1
|
Lam LY, Liang TR, Wu WJ, Lam HYP. Intestinal Lactobacillus johnsonii protects against neuroangiostrongyliasis in BALB/c mice through modulation of immune response. PLoS Negl Trop Dis 2025; 19:e0012977. [PMID: 40198714 PMCID: PMC11978024 DOI: 10.1371/journal.pntd.0012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 04/10/2025] Open
Abstract
Neuroangiostrongyliasis is characterized by eosinophilic meningoencephalitis with a robust onset of severe neurological symptoms, by which immunological factors and peripheral metabolites have been postulated to affect the course of the disease. The gut-brain axis provides a bidirectional communication between the gut and the central nervous system, and therefore, understanding the gut microbiome may provide us with a deeper insight into the pathogenesis of angiostrongyliasis. Using 16S rRNA sequencing, we identified an increase in the abundance of different Lactobacillus species in Angiostrongylus cantonensis-infected mice, which was correlated to the disease severity. However, attempts to inoculate L. johnsonii into A. cantonensis-infected mice surprisingly revealed an improvement in neuroinflammation and prolonged survival. RNA sequencing suggested an immune-modulatory effect of L. johnsonii, which was confirmed by ELISA, showing increased levels of IL-10 and reduced levels of IL-2, IL-4, IL-5, and MCP-1 in the brain. Nevertheless, L. johnsonii-associated improvements were not associated with microbiome-related metabolites, as UHPLC-MS/MS analysis revealed no change in short-chain fatty acids, tryptophan metabolites, and bile acids. Our results suggest that while intestinal L. johnsonii appears to be linked to the progression of neuroangiostrongyliasis, these bacteria are likely attempting to modulate the dysregulated immune response to combat the disease. This is one of the first studies to investigate the gut microbiome in mice with A. cantonensis infection, which extends our knowledge from the microbiome-point-of-view of the pathogenesis of angiostrongyliasis and how the body defends against A. cantonensis. This work also extends to possible treatment approaches using L. johnsonii as probiotics.
Collapse
Affiliation(s)
- Long Yin Lam
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Ting-Ruei Liang
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien, Taiwan
| | - Wen-Jui Wu
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Ho Yin Pekkle Lam
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
2
|
Hou S, Yu J, Li Y, Zhao D, Zhang Z. Advances in Fecal Microbiota Transplantation for Gut Dysbiosis-Related Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413197. [PMID: 40013938 PMCID: PMC11967859 DOI: 10.1002/advs.202413197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/22/2025] [Indexed: 02/28/2025]
Abstract
This article provides an overview of the advancements in the application of fecal microbiota transplantation (FMT) in treating diseases related to intestinal dysbiosis. FMT involves the transfer of healthy donor fecal microbiota into the patient's body, aiming to restore the balance of intestinal microbiota and thereby treat a variety of intestinal diseases such as recurrent Clostridioides difficile infection (rCDI), inflammatory bowel disease (IBD), constipation, short bowel syndrome (SBS), and irritable bowel syndrome (IBS). While FMT has shown high efficacy in the treatment of rCDI, further research is needed for its application in other chronic conditions. This article elaborates on the application of FMT in intestinal diseases and the mechanisms of intestinal dysbiosis, as well as discusses key factors influencing the effectiveness of FMT, including donor selection, recipient characteristics, treatment protocols, and methods for assessing microbiota. Additionally, it emphasizes the key to successful FMT. Future research should focus on optimizing the FMT process to ensure long-term safety and explore the potential application of FMT in a broader range of medical conditions.
Collapse
Affiliation(s)
- Shuna Hou
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
- Department of general surgeryThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Jiachen Yu
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Yongshuang Li
- Department of general surgeryThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Duoyi Zhao
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| | - Zhiyu Zhang
- Department of OrthopedicsThe Fourth Affiliated Hospital of China Medical UniversityChina Medical UniversityLiao NingShen Yang110032P. R. China
| |
Collapse
|
3
|
Xu W, Tan H, Hu P, Liu S, Song J, Chen X, Suo H. Advancements in fluorescent labeling in assessing the probiotic adhesion capacity - A review. World J Microbiol Biotechnol 2025; 41:73. [PMID: 40011303 DOI: 10.1007/s11274-024-04186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/29/2024] [Indexed: 02/28/2025]
Abstract
Adhesion capacity of probiotics is closely related to their intestinal-protective effects. The conventional techniques used to evaluate probiotic adhesion capacity have limitations in terms of imaging resolution and quantitative analysis. Fluorescent labelling technology has shown immense potential in recent years owing to its high specificity and sensitivity for resolving probiotic adhesion mechanisms. Although there are still problems with the fluorescence signal intensity and hysteresis effect, this technology has significantly advanced the accurate detection and evaluation of probiotic adhesion capacity. This review examines the critical role of probiotic adhesion and its detection methods, with a special focus on the application of fluorescent-labeling technology. Our objective was to identify more accurate and efficient approaches for evaluating the adhesion capacity of probiotic bacteria while promoting in-depth research into the underlying mechanisms that govern probiotic adhesion.
Collapse
Affiliation(s)
- Weiping Xu
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Han Tan
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Peiling Hu
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Shijian Liu
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, 400715, China
| | - Xiaoyong Chen
- College of Food Science, Southwest University, Chongqing, 400715, China.
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
4
|
Yaikhan T, Wonglapsuwan M, Pahumunto N, Nokchan N, Teanpaisan R, Surachat K. Probiogenomic analysis of Limosilactobacillus fermentum SD7, a probiotic candidate with remarkable aggregation abilities. Heliyon 2025; 11:e42451. [PMID: 40007772 PMCID: PMC11850171 DOI: 10.1016/j.heliyon.2025.e42451] [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: 09/02/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Limosilactobacillus fermentum has gained recognition as a probiotic due to its immunomodulatory properties. In this study, we characterized L. fermentum SD7, which was isolated from the human oral cavity. The genome of L. fermentum SD7 was approximately 2.27 Mb in size, with a 51.1 % GC content. Using comprehensive genome analysis, we compared the genome of L. fermentum SD7 with 153 available genome sequences of L. fermentum strains and categorized the 154 strains into six distinct clades based on core gene single nucleotide polymorphisms. Among the 12,598 orthologous proteins, we identified 910 core genes and 10,169 accessory genes. Our analysis revealed a close similarity between L. fermentum SD7, FS-10, and L13. In addition, L. fermentum SD7 genome contains four strain-specific putative CRISPR-associated genes and lacks antimicrobial resistance and virulence genes. Importantly, we identified 27 genes in L. fermentum SD7 genome that are linked to aggregation ability, which is supported by our probiogenomic analysis. This aggregation ability is considered crucial for the probiotic efficacy of L. fermentum SD7. These findings provide a comprehensive understanding of the genetic composition of L. fermentum and its potential probiotic properties, identifying L. fermentum SD7 as a promising probiotic candidate for use in the food and healthcare industries.
Collapse
Affiliation(s)
- Thunchanok Yaikhan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Monwadee Wonglapsuwan
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Nuntiya Pahumunto
- Research Center of Excellence for Oral Health, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Department of Oral Diagnostic Sciences, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Natakorn Nokchan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Rawee Teanpaisan
- Research Center of Excellence for Oral Health, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Medical Science Research and Innovation Institute, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Komwit Surachat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| |
Collapse
|
5
|
Li P, He X, Feng E, Wei J, Tu H, Chen T. Lactobacillus acidophilus JYLA-126 Ameliorates Obesity-Associated Metabolic Disorders by Positively Regulating the AMPK Signaling Pathway Through the Gut-Liver Axis. Probiotics Antimicrob Proteins 2025; 17:62-80. [PMID: 38051435 DOI: 10.1007/s12602-023-10190-3] [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] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
Obesity is a chronic metabolic disease worldwide and is considered a major health problem in contemporary society. Lactobacillus acidophilus have demonstrated beneficial effects on obesity, but the specific mechanism of how it exerts beneficial effects has not been elucidated. Here, we found that L. acidophilus JYLA-126 had good biological properties for intestinal health, such as antioxidation, acid tolerance, bile salt tolerance, antimicrobial activity, and gut colonization. We further identified that supplementation of L. acidophilus JYLA-126 obese mice possessed a dose-dependent amelioration of body weight, intestinal imbalance, and metabolic disorders compared to HFD-induced mice. Mechanistically, the excellent slimming effect of L. acidophilus JYLA-126 was achieved mainly by reversing HFD-induced gut dysbiosis, inhibiting inflammatory factors and balancing the homeostasis of the gut-liver axis. Specifically, L. acidophilus JYLA-126 improved hepatic glycogen synthesis, lowered oxidative stress, and facilitated lipid metabolism by regulating AMPK signaling pathway-related protein expression to restore the overall metabolic level. Accordingly, L. acidophilus JYLA-126 promoted energy uptake efficiency in obese mice, resulting in significant expression of uncoupling protein 1 (UCP1) protein in brown adipose tissue (BAT), and markedly reduced the size of adipocytes. These findings indicate that the anti-obesity activity of L. acidophilus JYLA-126 correlates with activation of the AMPK signaling pathway through improved gut-liver interactions.
Collapse
Affiliation(s)
- Ping Li
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Xia He
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Enxu Feng
- College of Food Science and Engineering, Shandong Agricultural University, Taian, 271018, People's Republic of China
| | - Jing Wei
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China
| | - Huaijun Tu
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China.
| | - Tingtao Chen
- Departments of Geriatrics, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, People's Republic of China.
- National Engineering Research Center for Bioengineering Drugst and the Technologies, Institute of Translational Medicine, the First Affiliated Hospital, Nanchang University, Nanchang, 330031, People's Republic of China.
| |
Collapse
|
6
|
Cerdán-Alduán M, Salvador-Erro J, Villegas-Remírez A, García-Yoldi D, Ceniceros A, Pastor Y, Gamazo C. Yeast Strains as Probiotic and Postbiotic Agents for the Agglutination of Enteric Pathogens: A Preventive Approach. Pathogens 2025; 14:113. [PMID: 40005490 PMCID: PMC11858296 DOI: 10.3390/pathogens14020113] [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: 12/20/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
This study evaluates the potential of various yeast strains as probiotic and postbiotic agents for agglutinating enteric pathogens, offering a preventive approach to gastrointestinal infections. Different yeast species were tested in vitro against a range of pathogenic bacteria, including enterotoxigenic Escherichia coli ETEC, Shigella flexneri, Salmonella enterica serovar Typhimurium, and Salmonella enterica serovar Enteritidis, to assess their capacity for pathogen agglutination. Additionally, inactivated yeasts were obtained using a novel chemical treatment and employed to explore their efficacy as postbiotic agents. The results suggest that both live and inactivated yeasts are able to agglutinate the different pathogens, potentially limiting bacterial colonization. Notably, we also demonstrated that Wickerhamomyces anomalus, Saccharomyces cerevisiae, and Pichia fermentans, exhibiting agglutination activity, were capable of reducing bacterial adhesion to HeLa cells in vitro. This research highlights yeast's probiotic and postbiotic potential and supports the development of novel yeast-based products for preventing enteric infections.
Collapse
Affiliation(s)
- Michelle Cerdán-Alduán
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.C.-A.); (J.S.-E.); (A.V.-R.); (Y.P.)
| | - Josune Salvador-Erro
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.C.-A.); (J.S.-E.); (A.V.-R.); (Y.P.)
| | - Ana Villegas-Remírez
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.C.-A.); (J.S.-E.); (A.V.-R.); (Y.P.)
| | - David García-Yoldi
- LEV2050, Polígono Industrial Plazaola, Manzana E, Nave 10, 31195 Aizoain, Spain; (D.G.-Y.); (A.C.)
| | - Ana Ceniceros
- LEV2050, Polígono Industrial Plazaola, Manzana E, Nave 10, 31195 Aizoain, Spain; (D.G.-Y.); (A.C.)
| | - Yadira Pastor
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.C.-A.); (J.S.-E.); (A.V.-R.); (Y.P.)
| | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.C.-A.); (J.S.-E.); (A.V.-R.); (Y.P.)
| |
Collapse
|
7
|
Xu H, Feng Y, Du Y, Han Y, Duan X, Jiang Y, Su L, Liu X, Qin S, He K, Huang J. Bacterial-host adhesion dominated by collagen subtypes remodelled by osmotic pressure. NPJ Biofilms Microbiomes 2024; 10:124. [PMID: 39532878 PMCID: PMC11557999 DOI: 10.1038/s41522-024-00600-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Environmental osmolarity plays a crucial role in regulating the functions and behaviors of both host cells and pathogens. However, it remains unclear whether and how environmental osmotic stimuli modulate bacterial‒host interfacial adhesion. Using single-cell force spectroscopy, we revealed that the interfacial adhesion force depended nonlinearly on the osmotic prestimulation of host cells but not bacteria. Quantitatively, the adhesion force increased dramatically from 25.98 nN under isotonic conditions to 112.45 or 93.10 nN after the host cells were treated with the hypotonic or hypertonic solution. There was a strong correlation between the adhesion force and the number of host cells harboring adherent/internalized bacteria. We further revealed that enhanced overexpression levels of collagen XV and II were responsible for the increases in interfacial adhesion under hypotonic and hypertonic conditions, respectively. This work provides new opportunities for developing host-directed antibacterial strategies related to interfacial adhesion from a mechanobiological perspective.
Collapse
Affiliation(s)
- Hongwei Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Yuting Feng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Yongtao Du
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yiming Han
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Xiaocen Duan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
| | - Ying Jiang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China
- Nanchang Innovation Institute of Peking University, Nanchang, China
| | - Liya Su
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Inner Mongolia Key Laboratory of Medical Cell Biology, Hohhot, Inner Mongolia, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Fifth Central Hospital of Tianjin, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People's Hospital, Huangnan Prefecture, Qinghai Province, China
| | - Siying Qin
- School of Life Sciences, Peking University, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing, China.
| |
Collapse
|
8
|
Gallina NLF, Irizarry Tardi N, Li X, Cai A, Horn MJ, Applegate BM, Reddivari L, Bhunia AK. Assessment of Biofilm Formation and Anti-Inflammatory Response of a Probiotic Blend in a Cultured Canine Cell Model. Microorganisms 2024; 12:2284. [PMID: 39597673 PMCID: PMC11596120 DOI: 10.3390/microorganisms12112284] [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: 09/10/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Gut dysbiosis and an inflamed bowel are growing concerns in mammals, including dogs. Probiotic supplements have been used to restore the natural microbial community and improve gastrointestinal health. Biofilm formation, antimicrobial activities, and immunological responses of probiotics are crucial to improving gut health. Thus, we tested a commercial probiotic blend (LabMAX-3), a canine kibble additive comprising Lactobacillus acidophilus, Lacticaseibacillus casei, and Enterococcus faecium for their ability to inactivate common enteric pathogens; their ability to form biofilms; epithelial cell adhesion; and their anti-inflammatory response in the Madin-Darby Canine Kidney (MDCK) cell line. Probiotic LabMAX-3 blend or individual isolates showed a strong inhibitory effect against Salmonella enterica, Listeria monocytogenes, enterotoxigenic Escherichia coli, and Campylobacter jejuni. LabMAX-3 formed biofilms comparable to Staphylococcus aureus. LabMAX-3 adhesion to the MDCK cell line (with or without lipopolysaccharide (LPS) pretreatment) showed comparable adhesion and biofilm formation (p < 0.05) to L. casei ATCC 334 used as a control. LabMAX-3 had no cytotoxic effects on the MDCK cell line during 1 h exposure. The interleukin-10 (IL-10) and tumor necrosis factor alpha (TNFα) ratio of LabMAX-3, compared to the L. casei control, showed a significant increase (p < 0.05), indicating a more pronounced anti-inflammatory response. The data show that LabMAX-3, a canine kibble supplement, can improve gastrointestinal health.
Collapse
Affiliation(s)
- Nicholas L. F. Gallina
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (N.L.F.G.); (N.I.T.); (X.L.); (A.C.)
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
| | - Nicole Irizarry Tardi
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (N.L.F.G.); (N.I.T.); (X.L.); (A.C.)
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
| | - Xilin Li
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (N.L.F.G.); (N.I.T.); (X.L.); (A.C.)
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
| | - Alvin Cai
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (N.L.F.G.); (N.I.T.); (X.L.); (A.C.)
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
| | - Mandy J. Horn
- CH2 Animal Solutions, 21 Bear Creek Estates Dr., Ottumwa, IA 52501, USA;
| | - Bruce M. Applegate
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
- Purdue University Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN 47907, USA
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Lavanya Reddivari
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
- Purdue University Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN 47907, USA
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K. Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (N.L.F.G.); (N.I.T.); (X.L.); (A.C.)
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA; (B.M.A.); (L.R.)
- Purdue University Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, IN 47907, USA
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Department of Comparative Pathology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
9
|
Zhou Y, Guo L, Xiao T, Chen Y, Lv T, Wang Y, Zhang S, Cai H, Chi X, Kong X, Zhou K, Shen P, Xiao Y. Characterization and dynamics of intestinal microbiota in patients with Clostridioides difficile colonization and infection. Microbes Infect 2024; 26:105373. [PMID: 38857786 DOI: 10.1016/j.micinf.2024.105373] [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: 10/22/2023] [Revised: 04/17/2024] [Accepted: 06/02/2024] [Indexed: 06/12/2024]
Abstract
Gut microbiota dysbiosis increases the susceptibility to Clostridioides difficile infection (CDI). In this study, we monitored C. difficile colonization (CDC) patients from no CDC status (CDN) to CDC status (CDCp) and CDI patients from asymptomatic status before CDI (PRECDI), CDI status (ONCDI), to asymptomatic status after CDI (POSTCDI). Based on metagenomic sequencing, we aimed to investigate the interaction pattern between gut microbiota and C. difficile. There was no significant difference of microbiota diversity between CDN and CDCp. In CDCp, Bacteroidetes and short-chain fatty acid (SCFA)-producing bacteria increased, with a positive correlation between SCFA-producing bacteria and C. difficile colonization. Compared with PRECDI, ONCDI and POSTCDI showed a significant decrease in microbiota diversity, particularly in Bacteroidetes and SCFA-producing bacteria, with a positive correlation between opportunistic pathogen and C. difficile. Fatty acid metabolism, and amino acid biosynthesis were enriched in CDN, CDCp, and PRECDI, while bile secretion was enriched in ONCDI and POSTCDI. Microbiota and metabolic pathways interaction networks in CDN and CDCp were more complex, particularly pathways in fatty acid and bile acid metabolism. Increasing of Bacteroidetes and SCFA-producing bacteria, affecting amino acid and fatty acid metabolism, is associated with colonization resistance to C. difficile and inhibiting the development of CDI.
Collapse
Affiliation(s)
- Yanzi Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Department of Rheumatology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310003, China
| | - Lihua Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tingting Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Tao Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shuntian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hongliu Cai
- Department of Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaohui Chi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xiaoyang Kong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, and Second Clinical Medical College, Jinan University, Shenzhen 518000, China
| | - Ping Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250022, China; Research Units of Infectious Disease and Microecology, Chinese Academy of Medical Sciences, Hangzhou 310003, China.
| |
Collapse
|
10
|
Dhakephalkar T, Pisu V, Margale P, Chandras S, Shetty D, Wagh S, Dagar SS, Kapse N, Dhakephalkar PK. Strain-Dependent Adhesion Variations of Shouchella clausii Isolated from Healthy Human Volunteers: A Study on Cell Surface Properties and Potential Probiotic Benefits. Microorganisms 2024; 12:1771. [PMID: 39338446 PMCID: PMC11434523 DOI: 10.3390/microorganisms12091771] [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: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
The probiotic potential of Shouchella clausii is widely recognized, but little is known about its adhesive properties. Hence, this study aims to investigate the adhesion potential and cell surface properties of four human-origin S. clausii strains (B619/R, B603/Nb, B106, and B637/Nm). We evaluated epithelial adhesion, Extracellular Matrix (ECM) binding, aggregation ability, and cell surface hydrophobicity and used genome analysis for validation. Our results demonstrate that adhesion capability is a strain-specific attribute, with significant variations observed among the four strains. B619/R, B603/Nb, and B106 displayed stronger adhesion properties than B637/Nm. Supplementary adhesion assays showed that B637/Nm displayed high hydrophobicity, significant auto-aggregation, and significant mucin-binding abilities. Conversely, B619/R, B603/Nb, and B106 had mildly hydrophobic surfaces and low aggregation abilities. Genome annotation revealed the presence of various adhesion proteins in four strains. Notably, the reduced adhesion potential of B637/Nm was supported by the absence of the cell wall surface anchor family protein (LPxTG motif), which is crucial for interactions with intestinal epithelial cells or mucus components. Further, docking studies provided insights into the interaction of adhesion proteins with gut mucins. These findings contribute to a better understanding of how S. clausii strains interact with the gut environment, facilitating the development of probiotic formulations tailored for improved gut health and well-being.
Collapse
Affiliation(s)
- Tanisha Dhakephalkar
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Vaidehi Pisu
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Prajakta Margale
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Siddhi Chandras
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Deepa Shetty
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Shilpa Wagh
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Sumit Singh Dagar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Neelam Kapse
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Prashant K Dhakephalkar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| |
Collapse
|
11
|
Zhou B, Elean M, Arce L, Fukuyama K, Tomotsune K, Dentice Maidana S, Saha S, Namai F, Nishiyama K, Vizoso-Pinto MG, Villena J, Kitazawa H. The Mucus-Binding Factor Mediates Lacticaseibacillus rhamnosus CRL1505 Adhesion but Not Immunomodulation in the Respiratory Tract. Microorganisms 2024; 12:1209. [PMID: 38930591 PMCID: PMC11205462 DOI: 10.3390/microorganisms12061209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/02/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Lacticaseibacillus rhamnosus CRL1505 possesses immunomodulatory activities in the gastrointestinal and respiratory tracts when administered orally. Its adhesion to the intestinal mucosa does not condition its beneficial effects. The intranasal administration of L. rhamnosus CRL1505 is more effective than the oral route at modulating immunity in the respiratory tract. Nonetheless, it has not yet been established whether the adherence of the CRL1505 strain to the respiratory mucosa is needed to provide the immune benefits to the host. In this study, we evaluated the role of adhesion to the respiratory mucosa of the mucus-binding factor (mbf) knock-out L. rhamnosus CRL1505 mutant (Δmbf CRL1505) in the context of a Toll-like receptor 3 (TLR3)-triggered innate immunity response. In vitro adhesion studies in porcine bronchial epitheliocytes (PBE cells) indicated that L. rhamnosus Δmbf CRL1505 adhered weakly compared to the wild-type strain. However, in vivo studies in mice demonstrated that the Δmbf CRL1505 also reduced lung damage and modulated cytokine production in the respiratory tract after the activation of TLR3 to a similar extent as the wild-type strain. In addition, the mutant and the wild-type strains modulated the production of cytokines and antiviral factors by alveolar macrophages in the same way. These results suggest that the Mbf protein is partially involved in the ability of L. rhamnosus CRL1505 to adhere to the respiratory epithelium, but the protein is not necessary for the CRL1505 strain to exert its immunomodulatory beneficial effects. These findings are a step forward in the understanding of molecular interactions that mediate the beneficial effects of nasally administered probiotics.
Collapse
Affiliation(s)
- Binghui Zhou
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina; (M.E.); (S.D.M.)
| | - Lorena Arce
- Infection Biology Laboratory, INSIBIO (CONICET-UNT), Tucuman CP4000, Argentina; (L.A.)
| | - Kohtaro Fukuyama
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Kae Tomotsune
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
| | - Stefania Dentice Maidana
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina; (M.E.); (S.D.M.)
| | - Sudeb Saha
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Department of Dairy Science, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Fu Namai
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Keita Nishiyama
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | | | - Julio Villena
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina; (M.E.); (S.D.M.)
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (B.Z.); (K.F.); (K.T.); (F.N.); (K.N.)
- Livestock Immunology Unit, International Education and Research Center for Food Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| |
Collapse
|
12
|
Jin P, Lin X, Xu W, Li K, Zhao X, Guo S, Zhao Z, Jiang L, Liao F, Chang L, Wang M, Liu Y, Huang S, Chen Z, Ji F. The feasibility of using pathobiome strains as live biotherapeutic products for human use. IMETA 2024; 3:e202. [PMID: 38898988 PMCID: PMC11183195 DOI: 10.1002/imt2.202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 06/21/2024]
Abstract
The evaluation of pathobiome strains should be conducted at the strain level, involving the identification of the functional genes, while considering the impact of ecological niche and drug interactions. The safety, efficacy, and quality management of live biotherapeutic products (LBPs), especially pathobiome strains, have certain peculiarities. Promising development methods include the recombinant LBP and active metabolites.
Collapse
Affiliation(s)
- Pengfei Jin
- Department of PharmacyBeijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital)BeijingChina
| | - Xiong Lin
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Wenfeng Xu
- Department of PharmacyBeijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital)BeijingChina
| | - Kangning Li
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Xiaoxiao Zhao
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Sirui Guo
- Department of PharmacyBeijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital)BeijingChina
| | - Zinan Zhao
- Department of PharmacyBeijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences; Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital)BeijingChina
| | - Lujie Jiang
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Feng Liao
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Longgang Chang
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Min Wang
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Yanmin Liu
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Shaolei Huang
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Zhangran Chen
- Shenzhen Wedge Microbiology Research Co., Ltd.ShenzhenChina
| | - Fusui Ji
- Department of Cardiology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
13
|
Kopczyńska J, Kowalczyk M. The potential of short-chain fatty acid epigenetic regulation in chronic low-grade inflammation and obesity. Front Immunol 2024; 15:1380476. [PMID: 38605957 PMCID: PMC11008232 DOI: 10.3389/fimmu.2024.1380476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Obesity and chronic low-grade inflammation, often occurring together, significantly contribute to severe metabolic and inflammatory conditions like type 2 diabetes (T2D), cardiovascular disease (CVD), and cancer. A key player is elevated levels of gut dysbiosis-associated lipopolysaccharide (LPS), which disrupts metabolic and immune signaling leading to metabolic endotoxemia, while short-chain fatty acids (SCFAs) beneficially regulate these processes during homeostasis. SCFAs not only safeguard the gut barrier but also exert metabolic and immunomodulatory effects via G protein-coupled receptor binding and epigenetic regulation. SCFAs are emerging as potential agents to counteract dysbiosis-induced epigenetic changes, specifically targeting metabolic and inflammatory genes through DNA methylation, histone acetylation, microRNAs (miRNAs), and long non-coding RNAs (lncRNAs). To assess whether SCFAs can effectively interrupt the detrimental cascade of obesity and inflammation, this review aims to provide a comprehensive overview of the current evidence for their clinical application. The review emphasizes factors influencing SCFA production, the intricate connections between metabolism, the immune system, and the gut microbiome, and the epigenetic mechanisms regulated by SCFAs that impact metabolism and the immune system.
Collapse
Affiliation(s)
- Julia Kopczyńska
- Laboratory of Lactic Acid Bacteria Biotechnology, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
14
|
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
|
15
|
Mazhar S, Simon A, Khokhlova E, Colom J, Leeuwendaal N, Deaton J, Rea K. In vitro safety and functional characterization of the novel Bacillus coagulans strain CGI314. Front Microbiol 2024; 14:1302480. [PMID: 38274758 PMCID: PMC10809412 DOI: 10.3389/fmicb.2023.1302480] [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: 09/26/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Bacillus coagulans species have garnered much interest in health-related functional food research owing to their desirable probiotic properties, including pathogen exclusion, antioxidant, antimicrobial, immunomodulatory and food fermentation capabilities coupled with their tolerance of extreme environments (pH, temperature, gastric and bile acid resistance) and stability due to their endosporulation ability. Methods In this study, the novel strain Bacillus coagulans CGI314 was assessed for safety, and functional probiotic attributes including resistance to heat, gastric acid and bile salts, the ability to adhere to intestinal cells, aggregation properties, the ability to suppress the growth of human pathogens, enzymatic profile, antioxidant capacity using biochemical and cell-based methods, cholesterol assimilation, anti-inflammatory activity, and attenuation of hydrogen peroxide (H2O2)-induced disruption of the intestinal-epithelial barrier. Results B. coagulans CGI314 spores display resistance to high temperatures (40°C, 70°C, and 90°C), and gastric and bile acids [pH 3.0 and bile salt (0.3%)], demonstrating its ability to survive and remain viable under gastrointestinal conditions. Spores and the vegetative form of this strain were able to adhere to a mucous-producing intestinal cell line, demonstrated moderate auto-aggregation properties, and could co-aggregate with potentially pathogenic bacteria. Vegetative cells attenuated LPS-induced pro-inflammatory cytokine gene expression in HT-29 intestinal cell lines and demonstrated broad antagonistic activity toward numerous urinary tract, intestinal, oral, and skin pathogens. Metabolomic profiling demonstrated its ability to synthesize several amino acids, vitamins and short-chain fatty acids from the breakdown of complex molecules or by de novo synthesis. Additionally, B. coagulans CGI314's strong antioxidant capacity was demonstrated using enzyme-based methods and was further supported by its cytoprotective and antioxidant effects in HepG2 and HT-29 cell lines. Furthermore, B. coagulans CGI314 significantly increased the expression of tight junction proteins and partially ameliorated the detrimental effects of H2O2 induced intestinal-epithelial barrier integrity. Discussion Taken together these beneficial functional properties provide strong evidence for B. coagulans CGI314 as a promising potential probiotic candidate in food products.
Collapse
Affiliation(s)
- Shahneela Mazhar
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| | - Annie Simon
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| | - Ekaterina Khokhlova
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| | - Joan Colom
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| | - Natasha Leeuwendaal
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| | - John Deaton
- ADM Deerland Probiotics and Enzymes, Kennesaw, GA, United States
| | - Kieran Rea
- ADM Cork H&W Limited, Bio-Innovation Unit, University College Cork, Cork, Ireland
| |
Collapse
|
16
|
Hyun JH, Yu HS, Woo IK, Lee GW, Lee NK, Paik HD. Anti-inflammatory activities of Levilactobacillus brevis KU15147 in RAW 264.7 cells stimulated with lipopolysaccharide on attenuating NF-κB, AP-1, and MAPK signaling pathways. Food Sci Biotechnol 2023; 32:2105-2115. [PMID: 37860733 PMCID: PMC10581997 DOI: 10.1007/s10068-023-01318-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 10/21/2023] Open
Abstract
Probiotics confer many beneficial effects on several illnesses, ranging from microbial diarrhea to inflammatory diseases. This study was conducted on whether Levilactobacillus brevis KU15147 obtained from kimchi has anti-inflammatory effects in RAW 264.7 cells stimulated with lipopolysaccharide (LPS) and antioxidant potential. L. brevis KU15147 reduced nitric oxide and prostaglandin E2 levels with decreasing the activation of inducible nitric oxide synthase and cyclooxygenase-2 without cell cytotoxicity. In addition, L. brevis KU15147 attenuated proinflammatory cytokine production including tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 in RAW 264.7 cells stimulated with LPS. Additionally, L. brevis KU15147 reduced the activity of nuclear factor-κB, activator protein-1, and mitogen-activated protein kinase signaling pathways. Furthermore, L. brevis KU15147 downregulated the production of reactive oxygen species. Therefore, L. brevis KU15147 was concluded that had an inhibition effect on LPS-induced inflammatory responses and can be used in functional foods to suppress inflammatory diseases. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-023-01318-w.
Collapse
Affiliation(s)
- Jun-Hyun Hyun
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029 Republic of Korea
| | - Hyung-Seok Yu
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029 Republic of Korea
| | - Im-Kyung Woo
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029 Republic of Korea
| | - Gil-Woong Lee
- View of Creativity, GHBio Co., Ltd., 120 Neungdong-Ro, Seoul, Republic of Korea
| | - Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029 Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
17
|
Selmi H, Rocchetti MT, Capozzi V, Semedo-Lemsaddek T, Fiocco D, Spano G, Abidi F. Lactiplantibacillus plantarum from Unexplored Tunisian Ecological Niches: Antimicrobial Potential, Probiotic and Food Applications. Microorganisms 2023; 11:2679. [PMID: 38004691 PMCID: PMC10673251 DOI: 10.3390/microorganisms11112679] [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: 10/12/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The continued exploration of the diversity of lactic acid bacteria in little-studied ecological niches represents a fundamental activity to understand the diffusion and biotechnological significance of this heterogeneous class of prokaryotes. In this study, Lactiplantibacillus plantarum (Lpb. plantarum) strains were isolated from Tunisian vegetable sources, including fermented olive and fermented pepper, and from dead locust intestines, which were subsequently evaluated for their antimicrobial activity against foodborne pathogenic bacteria, including Escherichia coli O157:H7 CECT 4267 and Listeria monocytogenes CECT 4031, as well as against some fungi, including Penicillium expansum, Aspergilus niger, and Botrytis cinerea. In addition, their resistance to oro-gastro-intestinal transit, aggregation capabilities, biofilm production capacity, adhesion to human enterocyte-like cells, and cytotoxicity to colorectal adenocarcinoma cell line were determined. Further, adhesion to tomatoes and the biocontrol potential of this model food matrix were analyzed. It was found that all the strains were able to inhibit the indicator growth, mostly through organic acid production. Furthermore, these strains showed promising probiotic traits, including in vitro tolerance to oro-gastrointestinal conditions, and adhesion to abiotic surfaces and Caco-2 cells. Moreover, all tested Lpb. plantarum strains were able to adhere to tomatoes with similar rates (4.0-6.0 LogCFU/g tomato). The co-culture of LAB strains with pathogens on tomatoes showed that Lpb. plantarum could be a good candidate to control pathogen growth. Nonetheless, further studies are needed to guarantee their use as probiotic strains for biocontrol on food matrices.
Collapse
Affiliation(s)
- Hiba Selmi
- Faculty of Sciences of Bizerte, University of Carthage, Zarzouna, Bizerte 7021, Tunisia
- Laboratory of Protein Engineering and Bioactive Molecules (LIP-MB), National Institute of Applied Sciences and Technology, University of Carthage, Carthage 1054, Tunisia;
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Vittorio Capozzi
- Institute of Sciences of Food Production, National Research Council (CNR) of Italy, c/o CS-DAT, Via Michele Protano, 71122 Foggia, Italy;
| | - Teresa Semedo-Lemsaddek
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, University of Lisbon, Av. da Universidade Técnica de Lisboa, 1300-477 Lisbon, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (D.F.)
| | - Giuseppe Spano
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy
| | - Ferid Abidi
- Laboratory of Protein Engineering and Bioactive Molecules (LIP-MB), National Institute of Applied Sciences and Technology, University of Carthage, Carthage 1054, Tunisia;
| |
Collapse
|
18
|
You Y, Kim SH, Kim CH, Kim IH, Shin Y, Kim TR, Sohn M, Park J. Immune-Stimulating Potential of Lacticaseibacillus rhamnosus LM1019 in RAW 264.7 Cells and Immunosuppressed Mice Induced by Cyclophosphamide. Microorganisms 2023; 11:2312. [PMID: 37764156 PMCID: PMC10535240 DOI: 10.3390/microorganisms11092312] [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: 08/14/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Probiotics, including Lacticaseibacillus rhamnosus (L. rhamnosus), have gained recognition for their potential health benefits, such as enhancing immune function, maintaining gut health, and improving nutrient absorption. This study investigated the effectiveness of L. rhamnosus LM1019 (LM1019) in enhancing immune function. In RAW 264.7 cells, LM1019 demonstrated dose-dependent immune stimulation by increasing nitric oxide production, gene expression of proinflammatory cytokines, and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). These effects were mediated through the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-κB) translocation without inducing cytotoxicity. Furthermore, orally administered LM1019 was evaluated in immunosuppressed mice induced by cyclophosphamide (CTX). High-dose administration of LM1019 significantly increased the subpopulations of lymphocytes, specifically helper T cells (CD4+), as well as two subtypes of natural killer (NK) cells, namely, IFN-γ+ and granzyme B+ NK cells. Additionally, LM1019 at a high dose led to elevated levels of proinflammatory cytokines, including IFN-γ and IL-12, compared to CTX-treated mice. These findings highlight the potential of LM1019 in enhancing the immune system. The study contributes to the growing body of research on the beneficial effects of probiotics on immune function.
Collapse
Affiliation(s)
- Yeji You
- Microbiome R&D Center, Lactomason Co., Ltd., Jinju 52840, Republic of Korea; (Y.Y.); (T.-R.K.); (M.S.)
| | - Sung-Hwan Kim
- Food Research Center, Binggrae Co., Ltd., Namyangju 12253, Republic of Korea; (S.-H.K.); (C.-H.K.); (I.-H.K.); (Y.S.)
| | - Chul-Hong Kim
- Food Research Center, Binggrae Co., Ltd., Namyangju 12253, Republic of Korea; (S.-H.K.); (C.-H.K.); (I.-H.K.); (Y.S.)
| | - In-Hwan Kim
- Food Research Center, Binggrae Co., Ltd., Namyangju 12253, Republic of Korea; (S.-H.K.); (C.-H.K.); (I.-H.K.); (Y.S.)
| | - YoungSup Shin
- Food Research Center, Binggrae Co., Ltd., Namyangju 12253, Republic of Korea; (S.-H.K.); (C.-H.K.); (I.-H.K.); (Y.S.)
| | - Tae-Rahk Kim
- Microbiome R&D Center, Lactomason Co., Ltd., Jinju 52840, Republic of Korea; (Y.Y.); (T.-R.K.); (M.S.)
| | - Minn Sohn
- Microbiome R&D Center, Lactomason Co., Ltd., Jinju 52840, Republic of Korea; (Y.Y.); (T.-R.K.); (M.S.)
| | - Jeseong Park
- Microbiome R&D Center, Lactomason Co., Ltd., Jinju 52840, Republic of Korea; (Y.Y.); (T.-R.K.); (M.S.)
| |
Collapse
|
19
|
Kiepś J, Juzwa W, Olejnik A, Sip A, Tomaszewska-Gras J, Dembczyński R. The Effects of Cellular Membrane Damage on the Long-Term Storage and Adhesion of Probiotic Bacteria in Caco-2 Cell Line. Nutrients 2023; 15:3484. [PMID: 37571422 PMCID: PMC10421378 DOI: 10.3390/nu15153484] [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: 07/13/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
Adhesion is one of the main factors responsible for the probiotic properties of bacteria in the human gut. Membrane proteins affected by cellular damage are one of the key aspects determining adhesion. Fluid-bed-dried preparations containing probiotic bacteria were analyzed in terms of their stability (temperature of glass transition) and shelf life in different conditions (modified atmosphere, refrigeration). Imaging flow cytometry was utilized to determine four subpopulations of cells based on their physiological and morphological properties. Lastly, adhesion was measured in bacteria cultured in optimal conditions and treated with heat shock. The results show that the subpopulations with no or low levels of cell membrane damage exhibit the ability to adhere to Caco-2 cells. The temperature of protein denaturation in bacteria was recorded as being between 65 °C and 70 °C. The highest glass transition temperature (Tg) value for hydroxypropyl methylcellulose (used as a coating substance) was measured at 152.6 °C. Drying and coating can be utilized as a sufficient treatment, allowing a long shelf-life (up to 12 months). It is, however, worth noting that technological processing, especially with high temperatures, may decrease the probiotic value of the preparation by damaging the bacterial cells.
Collapse
Affiliation(s)
- Jakub Kiepś
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Wojciech Juzwa
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Anna Olejnik
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Anna Sip
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| | - Jolanta Tomaszewska-Gras
- Department of Food Safety and Quality Management, Poznan University of Life Sciences, 60-624 Poznan, Poland;
| | - Radosław Dembczyński
- Department of Biotechnology and Food Microbiology, Poznan University of Life Sciences, 60-627 Poznan, Poland; (W.J.); (A.O.); (A.S.)
| |
Collapse
|
20
|
Hassan DS, Hasary HJ, Hassan ZS. Role of Probiotics in the Prevention and Treatment of GIT Cancers: Updated Review. AL-RAFIDAIN JOURNAL OF MEDICAL SCIENCES ( ISSN: 2789-3219 ) 2023; 4:52-59. [DOI: 10.54133/ajms.v4i.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Cancer, one of the leading causes of death worldwide, has been the subject of extensive study by many researchers. Cancer is affected by both genetic and immune system factors in the human body. The gut microbiota plays an important role in the body's capacity to maintain homeostasis. Because of their beneficial effects on human health and their ability to successfully prevent and treat various chronic diseases, such as cancer, probiotics are becoming increasingly important in medicine. A wealth of research has shown that probiotic consumption can significantly helpful in cancer prevention and treatment. The goal of this review is to provide a thorough overview of the research on the function of probiotic bacteria in the prevention and treatment of gastrointestinal cancers.
Collapse
|