delivery route, source, classification, preparation, dosage, state, concomitant treatment, efficacy, and safety. The panel defined less than 80% of all PRIM points (14 points) as low-quality reports. These guidelines are recommended for reporting on microbiotherapy in clinical studies and reports on compassionate use, including but not limited to fecal microbiota transplantation, phage therapy, probiotics, and synbiotics. These consistent and transparent reporting items can help researchers and practitioners better evaluate, compare, implement research findings in microbiotherapy.
Collapse
Affiliation(s)
- Faming Zhang
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Michael A Kamm
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Victoria, Australia
| | - Xia Wu
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Thomas J Borody
- Center for Digestive Diseases, Sydney, New South Wales, Australia
| | - Lea Ann Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers, New Brunswick, USA
| | - Xingxiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sunny H Wong
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, National Healthcare Group, Singapore
| | - Siew C Ng
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bota Cui
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Francis K-L Chan
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Yongzhan Nie
- National Clinical Research Center for Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ajit Sood
- Department of Gastroenterology, Dayanand Medical College and Hospital, Ludhiana, Punjab, India
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Beijing, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ishikawa Dai
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo, Japan
| | - Qiyi Chen
- Department of Colorectal Disease, Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital, Shanghai, China
| | - Muhan Lv
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zulun Zhang
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Yunsheng Yang
- National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Colleen R Kelly
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int J Mol Sci 2025; 26:3059. [PMID: 40243712 PMCID: PMC11988433 DOI: 10.3390/ijms26073059] [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: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn's disease. The gut microbiota's role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies-including prebiotics, probiotics, postbiotics, and symbiotics-have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients' quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Pierluigi Puca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Federica di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Alfredo Papa
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
| |
Collapse
|
36
|
Giambra V, Caldarelli M, Franza L, Rio P, Bruno G, di Iasio S, Mastrogiovanni A, Gasbarrini A, Gambassi G, Cianci R. The Role of Notch Signaling and Gut Microbiota in Autoinflammatory Diseases: Mechanisms and Future Views. Biomedicines 2025; 13:768. [PMID: 40299348 PMCID: PMC12024679 DOI: 10.3390/biomedicines13040768] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Notch signaling is an evolutionarily conserved, multifunctional pathway involved in cell fate determination and immune modulation and contributes to the pathogenesis of autoinflammatory diseases. Emerging evidence reveals a bidirectional interaction between Notch and the gut microbiota (GM), whereby GM composition is capable of modulating Notch signaling through the binding of microbial elements to Notch receptors, leading to immune modulation. Furthermore, Notch regulates the GM by promoting SCFA-producing bacteria while suppressing proinflammatory strains. Beneficial microbes, such as Lactobacillus and Akkermansia muciniphila, modulate Notch and reduce proinflammatory cytokine production (such as IL-6 and TNF-α). The interaction between GM and Notch can either amplify or attenuate inflammatory pathways in inflammatory bowel diseases (IBDs), Behçet's disease, and PAPA syndrome. Together, these findings provide novel therapeutic perspectives for autoinflammatory diseases by targeting the GM via probiotics or inhibiting Notch signaling. This review focuses on Notch-GM crosstalk and how GM-based and/or Notch-targeted approaches may modulate immune responses and promote better clinical outcomes.
Collapse
Affiliation(s)
- Vincenzo Giambra
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Laura Franza
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
- Department of Emergency Medicine, AOU Modena, 41125 Modena, Italy
| | - Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Gaja Bruno
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Serena di Iasio
- Institute for Stem Cell Biology, Regenerative Medicine and Innovative Therapies (ISBReMIT), Fondazione IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, Italy; (V.G.)
| | - Andrea Mastrogiovanni
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, 00168 Rome, Italy; (M.C.); (A.M.); (G.G.); (R.C.)
- Fondazione Policlinico Universitario A. Gemelli, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy
| |
Collapse
|
37
|
Gong P, Tang X. The impact of probiotic supplementation on gastric motility and nutrient absorption in elderly patients with Gastrointestinal disorders. BMC Gastroenterol 2025; 25:192. [PMID: 40114066 PMCID: PMC11927212 DOI: 10.1186/s12876-025-03740-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/27/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Gastrointestinal disorders (GIDs) in the elderly often lead to impaired gastric motility and nutrient absorption, exacerbating malnutrition. Probiotics, particularly Lactobacillus rhamnosus GG (LGG), may enhance gastric motility and nutrient absorption. This study evaluates the impact of LGG supplementation on gastric motility and nutrient absorption in elderly patients with GIDs. METHODS A retrospective analysis was conducted on 231 elderly patients with GIDs, divided into a probiotic supplementation (PS) group (n = 110) and a NPS group (n = 121). The PS group received LGG (1 × 1010 CFU, twice daily) for at least 7 days. Baseline and post-treatment measurements included gastric motility via ultrasonography, gastrointestinal hormone levels using radioimmunoassay, and nutrient absorption markers through ELISA and calorimetry. RESULTS Post-treatment, the PS group exhibited significantly improved gastric motility, with increased antral contraction amplitude (58.65 mm vs. 56.53 mm; P = 0.004), frequency (4.06 vs. 3.81 times/min; P = 0.009), and reduced gastric half-emptying time (28.15 min vs. 29.77 min; P = 0.007). Hormone analyses showed elevated motilin and neuropeptide Y levels and decreased vasoactive intestinal peptide levels in the PS group (P < 0.05). Nutrient absorption markers indicated decreased stool fat, protein, and carbohydrate content, enhanced intestinal permeability, increased weight and digestibility of energy, fat, and protein in the PS group (P < 0.05). CONCLUSION PS with LGG significantly enhances gastric motility and nutrient absorption in elderly patients with GIDs, indicating potential therapeutic benefits for addressing digestive dysfunction and malnutrition in this demographic.
Collapse
Affiliation(s)
- Pingting Gong
- Department of Geriatrics, Liangping District People's Hospital, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuehong Tang
- Department of Gastroenterology, Second People's Hospital of Banan District, No.14, Xincun, Huaxi Street, Banan District, Chongqing, 401320, China.
| |
Collapse
|
38
|
Gupta N, Al-Dossari M, El-Gawaad NSA, Alghamdi S, Qusty NF, Babalghith AO, Yadav VK, Niazi P, Mallasiy LO. Lactiplantibacillus plantarum Moderating Effect on Autoimmune Celiac Disease Triggers. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10514-5. [PMID: 40106190 DOI: 10.1007/s12602-025-10514-5] [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: 03/06/2025] [Indexed: 03/22/2025]
Abstract
The only approved preventive treatment option GFD remains insufficient to manage Celiac Disease (CeD). A cohort of probiotic bacteria recently indicated that probiotic bacteria such as L. plantarum (LP) have a protective effect on CeD. LP has been a prominent probiotic, studied for numerous modulating properties. This review highlights and summarizes LP's ameliorating effect on various triggers/drivers of CeD. Probiotic LP potential for CeD is noticeable, mainly involving gut microbiota modulation, gluten digestion, intestinal homeostasis, CeD-associated pathogens reduction, and CD4 + T cell regulation. LP supplementation maintains intestinal physiology by improving the ratio of intestinal villus height to crypt depth. Gut microbiota modulation also improves tight junction proteins and the intestinal barrier. LP increases the digestibility of immunoreactive 33-mer gliadin peptides and regulates immune triggers such as CD4 + T cells. LP supplementation may minimize the gastrointestinal symptoms of CeD. Nevertheless, the therapeutic applicability of LP is subjected to significant clinical and nonclinical studies.
Collapse
Affiliation(s)
- Nishant Gupta
- Medical Division, River Engineering Private Limited, Ecotech-3, Greater Noida, India.
| | - M Al-Dossari
- Muhayil Asir, Applied College, King Khalid University, 62529, Abha, Saudi Arabia
| | - N S Abd El-Gawaad
- Muhayil Asir, Applied College, King Khalid University, 62529, Abha, Saudi Arabia
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Naeem F Qusty
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University Makkah, Makkah, Saudi Arabia
| | - Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Parwiz Niazi
- Department of Biology, Faculty of Education, Kandahar University, Kandahar, 3801, Afghanistan
| | - L O Mallasiy
- Muhayil Asir, Applied College, King Khalid University, 61913, Abha, Saudi Arabia
| |
Collapse
|
39
|
Lin X, Xu M, Lan R, Hu D, Zhang S, Zhang S, Lu Y, Sun H, Yang J, Liu L, Xu J. Gut commensal Alistipes shahii improves experimental colitis in mice with reduced intestinal epithelial damage and cytokine secretion. mSystems 2025; 10:e0160724. [PMID: 39936902 PMCID: PMC11915872 DOI: 10.1128/msystems.01607-24] [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: 12/02/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
The commensal bacterium Alistipes shahii is a core microbe of the human gut microbiome and its abundance is negatively correlated with inflammatory bowel diseases (IBDs). However, its fundamental role in regulating inflammatory response remains unknown. Using a dextran sulfate sodium (DSS)-induced colitis mouse model, we examined the effect of A. shahii strain As360 intervention on host inflammatory response and found that A. shahii As360 alleviated disease activity index, colon shortening, and colonic histopathological lesion. The levels of tight junction proteins (mainly ZO1 and claudin-1) were decreased in DSS-induced colitis mice, whereas the levels of these proteins were elevated in colitis mice with A. shahii As360 treatment. In addition, A. shahii As360 treatment led to alterations in cytokine release, especially an increase of IL10. It also led to reduced expressions of mtor and Nlrp3 and increased expression of mTOR inhibitor Ddit4 at the transcriptional level. 16S rRNA amplicon sequencing found that Bacteroides, a producer of short-chain fatty acids (SCFAs), was enriched in the fecal samples of mice with A. shahii treatment. Metabolic analyses found that, following A. shahii As360 treatment, the SCFAs in the fecal content was increased whereas lactic acid was decreased in the cecal content. These findings suggest that supplementation with A. shahii As360 is a promising strategy to prevent colitis.IMPORTANCEAs one of the core microbes and keystone species in the human gut, Alistipes shahii has the potential to inhibit inflammation and improve inflammatory bowel diseases (IBDs) conditions. In this study, we experimentally demonstrated that oral administration of A. shahii As360 alleviated symptoms of colitis, altered the release of cellular inflammatory factors, reduced the intestinal epithelial barrier damage, and changed gut microbiota and fecal metabolites. These findings provide a deeper understanding of the beneficial effects of A. shahii and its perspective for better strategies to prevent IBD.
Collapse
Affiliation(s)
- Xiaoying Lin
- School of Public Health, Nanjing Medical University, Nanjing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mingchao Xu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Dalong Hu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Suping Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuwei Zhang
- School of Public Health, Nanjing Medical University, Nanjing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yao Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| | - Liyun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang, China
| | - Jianguo Xu
- School of Public Health, Nanjing Medical University, Nanjing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
40
|
Lai Y, Shen H, Wang S, Ouyang Y, Zhang X, Hu B, Zhang X, Li G, Xu L, Zhao J. Hydrogel-Transformable Probiotic Powder for Targeted Eradication of Helicobacter pylori with Enhanced Gastric Mucosal Repair and Microbiota Preservation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500478. [PMID: 40091425 DOI: 10.1002/advs.202500478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Lactobacillus reuteri (L. reuteri) therapies represent a potentially effective approach to eradicating Helicobacter pylori (H. pylori). However, the difficulty in bacterial viability preservation and harsh gastric environment compromises the survival and on-target delivery of L. reuteri. This study presents a novel bacterium-mediated bacterial elimination strategy using an edible L. reuteri@HTP probiotic powder for targeted bacterial elimination. The probiotic powder is obtained by grinding a lyophilized hydrogel composed of L. reuteri, hyaluronic acid (HA), tannic acid (TA), and polyvinyl alcohol (PVA). Upon contact with water, the powder quickly transforms into a hydrogel, enhancing L. reuteri's survival in the harsh gastric environment and ensuring selective release at H. pylori-infected inflammatory sites. L. reuteri targets and reduces H. pylori colonization while secreting reuterin to eliminate the bacteria. Additionally, TA's antioxidant properties help alleviate inflammation, and HA supports gastric mucosal repair. L. reuteri@HTP powder preserves the integrity of the gut microbiota, facilitating the restoration of a healthy microbiome. In particular, the probiotic powder remains stable at room temperature for at least six months, providing a promising alternative to traditional antibiotics for H. pylori treatment. This strategy combines targeted eradication, mucosal healing, and microbiome restoration, offering a new approach to treating gastric infections.
Collapse
Affiliation(s)
- Yongkang Lai
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, P. R. China
| | - Hanchun Shen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
| | - Yongliang Ouyang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Xinyuan Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Bin Hu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Xiaoyi Zhang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
| | - Guisheng Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Lizhi Xu
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
- Materials Innovation Institute for Life Sciences and Energy (MILES), The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, 518057, P. R. China
| | - Jiulong Zhao
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, P. R. China
| |
Collapse
|
41
|
Sun J, Song S, Liu J, Chen F, Li X, Wu G. Gut microbiota as a new target for anticancer therapy: from mechanism to means of regulation. NPJ Biofilms Microbiomes 2025; 11:43. [PMID: 40069181 PMCID: PMC11897378 DOI: 10.1038/s41522-025-00678-x] [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: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
In order to decipher the relationship between gut microbiota imbalance and cancer, this paper reviewed the role of intestinal microbiota in anticancer therapy and related mechanisms, discussed the current research status of gut microbiota as a biomarker of cancer, and finally summarized the reasonable means of regulating gut microbiota to assist cancer therapy. Overall, our study reveals that the gut microbiota can serve as a potential target for improving cancer management.
Collapse
Affiliation(s)
- Jiaao Sun
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shiyan Song
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiahua Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Feng Chen
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Xiaorui Li
- Department of oncology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
42
|
Zonjić J, Karačić A, Brodić I, Starčević A, Renko I, Krznarić Ž, Ivančić M, Liberati Pršo AM, Šatalić Z. The Short- and Long-Term Effects of a Short Course of Sauerkraut Supplementation on the Gut Microbiota of Active Athletes: A Pilot Follow-Up Study. Nutrients 2025; 17:929. [PMID: 40077799 PMCID: PMC11901685 DOI: 10.3390/nu17050929] [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: 02/01/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Objectives: The application of whole fermented foods in sports nutrition for gut microbiota personalization is a promising area of investigation. Sauerkraut, a traditional fermented food, has not been extensively studied in this context. This study aimed to replicate earlier findings of a proof-of-concept study on the potential of sauerkraut for gut microbiota personalization in active athletes. Methods: A pilot follow-up study was conducted on active recreational athletes who consumed 250 g of organic pasteurized sauerkraut daily for 10 days. Changes in gut microbiota composition and functionality were assessed using 16S rRNA sequencing and metabolic pathway analysis across three time points: pre-intervention, postintervention, and one-month postintervention. Laboratory parameters, bowel function, and side effects were monitored throughout the study. Results: In total, 11 male participants with an average age of 30 years completed the study. The pilot follow-up study did not replicate the original study's findings on sauerkraut's short-term effects on β-diversity and taxonomic and functional groups. However, long-term effects of sauerkraut supplementation were demonstrated, including a significant reduction in α-diversity variance and increased gut microbiota composition similarity (β-diversity) as well as several significant changes in bacterial taxa and metabolic pathways after a washout period. The intervention also induced a transient decrease in B12 vitamin levels and a short- and long-term increase in leukocyte concentrations. The probability for physiological types of stools increased after one week of sauerkraut supplementation as well as the incidence of gastrointestinal side effects, such as bloating, diarrhea, pain, nausea, and constipation. Conclusions: This study suggests that the short-term effects on the gut microbiota of sauerkraut supplementation depend on its baseline status, but it can induce long-term effects. Sauerkraut supplementation requires a seven-day adaptation period. Further research is needed to explore the mechanisms behind the short- and long-term effects of sauerkraut supplementation.
Collapse
Affiliation(s)
- Jadran Zonjić
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
| | - Andrija Karačić
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
- The Gut Microbiome Center (CCM), Jablanska 82, 10000 Zagreb, Croatia
- Department of Internal Medicine, University Hospital “Sveti Duh”, Sveti Duh 64, 10000 Zagreb, Croatia;
| | - Ivona Brodić
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
| | - Antonio Starčević
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
| | - Ira Renko
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
| | - Željko Krznarić
- Department of Internal Medicine, Faculty of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Matija Ivančić
- Department of Internal Medicine, University Hospital “Sveti Duh”, Sveti Duh 64, 10000 Zagreb, Croatia;
| | - Ana-Marija Liberati Pršo
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
- Department of Internal Medicine, University Hospital “Sveti Duh”, Sveti Duh 64, 10000 Zagreb, Croatia;
| | - Zvonimir Šatalić
- Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (J.Z.); (A.K.); (I.B.); (A.S.); (I.R.); (Z.Š.)
| |
Collapse
|
43
|
Yang Y, Zhang Y, Sun R, Du W, Liu Y, Zheng L, Ren Z, Li MD, Xu J. Preclinical Safety Assessment of the Oral Administration of Lactobacillus plantarum GUANKE in Animal Models. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10498-2. [PMID: 40032753 DOI: 10.1007/s12602-025-10498-2] [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: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Probiotics have a long history as fermented food or food supplements. The health benefits and safety profiles of probiotics are strain-specific and should be evaluated individually. The aim of this study was to assess the safety of the Lactobacillus plantarum GUANKE (GUANKE) strain by conducting pharmacological studies, oral toxicity assessments, and investigating the colonization and translocation of GUANKE in experimental animal models. Three pharmacological studies were conducted to examine the effects of oral administration of GUANKE on gastric emptying, bile secretion, and gastric juice secretion. In an acute toxicity study, rats were orally administrated with different doses of GUANKE and monitored for 14 days. In the subacute toxicity study, both rats and beagles were administrated with varying doses of GUANKE for 28 consecutive days to evaluate hematologic, biochemical, and histological effects. The results showed that GUANKE administration did not result in any adverse effect on hematological parameters, biochemical parameters, urinary parameters, and organ indices. Importantly, no translocation of GUANKE to extra-intestinal organs or blood was observed following administration of the CFDA-SE labeled strain. In summary, this study demonstrated the safety of GUANKE intake, which encourages its potential application as a probiotic in clinical trials.
Collapse
Affiliation(s)
- Yuewen Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Beijing, 102206, China
| | - Yanlin Zhang
- JOINN Laboratories (China) Co. Ltd, Beijing Economic-Technological Development Area, No. 5, Rongjingdong Street, Beijing, 100176, China
| | - Ruixiang Sun
- Maiyata Research Institute for Beneficial Bacteria, Shaoxing, Zhejiang, China
| | - Wenjuan Du
- Maiyata Research Institute for Beneficial Bacteria, Shaoxing, Zhejiang, China
| | | | - Lijun Zheng
- Guangzhou Zhiyi Biotech Inc., Guangzhou, China
| | - Zhihong Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Beijing, 102206, China.
| | - Ming Ding Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Jianguo Xu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Beijing, 102206, China.
| |
Collapse
|
44
|
Mahamud AGMSU, Tanvir IA, Kabir ME, Samonty I, Chowdhury MAH, Rahman MA. Gerobiotics: Exploring the Potential and Limitations of Repurposing Probiotics in Addressing Aging Hallmarks and Chronic Diseases. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10501-w. [PMID: 40029460 DOI: 10.1007/s12602-025-10501-w] [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: 02/20/2025] [Indexed: 03/05/2025]
Abstract
As unhealthy aging continues to rise globally, there is a pressing need for effective strategies to promote healthy aging, extend health span, and address aging-related complications. Gerobiotics, an emerging concept in geroscience, offers a novel approach to repurposing selective probiotics, postbiotics, and parabiotics to modulate key aging processes and enhance systemic health. This review explores recent advancements in gerobiotics research, focusing on their role in targeting aging hallmarks, regulating longevity-associated pathways, and reducing risks of multiple age-related chronic conditions. Despite their promise, significant challenges remain, including optimizing formulations, ensuring safety and efficacy across diverse populations, and achieving successful clinical translation. Addressing these gaps through rigorous research, well-designed clinical trials, and advanced biotechnologies can establish gerobiotics as a transformative intervention for healthy aging and chronic disease prevention.
Collapse
Affiliation(s)
| | | | - Md Ehsanul Kabir
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Ismam Samonty
- Department of Agricultural Chemistry, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Md Anamul Hasan Chowdhury
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Md Ashikur Rahman
- Department of Food Safety and Regulatory Science, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| |
Collapse
|
45
|
Pammi M, Preidis GA. How accurate are labels of probiotic products marketed to infants? Pediatr Res 2025:10.1038/s41390-025-03971-9. [PMID: 40025117 DOI: 10.1038/s41390-025-03971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 03/04/2025]
Affiliation(s)
- Mohan Pammi
- Department of Pediatrics, Division of Neonatology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.
| | - Geoffrey A Preidis
- Department of Pediatrics, Division of Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
- USDA/ARS Children's Nutrition Research Center, Houston, TX, USA
| |
Collapse
|
46
|
Younge N, Patel RM. Probiotics and the Risk of Infection. Clin Perinatol 2025; 52:87-100. [PMID: 39892956 PMCID: PMC11789005 DOI: 10.1016/j.clp.2024.10.006] [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] [Indexed: 02/04/2025]
Abstract
Probiotic use has increased in preterm infants and may reduce the risk of necrotizing enterocolitis. Probiotic-associated infection is a concern for infants receiving probiotic supplementation in the neonatal intensive care unit, as highlighted by a recent case and subsequent action by the United States Food and Drug Administration. Based on reports to date, invasive infection is an infrequent but known risk of probiotic supplementation. In this article, we discuss the epidemiology and pathophysiology of invasive infection in preterm infants, review the benefits and risks of probiotic as regulations and available products continue to evolve.
Collapse
Affiliation(s)
- Noelle Younge
- Department of Pediatrics, Duke University, 2400 Pratt Street, DUMC Box 2739, Durham, NC 27705, USA
| | - Ravi M Patel
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Road NE, CL.06323, Atlanta, GA 30329, USA.
| |
Collapse
|
47
|
Patki A, Kunjimoideen K, Sawankar S, Tyagi R, Hegde V, Budi J. Expert Opinion on the Use of Probiotics in Recurrent Pregnancy Loss. Cureus 2025; 17:e81056. [PMID: 40271290 PMCID: PMC12015142 DOI: 10.7759/cureus.81056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2025] [Indexed: 04/25/2025] Open
Abstract
Recurrent pregnancy loss (RPL) involves multiple consecutive miscarriages in early pregnancy, affecting a significant number of Indian women and placing substantial physical and emotional stress on expecting couples. This expert consensus aims to highlight probiotics as a promising option for enhancing fertility and supporting successful pregnancy outcomes, offering hope to individuals and couples affected by RPL. A group of fourteen experts with diverse expertise in gynecology, obstetrics, and fertility from across India gathered between June 29 and June 30, 2024. According to the experts, advanced maternal age emerges as an independent risk factor for miscarriage, with increased risks among older Indian women. The major contributors to RPL include thyroid disease and polycystic ovarian disease. Experts emphasize that the vaginal microbiome dysbiosis, characterized by the reduced dominance of Lactobacilli, is associated with adverse pregnancy outcome, such as preterm birth, early pregnancy loss, and increased events of RPL. Oral probiotic supplementation, particularly strains like L. acidophilus and L. rhamnosus, may improve embryo implantation, reduce miscarriage risk, and support pregnancy maintenance. A healthy lifestyle choice and minimal use of antibiotics are important in creating a positive reproductive outcome. The present expert opinion supports the potential benefits of probiotics, particularly Lactobacillus species, in managing RPL and improving reproductive outcomes. By promoting a balanced microbiota, reducing inflammation, and modulating immune responses, probiotics may play a critical role in enhancing reproductive success.
Collapse
Affiliation(s)
- Ameet Patki
- Obstetrics and Gynecology, Indian Society for Assisted Reproduction (ISAR), Mumbai, IND
| | - K Kunjimoideen
- Obstetrics and Gynecology, Asian Reproductive Medicine Centre, Kochi, IND
| | - Sheetal Sawankar
- Obstetrics and Gynecology, Avisa IVF and Fertility Center, Mumbai, IND
| | - Rajul Tyagi
- Obstetrics and Gynecology, Javitri Hospital and Test Tube Baby Centre, Lucknow, IND
| | - Vandana Hegde
- Obstetrics and Gynecology, Hegde Fertility, Hyderabad, IND
| | - Jyoti Budi
- Obstetrics and Gynecology, Ferty9 Fertility Center, Hyderabad, IND
| |
Collapse
|
48
|
Yang M, Zhong P, Wei P. Living Bacteria: A New Vehicle for Vaccine Delivery in Cancer Immunotherapy. Int J Mol Sci 2025; 26:2056. [PMID: 40076679 PMCID: PMC11900161 DOI: 10.3390/ijms26052056] [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: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer vaccines, aimed at evolving the human immune system to eliminate tumor cells, have long been explored as a method of cancer treatment with significant clinical potential. Traditional delivery systems face significant challenges in directly targeting tumor cells and delivering adequate amounts of antigen due to the hostile tumor microenvironment. Emerging evidence suggests that certain bacteria naturally home in on tumors and modulate antitumor immunity, making bacterial vectors a promising vehicle for precision cancer vaccines. Live bacterial vehicles offer several advantages, including tumor colonization, precise drug delivery, and immune stimulation, making them a compelling option for cancer immunotherapy. In this review, we explore the mechanisms of action behind living bacteria-based vaccines, recent progress in popular bacterial chassis, and strategies for specific payload delivery and biocontainment to ensure safety. These approaches will lay the foundation for developing an affordable, widely applicable cancer vaccine delivery system. This review also discusses the challenges and future opportunities in harnessing bacterial-based vaccines for enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
| | | | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (M.Y.); (P.Z.)
| |
Collapse
|
49
|
Tang Y, Zhang Y, Chen C, Cao Y, Wang Q, Tang C. Gut microbiota: A new window for the prevention and treatment of neuropsychiatric disease. J Cent Nerv Syst Dis 2025; 17:11795735251322450. [PMID: 39989718 PMCID: PMC11846125 DOI: 10.1177/11795735251322450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
Under normal physiological conditions, gut microbiota and host mutually coexist. They play key roles in maintaining intestinal barrier integrity, absorption, and metabolism, as well as promoting the development of the central nervous system (CNS) and emotional regulation. The dysregulation of gut microbiota homeostasis has attracted significant research interest, specifically in its impact on neurological and psychiatric disorders. Recent studies have highlighted the important role of the gut- brain axis in conditions including Alzheimer's Disease (AD), Parkinson's Disease (PD), and depression. This review aims to elucidate the regulatory mechanisms by which gut microbiota affect the progression of CNS disorders via the gut-brain axis. Additionally, we discuss the current research landscape, identify gaps, and propose future directions for microbial interventions against these diseases. Finally, we provide a theoretical reference for clinical treatment strategies and drug development for AD, PD, and depression.
Collapse
Affiliation(s)
- Yali Tang
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Yizhu Zhang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Chen Chen
- Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ying Cao
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, People’s Republic of China
| | - Qiaona Wang
- School of Ecology and Applied Meteorology, Nanjing University of Information Science & Technology, Nanjing, People’s Republic of China
| | - Chuanfeng Tang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
50
|
Zhang Y, Xu Y, Hu L, Wang X. Advancements related to probiotics for preventing and treating recurrent respiratory tract infections in children. Front Pediatr 2025; 13:1508613. [PMID: 39981209 PMCID: PMC11839809 DOI: 10.3389/fped.2025.1508613] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Recurrent respiratory tract infections (RRTIs) are a common condition in pediatrics and significantly impact children's quality of life; however, their pathogenesis and contributing factors are not yet fully elucidated. Probiotics have recently emerged as promising agents for modulating intestinal microecology and have gained considerable attention in clinical research on preventing and treating RRTIs in children. This article provides an initial overview of the concept, classification, and mechanisms underlying probiotics. It emphasizes their beneficial effects on respiratory health by modulating intestinal microbial equilibrium, augmenting immune system functionality, and attenuating inflammatory responses. Subsequently, we examine existing research regarding the use of probiotics in pediatric RRTIs. Numerous clinical trials have unequivocally demonstrated that supplementing with probiotics can significantly reduce both the frequency and severity of RRTIs in children while also simultaneously decreasing antibiotic usage. However, there are ongoing controversies and challenges in current research concerning the influence of probiotic type, dosage, duration of use, and other factors on efficacy. Furthermore, variations have been observed across different studies. Additionally, it is crucial to further evaluate the safety and potential long-term side effects associated with probiotic use in children with RRTIs. In conclusion, we propose future research directions including conducting more high-quality randomized controlled trials to optimize application strategies for probiotics alongside other treatments while considering variations based on age and health conditions among pediatric populations. Finally, in summary although probiotics exhibit promising benefits in preventing and treating RRTIs in children; additional studies are necessary to refine their application strategies ensuring both safety and effectiveness.
Collapse
Affiliation(s)
- Yali Zhang
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yingying Xu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ling Hu
- Tianyou Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xiaomei Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|