1
|
Bonazzi E, De Barba C, Lorenzon G, Maniero D, Bertin L, Barberio B, Facciotti F, Caprioli F, Scaldaferri F, Zingone F, Savarino EV. Recent developments in managing luminal microbial ecology in patients with inflammatory bowel disease: from evidence to microbiome-based diagnostic and personalized therapy. Expert Rev Gastroenterol Hepatol 2025:1-14. [PMID: 40247656 DOI: 10.1080/17474124.2025.2495087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/21/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic condition characterized by abnormal immune responses and intestinal inflammation. Emerging evidence highlights the vital role of gut microbiota in IBD's onset and progression. Recent advances have shaped diagnostic and therapeutic strategies, increasingly focusing on microbiome-based personalized care. Methodology: this review covers studies from 2004 to 2024, reflecting the surge in research on luminal microbial ecology in IBD. Human studies were prioritized, with select animal studies included for mechanistic insights. Only English-language, peer-reviewed articles - clinical trials, systematic reviews, and meta-analyses - were considered. Studies without clinical validation were excluded unless offering essential insights. Searches were conducted using PubMed, Scopus, and Web of Science. AREAS COVERED we explore mechanisms for managing IBD-related microbiota, including microbial markers for diagnosis and novel therapies such as fecal microbiota transplantation, metabolite-based treatments, and precision microbiome modulation. Additionally, we review technologies and diagnostic tools used to analyze gut microbiota composition and function in clinical settings. Emerging data supporting personalized therapeutic strategies based on individual microbial profiles are discussed. EXPERT OPINION Standardized microbiome research integration into clinical practice will enhance precision in IBD care, signaling a shift toward microbiota-based personalized medicine.
Collapse
Affiliation(s)
- Erica Bonazzi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Caterina De Barba
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Greta Lorenzon
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Daria Maniero
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Luisa Bertin
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Gastroenterology Unit, Azienda Ospedale-Università Padova, Padua, Italy
| | - Brigida Barberio
- Gastroenterology Unit, Azienda Ospedale-Università Padova, Padua, Italy
| | - Federica Facciotti
- INGM-National Institute of Molecular Genetics 'Romeo ed Enrica Invernizzi', Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Franco Scaldaferri
- Department of Gastroenterological Area, "A. Gemelli" Hospital, Catholic University of the Sacred Heart, Rome, Italy
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Gastroenterology Unit, Azienda Ospedale-Università Padova, Padua, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Gastroenterology Unit, Azienda Ospedale-Università Padova, Padua, Italy
| |
Collapse
|
2
|
Kamlárová A, Kvaková M, Ambro Ľ, Link R, Bertková I, Hertelyová Z, Janíčko M, Hijová E, Štofilová J. Improvement of the inflammation-damaged intestinal barrier and modulation of the gut microbiota in ulcerative colitis after FMT in the SHIME® model. BMC Complement Med Ther 2025; 25:145. [PMID: 40259351 PMCID: PMC12013018 DOI: 10.1186/s12906-025-04889-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 04/09/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) seems to be a promising approach in ulcerative colitis (UC) management with the aim of repopulating a patient's dysbiotic microbiota with beneficial bacteria and restore its metabolic activity to its healthy characteristics. Metabolites present after FMT may improve the function and integrity of the intestinal barrier, reduce inflammation, and thus induce remission in an UC patient. In this study we evaluated whether the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®) model may be a suitable non-invasive alternative for studying and modifying the dysbiotic microbiota in UC by FMT application. METHODS SHIME® model was used to investigate microbial and metabolic changes in the gut microbiota of UC patient induced by FMT application. FMT-modified metabolites from SHIME® were applied to an in vitro model of the intestinal barrier (differentiated Caco-2 and HT-29-MTX-E12 cell lines) compromised by pro-inflammatory cytokines to study the effect of FMT on the intestinal barrier. RESULTS Qualitative and quantitative microbial analyses showed that FMT increased the diversity and variability of the microbiota in UC patient associated with a significant increase in total bacteria, Bacteroidota and Lactobacillus, as well as an increase in butyrate levels. In addition, an increase in the relative abundance of some important species such as Faecalibacterium prausnitzii and Bifidobacterium longum was observed, and there was also an enrichment of the microbiota with new species such as Blautia obeum, Roseburia faecis, Bifidobacterium adolescentis, Fusicatenibacter saccharivorans and Eubacterium rectale. Furthermore, microbial metabolites modulated by FMT from the SHIME® model prevented intestinal barrier damage and inhibited interleukin 8 (IL-8) and monocyte chemoattractant protein 1 (MCP-1) secretion when cell barriers were pretreated with FMT medium for 24 h. In summary, this study confirmed that a single dose of FMT beneficially modulated the composition and metabolic activity of the UC microbiota in the SHIME® model. CONCLUSIONS FMT favorably modulates the gut microbiota of UC patient cultured in the SHIME® model. FMT-modulated SHIME-derived microbial metabolites improve intact and inflamed intestinal barrier properties in vitro. Repeated applications are necessary to maintain the beneficial effect of FMT in SHIME® model.
Collapse
Affiliation(s)
- Anna Kamlárová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Monika Kvaková
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Ľuboš Ambro
- Center for Interdisciplinary Biosciences, Technology and Innovation Park, P.J. Šafárik University, Jesenna 5, Košice, 040 01, Slovakia
| | - René Link
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Izabela Bertková
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Zdenka Hertelyová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Martin Janíčko
- 2nd Department of Internal Medicine, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Emília Hijová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Jana Štofilová
- Center of Clinical and Preclinical Research - MediPark, Faculty of Medicine, P. J. Šafárik University, Trieda SNP 1, Košice, 040 11, Slovakia.
| |
Collapse
|
3
|
Ozma MA, Fadaee M, Hosseini HM, Ataee MH, Mirhosseini SA. A Critical Review of Postbiotics as Promising Novel Therapeutic Agents for Clostridial Infections. Probiotics Antimicrob Proteins 2025; 17:656-667. [PMID: 39546182 DOI: 10.1007/s12602-024-10406-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Clostridial infections, known for their severity and rapid progression, present significant challenges in both clinical and veterinary fields. These bacteria, which can survive without oxygen and produce protective spores, cause many diseases, ranging from simple gastrointestinal disorders to severe and potentially fatal infections including botulism, tetanus, and gas gangrene. The rising occurrence of antibiotic-resistant strains and the repetitive character of some Clostridial illnesses, including Clostridioides difficile infections (CDI), highlight the immediate need for alternate treatment approaches. Postbiotics, which are metabolites derived from probiotics, are showing great potential as effective agents against these diseases. The current study offers a comprehensive investigation of the potential of postbiotics as therapeutic agents for treating Clostridial infections, including C. difficile, Clostridium perfringens, Clostridium botulinum, and Clostridium tetani. It also examines the processes by which postbiotics exert their effects. Preliminary investigations have shown that postbiotics have promising antibacterial and antibiofilm properties, indicating their potential as adjunct agents in methods for controlling microbial growth. Nevertheless, more study is required to thoroughly demonstrate their medicinal uses.
Collapse
Affiliation(s)
- Mahdi Asghari Ozma
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ataee
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Goya-Jorge E, Gonza I, Bondue P, Druart G, Al-Chihab M, Boutaleb S, Douny C, Scippo ML, Thonart P, Delcenserie V. Evaluation of Four Multispecies Probiotic Cocktails in a Human Colonic Fermentation Model. Probiotics Antimicrob Proteins 2024; 16:2102-2115. [PMID: 37725305 DOI: 10.1007/s12602-023-10162-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 09/21/2023]
Abstract
Bacteriotherapy represents an attractive approach for both prophylaxis and treatment of human diseases. However, combining probiotic bacteria in "cocktails" is underexplored, despite its potential as an alternative multi-target therapy. Herein, three-strain probiotic mixtures containing different combinations of Bacillus (Bc.) coagulans [ATB-BCS-042], Levilactobacillus (Lv.) brevis [THT 0303101], Lacticaseibacillus (Lc.) paracasei [THT 031901], Bacillus subtilis subsp. natto [ATB-BSN-049], Enterococcus faecium [ATB-EFM-030], and Bifidobacterium (Bf.) animalis subsp. lactis [THT 010802] were prepared. Four cocktails (PA: Bc. coagulans + Lv. brevis + Lc. paracasei, PB: Bc. subtilis subsp. natto + Lv. brevis + Lc. paracasei, PC: E. faecium + Lv. brevis + Lc. paracasei, PD: Bc. coagulans + Lv. brevis + Bf. animalis subsp. lactis) were tested using a short-term (72 h) simulation of the human colonic microbiota in a final dose of 6 × 109 CFU. All these probiotic mixtures significantly increased butyrate production compared to the parallel control experiment. PA and PB promoted a bifidogenic effect and facilitated lactobacilli colonization. Furthermore, reporter gene assays using the AhR_HT29-Lucia cell line revealed that fermentation supernatants from PA and PB notably induced AhR transactivity. Subsequent examination of the metabolic outputs of PA and PB in intestinal epithelial models using cell culture inserts suggested no significant impact on the transepithelial electrical resistance (TEER). Assessment of the expression of proinflammatory and anti-inflammatory cytokines, as well as AhR-related target genes in the Caco-2 cell monolayers indicated that PB's metabolic output upregulated most of the measured endpoints. This in vitro investigation evaluated the potential impact of four multispecies probiotic mixtures in the human colonic microbiota and identified a promising formulation comprising a combination of Bc. subtilis subsp. natto, Lv. brevis, and Lc. paracasei as a promising formulation for further study.
Collapse
Affiliation(s)
- Elizabeth Goya-Jorge
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Irma Gonza
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Pauline Bondue
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Germain Druart
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Mohamed Al-Chihab
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Samiha Boutaleb
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Caroline Douny
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Marie-Louise Scippo
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Philippe Thonart
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Véronique Delcenserie
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium.
| |
Collapse
|
5
|
Bahrami S, Babaei N, Esmaeili Gouvarchin Ghaleh H, Mohajeri Borazjani J, Farzanehpour M. Investigating the effects of combined treatment of mesalazine with Lactobacillus casei in the experimental model of ulcerative colitis. Front Mol Biosci 2024; 11:1456053. [PMID: 39421689 PMCID: PMC11484277 DOI: 10.3389/fmolb.2024.1456053] [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: 07/04/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Ulcerative colitis (UC), a common gastrointestinal disorder in affluent nations, involves chronic intestinal mucosal inflammation. This research investigated the effects of combined probiotic treatment of Lactobacillus casei (L. casei) and mesalazine on disease activity index and inflammatory factors in the UC model. Methods 20 male BALB/c mice were utilized and divided into four groups. To induce UC, all groups received 100 μL of 4% acetic acid (AA) intra-rectally. The first group received phosphate-buffered saline (PBS) (as a control group), the second group was treated with L. casei, the third group was treated with mesalazine and, the fourth group was treated with L. casei and mesalazine. Treatment with L. Casei and mesalazine commenced after the manifestation of symptoms resulting from UC induction. Finally, the mice were euthanized and the disease activity index, myeloperoxidase activity, nitric oxide rate, cytokines level (IL-1β, IL-6, TNF-α) and, gene expression (iNOS, COX-2, and cytokines) were evaluated. Results The combined treatment of L. casei and mesalazine led to a significant decrease in the levels of NO, MPO and inflammatory cytokines. In addition, the expression of cytokines, iNOS and COX-2 genes decreased in mice treated with the combination. Discussion This study shows that combined treatment of L. casei and mesalazine improves of experimental UC, which can be attributed to the anti-inflammatory properties of L. casei and mesalazine. In conclusion, this combination therapy can be considered a suitable option for the management of UC.
Collapse
Affiliation(s)
- Shabnam Bahrami
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Nahid Babaei
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jaleh Mohajeri Borazjani
- Department of Fisheries and Natural Resources, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Mahdieh Farzanehpour
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhang Y, Wang Y, Zhang X, Wang P, Shi F, Zhang Z, Wang R, Wu D, She J. Gastrointestinal Self-Adaptive and Nutrient Self-Sufficient Akkermansia muciniphila-Gelatin Porous Microgels for Synergistic Therapy of Ulcerative Colitis. ACS NANO 2024; 18:26807-26827. [PMID: 39301762 DOI: 10.1021/acsnano.4c07658] [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: 09/22/2024]
Abstract
To realize effective and long-term synergistic therapy of ulcerative colitis (UC) with probiotics, we developed gastrointestinal self-adaptive and nutrient self-sufficient Akkermansia muciniphila (AKK)-gelatin porous microgels (AKK@GPMGs). In AKK@GPMGs, AKK was covered with sequential layers of proanthocyanidins (PAs), mucin (MUC), and phosphatidylcholine (PC) to obtain AKK@PAs-MUC-PC (AKK@PMP), and then encapsulated within the methacrylate-modified gelatin porous microgels. AKK@GPMGs provide sufficient mucus as a nutrition source for AKK and boost resistance to stomach acid by 30.49-fold, and colonization in the intestines is enhanced by 83.46 times. The microgels can be dissociated by matrix metalloproteinase at the inflammatory sites of the intestine, and release AKK@PMP, which acts as "band-aid" that adheres to the inflamed colon for a long time and offers improved synergistic therapy for UC. Compared to uncoated AKK, AKK@GPMGs increase reactive oxygen species scavenging capacity by 26.47 times, improve the intestinal mucus layer thickness by 5.63 times, increase the goblet cells abundance by 3.93 times, reduce intestinal permeability by 5.60 times and significantly enhance beneficial gut microbiota while repressing harmful microbiota. These results indicate that AKK@GPMGs can restore mucus layer and tight junction integrity, reduce inflammation and oxidative stress, and regulate gut microbiota homeostasis to effectively treat intestinal inflammation.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Pengqian Wang
- Department of Chemical Engineering, School of Water and Environment, Chang'an University, Xi'an 710064, P.R. China
| | - Feiyu Shi
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
| | - Ruochen Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P.R. China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
7
|
Sun R, Du S, Wang M, Chen Z, Yan Q, Yuan B, Jin Y. Colonic long-term retention and colonization of probiotics by double-layer chitosan/tannic acid coating and microsphere embedding for treatment of ulcerative colitis and radiation enteritis. Int J Biol Macromol 2024; 280:135757. [PMID: 39299414 DOI: 10.1016/j.ijbiomac.2024.135757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Oral probiotics can alleviate enteric inflammations but their rapid transit through the gut limits their retention and colonization in the colon. Here, a novel strategy integrating the bacterial double-layer coating and hydrogel microsphere embedding techniques was used to highly enhance the colonic retention and colonization efficiency of Lactobacillus rhamnosus GG (LGG). LGG was coated by the double layers of chitosan (CS) and tannic acid (TA), and then embedded in calcium alginate (CA) hydrogel microspheres to form LGG@CT@CA. The microspheres resisted gastric liquids, improving LGG safe transit through the stomach to reach the colon. LGG@CT rapidly released in the colon due to the good swelling of hydrogel microspheres. More importantly, LGG exhibited long-term retention up to 7 days in the colon, and colonized the deep site of the colonic mucosa. LGG@CT@CA had a high therapeutic efficiency of ulcer colitis with the long colon and the low intestinal permeability of colonic tissues. LGG@CT@CA also alleviated the small intestinal damage induced by irradiation and the survival rates were improved. The mechanisms included local ROS decrease, IL-10 increase, and ferroptosis reduction in the small intestine. The oral colon-targeted system holds promise for oral probiotic therapy by the long-term retention and colonization in the colon.
Collapse
Affiliation(s)
- Rui Sun
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Shumin Du
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Minting Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Qiucheng Yan
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China.
| |
Collapse
|
8
|
Ottria R, Xynomilakis O, Casati S, Ciuffreda P. Pre- to Postbiotics: The Beneficial Roles of Pediatric Dysbiosis Associated with Inflammatory Bowel Diseases. Microorganisms 2024; 12:1582. [PMID: 39203424 PMCID: PMC11356122 DOI: 10.3390/microorganisms12081582] [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: 06/28/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Probiotics are "live microorganisms which, when administered in adequate amount, confer health benefits on the host". They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.
Collapse
Affiliation(s)
- Roberta Ottria
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, 20157 Milan, Italy; (O.X.); (S.C.); (P.C.)
| | | | | | | |
Collapse
|
9
|
Wang X, Zhou C, Zhang S, Ma Y, Xiao W, Guo Y. Additive efficacy and safety of probiotics in the treatment of ulcerative colitis: a systematic review and meta-analysis. Eur J Nutr 2024; 63:1395-1411. [PMID: 38446227 DOI: 10.1007/s00394-023-03307-5] [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: 07/11/2023] [Accepted: 12/08/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND We aim to report the latest pooled analyses to evaluate the additive efficacy and safety of probiotics in the treatment of ulcerative colitis (UC). METHODS We systematically searched the relevant literature investigating the efficacy and/or safety of probiotics in patients with UC from PubMed, Embase and Web of Science up to January 2023. Two researchers independently screened the literature, extracted data, and evaluated the quality of the included studies according to the inclusion and exclusion criteria. Any discrepancies throughout these processes were solved by consensus. All statistical analyses were performed by Review Manager version 5.4 and Stata version 15.0. RESULTS A total of 13 articles were included in the pooled analyses, and the studies were all randomized controlled trials with a total of 930 patients. There were no significant differences between the probiotics and placebo groups concerning demographic and baseline characteristics. For patients with active UC, the probiotic group boosted the remission rate by 87% compared to the placebo group, but failed to reach a statistical difference (OR: 1.87; 95% CI 0.98, 3.57; P = 0.06, I2 = 67%); furthermore, there were no statistical differences in maintenance of clinical remission, clinical response, change in UCDAI scores, or mucosal healing outcomes in the probiotic group compared to the placebo group. For patients in clinical remission, the clinical relapse rates were significantly lower in the probiotic group than in the placebo group (OR: 0.34; 95% CI 0.14, 0.79; P = 0.01). Moreover, this study did not observe a significant difference between the two groups for general adverse events rate (OR: 1.98; 95% CI 0.69, 5.68; P = 0.20). CONCLUSION Probiotic-assisted therapy may be effective in inhibiting UC recurrence in patients in clinical remission without increasing the risk of treatment-related adverse events; furthermore, probiotics may increase the rate of clinical remission in patients with active UC. However, caution is needed when interpreting the clinical efficacy of probiotics in improving the clinical outcome of patients with active UC.
Collapse
Affiliation(s)
- Xinyue Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Chunyu Zhou
- Medical Management Office of Beijing University of Chinese Medicine, Beijing, China.
| | - Shaohui Zhang
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yixiang Ma
- Beijing University of Chinese Medicine, Beijing, China
| | - Wenqin Xiao
- Beijing University of Chinese Medicine, Beijing, China
| | - Yanmei Guo
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Ebrahiminejad A, Sepahi AA, Yadegar A, Meyfour A. Pasteurized form of a potential probiotic lactobacillus brevis IBRC-M10790 exerts anti-inflammatory effects on inflammatory bowel disease in vitro. BMC Complement Med Ther 2024; 24:258. [PMID: 38987744 PMCID: PMC11234635 DOI: 10.1186/s12906-024-04576-1] [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: 03/16/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal system. So far, no treatment has been identified that can completely cure IBD. Lactobacillus brevis is hypothesized to be beneficial in preventing inflammation. This study aimed to evaluate the potential probiotic effects of live and pasteurized L. brevis IBRC-M10790 on the in vitro cell co-culture model of IBD. METHODS An in vitro intestinal model was established using a transwell co-culture system of Caco-2 intestinal epithelial cells and RAW264.7 macrophages. Inflammatory conditions were induced in RAW264.7 cells using lipopolysaccharide. The effects of live and pasteurized L. brevis IBRC-M10790 on inflammatory mediators and epithelial barrier markers were investigated. RESULTS L. brevis IBRC-M10790 was able to significantly decrease the proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and increase the anti-inflammatory cytokine (IL-10) in the in vitro co-culture system. In addition, L. brevis increased adherens and tight junction (TJ) markers (ZO-1, E-cadherin, and Occludin) in Caco-2 intestinal epithelial cells. Based on the results, pasteurized L. brevis showed a higher protective effect than live L. brevis. CONCLUSIONS Our findings suggest that live and pasteurized forms of L. brevis possess probiotic properties and can mitigate inflammatory conditions in IBD.
Collapse
Affiliation(s)
- Ardeshir Ebrahiminejad
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Abbas Akhavan Sepahi
- Department of Microbiology, Faculty of Biological Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
11
|
Štofilová J, Kvaková M, Kamlárová A, Hijová E, Bertková I, Guľašová Z. Correction: Štofilová et al. Probiotic-Based Intervention in the Treatment of Ulcerative Colitis: Conventional and New Approaches. Biomedicines 2022, 10, 2236. Biomedicines 2024; 12:797. [PMID: 38672283 PMCID: PMC11048518 DOI: 10.3390/biomedicines12040797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
The authors would like to add the following clarification regarding the clinical trials evaluating the probiotic product VSL#3 cited in the published paper [...].
Collapse
Affiliation(s)
- Jana Štofilová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, Trieda SNP 1, 040 11 Kosice, Slovakia; (M.K.); (A.K.); (E.H.); (I.B.); (Z.G.)
| | | | | | | | | | | |
Collapse
|
12
|
Vesci L, Tundo G, Soldi S, Galletti S, Stoppoloni D, Bernardini R, Modolea AB, Luberto L, Marra E, Giorgi F, Marini S. A Novel Lactobacillus brevis Fermented with a Vegetable Substrate (AL0035) Counteracts TNBS-Induced Colitis by Modulating the Gut Microbiota Composition and Intestinal Barrier. Nutrients 2024; 16:937. [PMID: 38612971 PMCID: PMC11013894 DOI: 10.3390/nu16070937] [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: 02/07/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Crohn's and ulcerative colitis are common conditions associated with inflammatory bowel disease as well as intestinal flora and epithelial barrier dysfunction. A novel fermented Lactobacillus brevis (AL0035) herein assayed in a trinitro benzene sulfonic acid (TNBS)-induced colitis mice model after oral administration significantly counteracted the body weight loss and improves the disease activity index and histological injury scores. AL0035 significantly decreased the mRNA and protein expression of different pro-inflammatory cytokines (TNFalpha, IL-1beta, IL-6, IL-12, IFN-gamma) and enhanced the expression of IL-10. In addition, the probiotic promoted the expression of tight junction proteins, such as ZO-1, keeping the intestinal mucosal barrier function to attenuate colitis symptoms in mice. Markers of inflammation cascade such as myeloperoxidase (MPO) and PPAR-gamma measured in the colon were also modified by AL0035 treatment. AL0035 was also able to reduce different lymphocyte markers' infiltration in the colon (GATA-3, T-Bet, NK1.1) and monocyte chemoattractant protein-1 (MCP-1/CCL2), a key chemokine involved in the migration and infiltration of monocytes/macrophages in the immunological surveillance of tissues and inflammation. In colonic microbiota profile analysis through 16S rRNA sequencing, AL0035 increased the microbial diversity depleted by TNBS administration and the relative abundance of the Lactobacillaceae and Lachnospiraceae families, whereas it decreased the abundance of Proteobacteria. Altogether, these data indicated that AL0035 could lower the severity of colitis induced by TNBS by regulating inflammatory cytokines, increasing the expression of tight junction proteins and modulating intestinal microbiota, thus preventing tissue damage induced by colitis.
Collapse
Affiliation(s)
- Loredana Vesci
- Corporate R&D, Alfasigma S.p.A., Via Pontina km 30.400, Pomezia, 00071 Rome, Italy;
| | - Grazia Tundo
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| | - Sara Soldi
- AAT Advanced Analytical Technologies Srl, Via P. Majavacca 12, 29017 Fiorenzuola d’Arda, Italy; (S.S.); (S.G.)
| | - Serena Galletti
- AAT Advanced Analytical Technologies Srl, Via P. Majavacca 12, 29017 Fiorenzuola d’Arda, Italy; (S.S.); (S.G.)
| | | | - Roberta Bernardini
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
- Centro Interdipartimentale di Medicina Comparata, Tecniche Alternative ed Acquacoltura (CIMETA), University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Anamaria Bianca Modolea
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| | - Laura Luberto
- Takis Castel Romano, 00128 Rome, Italy; (D.S.); (L.L.); (E.M.)
| | - Emanuele Marra
- Takis Castel Romano, 00128 Rome, Italy; (D.S.); (L.L.); (E.M.)
| | - Fabrizio Giorgi
- Corporate R&D, Alfasigma S.p.A., Via Pontina km 30.400, Pomezia, 00071 Rome, Italy;
| | - Stefano Marini
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| |
Collapse
|
13
|
Lau LYJ, Quek SY. Probiotics: Health benefits, food application, and colonization in the human gastrointestinal tract. FOOD BIOENGINEERING 2024; 3:41-64. [DOI: 10.1002/fbe2.12078] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/29/2024] [Indexed: 01/04/2025]
Abstract
AbstractProbiotics have become increasingly popular over the past two decades due to the continuously expanding scientific evidence indicating their beneficial effects on human health. Therefore, they have been applied in the food industry to produce functional food, which plays a significant role in human health and reduces disease risk. However, maintaining the viability of probiotics and targeting the successful delivery to the gastrointestinal tract remain two challenging tasks in food applications. Specifically, this paper reviews the potentially beneficial properties of probiotics, highlighting the use and challenges of probiotics in food application and the associated health benefits. Of foremost importance, this paper also explores the potential underlying molecular mechanisms of the enhanced effect of probiotics on gastrointestinal epithelial cells, including a discussion on various surface adhesion‐related proteins on the probiotic cell surface that facilitate colonization.
Collapse
Affiliation(s)
- Li Ying Jessie Lau
- Food Science, School of Chemical Sciences The University of Auckland Auckland New Zealand
| | - Siew Young Quek
- Food Science, School of Chemical Sciences The University of Auckland Auckland New Zealand
| |
Collapse
|
14
|
Jakubczyk D, Leszczyńska K, Pacyga-Prus K, Kozakiewicz D, Kazana-Płuszka W, Gełej D, Migdał P, Kruszakin R, Zabłocka A, Górska S. What happens to Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with eukaryotic cells? BMC Microbiol 2024; 24:60. [PMID: 38373929 PMCID: PMC10875879 DOI: 10.1186/s12866-023-03179-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND The impact of probiotic strains on host health is widely known. The available studies on the interaction between bacteria and the host are focused on the changes induced by bacteria in the host mainly. The studies determining the changes that occurred in the bacteria cells are in the minority. Within this paper, we determined what happens to the selected Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with the intestinal epithelial layer. For this purpose, we tested the bacteria cells' viability, redox activity, membrane potential and enzymatic activity in different environments, including CaCo-2/HT-29 co-culture, cell culture medium, presence of inflammatory inductor (TNF-α) and oxygen. RESULTS We indicated that the external milieu impacts the viability and vitality of bacteria. Bifidobacterium adolescentis decrease the size of the live population in the cell culture medium with and without TNF-α (p < 0.001 and p < 0.01 respectively). In contrast, Bifidobacterium longum ssp. longum significantly increased survivability in contact with the eukaryotic cells and cell culture medium (p < 0.001). Bifidobacterium adolescentis showed significant changes in membrane potential, which was decreased in the presence of eukaryotic cells (p < 0.01), eukaryotic cells in an inflammatory state (p < 0.01), cell culture medium (p < 0.01) and cell culture medium with TNF-α (p < 0.05). In contrast, Bifidobacterium longum ssp. longum did not modulate membrane potential. Instead, bacteria significantly decreased the redox activity in response to milieus such as eukaryotic cells presence, inflamed eukaryotic cells as well as the culture medium (p < 0.001). The redox activity was significantly different in the cells culture medium vs the presence of eukaryotic cells (p < 0.001). The ability to β-galactosidase production was different for selected strains: Bifidobacterium longum ssp. longum indicated 91.5% of positive cells, whereas Bifidobacterium adolescentis 4.34% only. Both strains significantly reduced the enzyme production in contact with the eukaryotic milieu but not in the cell culture media. CONCLUSION The environmental-induced changes may shape the probiotic properties of bacterial strains. It seems that the knowledge of the sensitivity of bacteria to the external environment may help to select the most promising probiotic strains, reduce research costs, and contribute to greater reproducibility of the obtained probiotic effects.
Collapse
Affiliation(s)
- Dominika Jakubczyk
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | - Katarzyna Leszczyńska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Katarzyna Pacyga-Prus
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Kozakiewicz
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Wioletta Kazana-Płuszka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Gełej
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paweł Migdał
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Roksana Kruszakin
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Agnieszka Zabłocka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
15
|
Ragavan ML, Hemalatha S. The functional roles of short chain fatty acids as postbiotics in human gut: future perspectives. Food Sci Biotechnol 2024; 33:275-285. [PMID: 38222911 PMCID: PMC10786766 DOI: 10.1007/s10068-023-01414-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 01/16/2024] Open
Abstract
The significance of gut microbiome and their metabolites (postbiotics) on human health could be a promising approach to treat various diseases that includes inflammatory bowel diseases, colon cancer, and many neurological disorders. Probiotics with potential mental health benefits (psychobiotics) can alter the gut-brain axis via immunological, humoral, neuronal, and metabolic pathways. Recently, probiotic bacteria like Lactobacillus and Bifidobacterium have been demonstrated for SCFAs production, which play a crucial role in a variety of diseases. These acids could enhance the production of mucins, antimicrobial proteins (bacteriocins and peptides), cytokines (Interleukin 10 and 18) and neurotransmitters (serotonin) in the intestine to main the gut microbiota, intestinal barrier system and other immune functions. In this review, we discuss about two mechanisms such as (i) SCFAs mediated intestinal barrier system, and (ii) SCFAs mediated gut-brain axis to elucidate the therapeutic options for the treatment/prevention of various diseases.
Collapse
Affiliation(s)
| | - S. Hemalatha
- School of Life Sciences, BSACIST, Vandalur, Chennai, Tamil Nadu India
| |
Collapse
|
16
|
Krawczyk A, Gosiewski T, Zapała B, Kowalska-Duplaga K, Salamon D. Alterations in intestinal Archaea composition in pediatric patients with Crohn's disease based on next-generation sequencing - a pilot study. Gut Microbes 2023; 15:2276806. [PMID: 37955638 PMCID: PMC10653639 DOI: 10.1080/19490976.2023.2276806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Intestinal dysbiosis can lead to the induction of systemic immune-mediated inflammatory diseases, such as Crohn's disease Although archaea are part of the commensal microbiota, they are still one of the least studied microorganisms. The aim of our study was the standardization of the optimal conditions and primers for sequencing of the gut archaeome using Next Generation Sequencing, and evaluation of the differences between the composition of archaea in patients and healthy volunteers, as well as analysis of the changes that occur in the archaeome of patients depending on disease activity. Newly diagnosed patients were characterized by similar archeal profiles at every taxonomic level as in healthy individuals (the dominance of Methanobacteria at the class level, and Methanobrevibacter at the genus level). In turn, in patients previously diagnosed with Crohn's disease (both in active and remission phase), an increased prevalence of Thermoplasmata, Thermoprotei, Halobacteria (at the class level), and Halococcus, Methanospaera or Picrophilus (at the genus level) were observed. Furthermore, we have found a significant correlation between the patient's parameters and the individual class or species of Archaea. Our study confirms changes in archaeal composition in pediatric patients with Crohn's disease, however, only in long-standing disease. At the beginning of the disease, the archeal profile is similar to that of healthy people. However, in the chronic form of the disease, significant differences in the composition of archaeome begin to appear. It seems that some archaea may be a good indicator of the chronicity and activity of Crohn's disease.
Collapse
Affiliation(s)
- A. Krawczyk
- Department of Molecular Medical Microbiology, Division of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - T. Gosiewski
- Department of Molecular Medical Microbiology, Division of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - B. Zapała
- Department of Pharmaceutical Microbiology, Jagiellonian University Medical College, Krakow, Poland
- Jagiellonian University Hospital in Krakow, Krakow, Poland
| | - K. Kowalska-Duplaga
- Department of Pediatrics, Gastroenterology and Nutrition,Jagiellonian University Medical College, Krakow, Poland
| | - D. Salamon
- Department of Molecular Medical Microbiology, Division of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
17
|
Khajah MA, Hawai S. Effect of minocycline, methyl prednisolone, or combination treatment on the colonic bacterial population in a state of colonic inflammation using the murine dextran sulfate sodium model. Microb Cell Fact 2023; 22:232. [PMID: 37950185 PMCID: PMC10636938 DOI: 10.1186/s12934-023-02242-8] [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: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Several reports demonstrated anti-inflammatory properties of minocycline in various inflammatory disorders including colitis. We have experimental evidence suggesting synergistic anti-inflammatory effect of minocycline with methyl prednisolone in reducing colitis severity in mice, but if this effect is in part related to modulating the composition of colonic microbiota is still unknown. METHODS the effect of vehicle (V), minocycline (M), methyl prednisolone (MP), or combination (C) regimen on the composition of the microbiota of mice in a state of colon inflammation compared to untreated (UT) healthy mice was determined using 16s metagenomic sequencing, and the taxonomic and functional profiles were summarized. RESULTS Overall, the bacterial flora from the phylum Firmicutes followed by Bacteroidota were found to be predominant in all the samples. However, the composition of Firmicutes was decreased relatively in all the treatment groups compared to UT group. A relatively higher percentage of Actinobacteriota was observed in the samples from the C group. At the genus level, Muribaculaceae, Bacteroides, Bifidobacterium, and Lactobacillus were found to be predominant in the samples treated with both drugs (C). Whereas "Lachnospiraceae NK4A136 group" and Helicobacter in the M group, and Helicobacter in the MP group were found to be predominant. But, in the UT group, Weissella and Staphylococcus were found to be predominant. Eubacterium siraeum group, Clostridia vadinBB60 group, Erysipelatoclostridium and Anaeroplasma genera were identified to have a significant (FDR p < 0.05) differential abundance in V compared to C and UT groups. While at the species level, the abundance of Helicobacter mastomyrinus, Massiliomicrobiota timonensis and uncultured Anaeroplasma were identified as significantly low in UT, C, and M compared to V group. Functional categories related to amino acid, carbohydrate, and energy metabolism, cell motility and cell cycle control were dominated overall across all the samples. Methane metabolism was identified as an enriched pathway. For the C group, "Colitis (decrease)" was among the significant (p = 1.81E-6) associations based on the host-intrinsic taxon set. CONCLUSION Combination regimen of minocycline plus methyl prednisolone produces a synergistic anti-inflammatory effect which is part related to alternation in the colonic microbiota composition.
Collapse
Affiliation(s)
- Maitham A Khajah
- College of Pharmacy, Kuwait University, PO Box 24923, 13110, Safat, Kuwait.
| | - Sanaa Hawai
- College of Pharmacy, Kuwait University, PO Box 24923, 13110, Safat, Kuwait
| |
Collapse
|
18
|
Xu L, Li Y, He Y. The variation characteristics of fecal microbiota in remission UC patients with anxiety and depression. Front Microbiol 2023; 14:1237256. [PMID: 37744915 PMCID: PMC10517179 DOI: 10.3389/fmicb.2023.1237256] [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: 06/12/2023] [Accepted: 08/16/2023] [Indexed: 09/26/2023] Open
Abstract
Background Ulcerative colitis (UC) patients with relapsed disease are most likely to suffer from anxiety and depression. Increasing data indicates that psychological issues can change the composition of intestinal flora. Thus, we aim to seek the variation of intestinal microbiota composition in remission UC patients with anxiety and depression in Northwest China. Results In this study, 45 UC patients in remission were enrolled. The incidence of anxiety was 33.3%, and the prevalence of depression was 22.2%. There was no statistical difference in the alpha diversity of fecal microbiota, while beta diversity had a significant difference between the anxiety group and the non-anxiety group and the depression group and the non-depression group. Species composition analysis results showed that the ratio of Bifidobacterium and Lactobacilales significantly decreased. At the same time, the proportion of Escherichia-Shigella and Proteus_mirabilis increased in the anxiety group, and the ratio of Faecalibacterium and Bifidobacterium significantly decreased. In contrast, Escherichia-Shigella increased in the depression group at the gene levels. Conclusion Anxiety and depression still exist in UC patients even in the remission period. We first identify that the proportion of probiotics decreases while the proportion of pathogens increases in UC patients with anxiety and depression. These findings may provide a new pathophysiological mechanism for the recurrence of disease caused by impaired psychological function and a new method for the treatment strategy of UC patients with psychological issues.
Collapse
Affiliation(s)
- Lingyun Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yingchao Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yingli He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
19
|
Liu C, Ma N, Feng Y, Zhou M, Li H, Zhang X, Ma X. From probiotics to postbiotics: Concepts and applications. ANIMAL RESEARCH AND ONE HEALTH 2023; 1:92-114. [DOI: 10.1002/aro2.7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/24/2023] [Indexed: 01/05/2025]
Abstract
AbstractIn recent years, the important role of gut microbiota in promoting animal health and regulating immune function in livestock and poultry has been widely reported. The issue of animal health problems causes significant economic losses each year. Probiotics and postbiotics have been widely developed as additives due to their beneficial effects in balancing host gut microbiota, enhancing intestinal epithelial barrier, regulating immunity, and whole‐body metabolism. Probiotics and postbiotics are composed of complex ingredients, with different components and compositions having different effects, requiring classification for discussing their mechanisms of action. Probiotics and postbiotics have considerable prospects in preventing various diseases in the livestock industry and animal feed and medical applications. This review highlights the application value of probiotics and postbiotics as potential probiotic products, emphasizing their concept, mechanism of action, and application, to improve the productivity of livestock and poultry.
Collapse
Affiliation(s)
- Chunchen Liu
- College of Public Health North China University of Science and Technology Qinhuangdao Hebei China
- State Key Laboratory of Animal Nutrition College of Animal Science and Technology China Agricultural University Beijing China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition College of Animal Science and Technology China Agricultural University Beijing China
| | - Yue Feng
- State Key Laboratory of Animal Nutrition College of Animal Science and Technology China Agricultural University Beijing China
| | - Min Zhou
- State Key Laboratory of Animal Nutrition College of Animal Science and Technology China Agricultural University Beijing China
| | - Huahui Li
- College of Public Health North China University of Science and Technology Qinhuangdao Hebei China
| | - Xiujun Zhang
- College of Public Health North China University of Science and Technology Qinhuangdao Hebei China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition College of Animal Science and Technology China Agricultural University Beijing China
| |
Collapse
|
20
|
Ashique S, Mishra N, Garg A, Sibuh BZ, Taneja P, Rai G, Djearamane S, Wong LS, Al-Dayan N, Roychoudhury S, Kesari KK, Slama P, Roychoudhury S, Gupta PK. Recent updates on correlation between reactive oxygen species and synbiotics for effective management of ulcerative colitis. Front Nutr 2023; 10:1126579. [PMID: 37545572 PMCID: PMC10400011 DOI: 10.3389/fnut.2023.1126579] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 06/30/2023] [Indexed: 08/08/2023] Open
Abstract
Ulcerative colitis (UC) is presently considered a multifactorial pathology, which may lead to persistent inflammatory action of the gastrointestinal tract (GIT) because of an improperly managed immunological reactivity to the intestinal microbiota found in the GIT. The immune response to common commensal microbes plays an essential role in intestinal inflammation related to UC synbiotics, and it is an important element in the optimal therapy of UC. Therefore, synbiotics, i.e., a mixture of prebiotics and probiotics, may help control the diseased state. Synbiotics alleviate the inflammation of the colon by lowering the reactive oxygen species (ROS) and improving the level of antioxidant enzymes such as catalase (CAT), glutathione peroxidase (GPX), and superoxide dismutase (SOD). Prebiotic supplementation is not a common practice at the moment, despite numerous research findings proving that the benefits of both probiotics and prebiotics encourage their continued existence and positioning in the GIT, with positive effects on human health by managing the inflammatory response. However, the fact that there have been fewer studies on the treatment of UC with different probiotics coupled with selected prebiotics, i.e., synbiotics, and the outcomes of these studies have been very favorable. This evidence-based study explores the possible role of ROS, SOD, and synbiotics in managing the UC. The proposed review also focuses on the role of alteration of gut microbiota, antioxidant defense in the gastrointestinal tract, and the management of UC. Thus, the current article emphasizes oxidative stress signaling in the GI tract, oxidative stress-based pathomechanisms in UC patients, and UC therapies inhibiting oxidative stress' effects.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Pandaveswar School of Pharmacy, Pandaveswar, West Bengal, India
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, India
| | - Ashish Garg
- Department of P.G. Studies and Research in Chemistry and Pharmacy, Rani Durgavati University, Jabalpur, India
| | - Belay Zeleke Sibuh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Pankaj Taneja
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Gopal Rai
- Department of Pharmaceutics, Guru Ramdas Institute of Science and Technology, Jabalpur, India
| | - Sinouvassane Djearamane
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Kampar, Malaysia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
| | - Noura Al-Dayan
- Department of Medical Lab Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Kavindra Kumar Kesari
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
- Department of Applied Physics, Aalto University, Espoo, Finland
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czechia
| | | | - Piyush Kumar Gupta
- Faculty of Health and Life Sciences, INTI International University, Nilai, Malaysia
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, India
| |
Collapse
|
21
|
Golpour F, Abbasi-Alaei M, Babaei F, Mirzababaei M, Parvardeh S, Mohammadi G, Nassiri-Asl M. Short chain fatty acids, a possible treatment option for autoimmune diseases. Biomed Pharmacother 2023; 163:114763. [PMID: 37105078 DOI: 10.1016/j.biopha.2023.114763] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/09/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Gut microbiota can interact with the immune system through its metabolites. Short-chain fatty acids (SCFAs), as one of the most abundant metabolites of the resident gut microbiota play an important role in this crosstalk. SCFAs (acetate, propionate, and butyrate) regulate nearly every type of immune cell in the gut's immune cell repertoire regarding their development and function. SCFAs work through several pathways to impose protection towards colonic health and against local or systemic inflammation. Additionally, SCFAs play a role in the regulation of immune or non-immune pathways that can slow the development of autoimmunity either systematically or in situ. The present study aims to summarize the current knowledge on the immunomodulatory roles of SCFAs and the association between the SCFAs and autoimmune disorders such as celiac disease (CD), inflammatory bowel disease (IBD), rheumatoid arthritis (RA), multiple sclerosis (MS), systemic lupus erythematosus (SLE), type 1 diabetes (T1D) and other immune-mediated diseases, uncovering a brand-new therapeutic possibility to prevent or treat autoimmunity.
Collapse
Affiliation(s)
- Faezeh Golpour
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrsa Abbasi-Alaei
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Babaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Mirzababaei
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Mohammadi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Marjan Nassiri-Asl
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Hu Q, Yu L, Zhai Q, Zhao J, Tian F. Anti-Inflammatory, Barrier Maintenance, and Gut Microbiome Modulation Effects of Saccharomyces cerevisiae QHNLD8L1 on DSS-Induced Ulcerative Colitis in Mice. Int J Mol Sci 2023; 24:ijms24076721. [PMID: 37047694 PMCID: PMC10094816 DOI: 10.3390/ijms24076721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/24/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
The use of probiotics has been considered as a new therapy option for ulcerative colitis (UC), and yeast has recently received widespread recommendation for human health. In this study, the probiotic characteristics of four yeast strains, Saccharomyces boulardii CNCMI-745, Kluyveromyces marxianus QHBYC4L2, Saccharomyces cerevisiae QHNLD8L1, and Debaryomyces hansenii QSCLS6L3, were evaluated in vitro; their ability to ameliorate dextran sulfate sodium (DSS)-induced colitis was investigated. Among these, S. cerevisiae QHNLD8L1 protected against colitis, which was reflected by increased body weight, colon length, histological injury relief, decreased gut inflammation markers, and intestinal barrier restoration. The abundance of the pathogenic bacteria Escherichia–Shigella and Enterococcaceae in mice with colitis decreased after S. cerevisiae QHNLD8L1 treatment. Moreover, S. cerevisiae QHNLD8L1 enriched beneficial bacteria Lactobacillus, Faecalibaculum, and Butyricimonas, enhanced carbon metabolism and fatty acid biosynthesis function, and increased short chain fatty acid (SCFAs) production. Taken together, our results indicate the great potential of S. cerevisiae QHNLD8L1 supplementation for the prevention and alleviation of UC.
Collapse
Affiliation(s)
- Qianjue Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
23
|
Kanang ILD. Effect of Intestinal Microbiota Transplantation on Intestinal Flora and Inflammatory Factor Levels in Patients with Ulcerative Colitis [Letter]. Infect Drug Resist 2023; 16:1713-1714. [PMID: 36999127 PMCID: PMC10046103 DOI: 10.2147/idr.s412512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 03/22/2023] [Indexed: 04/01/2023] Open
Affiliation(s)
- Indah Lestari Daeng Kanang
- Department of Internal Medicine, Faculty of Medicine, Universitas Muslim Indonesia, Makassar, Indoensia
- Correspondence: Indah Lestari Daeng Kanang, Department of Internal Medicine, Faculty of Medicine, Universitas Muslim Indonesia, Makassar, Indoensia, Email
| |
Collapse
|
24
|
Talapko J, Včev A, Meštrović T, Pustijanac E, Jukić M, Škrlec I. Homeostasis and Dysbiosis of the Intestinal Microbiota: Comparing Hallmarks of a Healthy State with Changes in Inflammatory Bowel Disease. Microorganisms 2022; 10:2405. [PMID: 36557658 PMCID: PMC9781915 DOI: 10.3390/microorganisms10122405] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota, which represent a community of different microorganisms in the human intestinal tract, are crucial to preserving human health by participating in various physiological functions and acting as a metabolic organ. In physiological conditions, microbiota-host partnership exerts homeostatic stability; however, changes in intestinal microbiota composition (dysbiosis) are an important factor in the pathogenesis of inflammatory bowel disease and its two main disease entities: ulcerative colitis and Crohn's disease. The incidence and prevalence of these inflammatory conditions have increased rapidly in the last decade, becoming a significant problem for the healthcare system and a true challenge in finding novel therapeutic solutions. The issue is that, despite numerous studies, the etiopathogenesis of inflammatory bowel disease is not completely clear. Based on current knowledge, chronic intestinal inflammation occurs due to altered intestinal microbiota and environmental factors, as well as a complex interplay between the genetic predisposition of the host and an inappropriate innate and acquired immune response. It is important to note that the development of biological and immunomodulatory therapy has led to significant progress in treating inflammatory bowel disease. Certain lifestyle changes and novel approaches-including fecal microbiota transplantation and nutritional supplementation with probiotics, prebiotics, and synbiotics-have offered solutions for dysbiosis management and paved the way towards restoring a healthy microbiome, with only minimal long-term unfavorable effects.
Collapse
Affiliation(s)
- Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Aleksandar Včev
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| | - Tomislav Meštrović
- University Centre Varaždin, University North, 42000 Varaždin, Croatia
- Institute for Health Metrics and Evaluation and the Department of Health Metrics Sciences, University of Washington, Seattle, WA 98195, USA
| | - Emina Pustijanac
- Faculty of Natural Sciences, Juraj Dobrila University of Pula, 52100 Pula, Croatia
| | - Melita Jukić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
- General Hospital Vukovar, Županijska 35, 32000 Vukovar, Croatia
| | - Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Crkvena 21, 31000 Osijek, Croatia
| |
Collapse
|