1
|
Chornchoem P, Tandhavanant S, Saiprom N, Preechanukul A, Thongchompoo N, Sensorn I, Chantratita W, Chantratita N. Metagenomic evaluation, antimicrobial activities, and immune stimulation of probiotics from dietary supplements and dairy products. Sci Rep 2025; 15:11537. [PMID: 40185827 DOI: 10.1038/s41598-025-95664-w] [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: 01/05/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Probiotics are widely marketed as dietary supplements and dairy products for their purported antimicrobial and immunomodulatory activities, often with limited supporting evidence. We identified and isolated probiotics from commercial dietary supplements and dairy products using metagenomics and cultured-based methods. We assessed their anti-bacterial activity against diverse pathogens and investigated their immunomodulatory effects on phagocytes and natural killer (NK) cells. Metagenomic analysis revealed that Lactobacillus and Bifidobacterium were the predominant genera in dietary supplements, while Streptococcus spp. was dominated in dairy products. However, only 37% of the predominant microorganisms identified by metagenomics were accurately listed on product labels. Among 70 representative probiotic strains, 4.3-17.1% probiotic strains demonstrated strong antibacterial-effects against pathogenic bacteria. Notably, specific strains of Bifidobacterium longum and Lactobacillus plantarum exhibited strong antagonistic activity against extended-spectrum beta-lactamase-producing and carbapenem-resistant Escherichia coli. Some strains of Lactobacillus spp. significantly enhanced phagocytic activity in monocytes and increased IFN-γ production in NK cells, while members of Lactobacillus rhamnosus significantly suppressed TNF-α, IL-6, and IL-8 production in lipopolysaccharide-stimulated macrophages. In contrast, Bifidobacterium animalis stimulated the production of anti-inflammatory cytokines. This study highlights discrepancies in probiotic labeling and demonstrates the antimicrobial and immunomodulatory potential of specific probiotic strains, suggesting their utility in enhancing health and wellness.
Collapse
Affiliation(s)
- Piyaorn Chornchoem
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Sarunporn Tandhavanant
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Natnaree Saiprom
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
| | - Anucha Preechanukul
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand
- Division of Infection and Immunity, University College London, London, UK
| | - Nartthawee Thongchompoo
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Insee Sensorn
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Wasun Chantratita
- Center for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, 420/6 Rajvithi Road, Bangkok, 10400, Thailand.
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
2
|
Dibakoane SR, Mhlongo G, Moonsamy G, Wokadala OC, Mnisi CM, Mlambo V. Phenomenological and mechanistic insights into potential dietary nucleotide - probiotic synergies in layer chickens: A review. Poult Sci 2025; 104:105049. [PMID: 40106904 PMCID: PMC11964621 DOI: 10.1016/j.psj.2025.105049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025] Open
Abstract
Despite their growing popularity as alternatives to antibiotic growth promoters (AGPs), the individual effects of nucleotides and probiotics on poultry gut functionality remain poorly understood. In addition, inconsistent outcomes are quite common in studies where these two additives have been used separately to modify gut function and related parameters in birds. These inconsistencies, which have limited the potential of probiotics and nucleotides as AGP replacements, stem from various factors and need to be addressed. Combining probiotics and nucleotides could potentially enhance their effectiveness and lead to more consistent outcomes in layer chickens. Since their mechanisms of action complement each other, some level of synergy is expected when used together. Both additives have been shown to support gut health, boost immune function, and improve performance in chickens when used individually. However, no studies have investigated the possible synergistic effects of nucleotides and probiotics in poultry. This review makes the case for combined use of probiotics and nucleotides in layer chickens by providing phenomenological and mechanistic insights into hypothetical synergistic effects. This paper highlights the need for AGP alternatives and reviews studies on the effects and mechanisms of probiotics and nucleotides in layer chickens when used individually. We then propose potential mechanisms for their synergistic effects on gut health, performance, and egg quality based on logical deductions from observed biological responses. These proposed mechanisms are hypothetical and require experimental validation. Finally, the review explores how this synergy could lead to more consistent outcomes and enhance the feasibility of AGP-free egg production.
Collapse
Affiliation(s)
- Siphosethu R Dibakoane
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Godfrey Mhlongo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Ghaneshree Moonsamy
- Council for Scientific and Industrial Research (CSIR); Future production: Chemicals, Meiring Naude Drive, Pretoria 0081, South Africa
| | - Obiro Cuthbert Wokadala
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Caven Mguvane Mnisi
- Department of Animal Science, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa; Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa
| | - Victor Mlambo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa.
| |
Collapse
|
3
|
Javanshir N, Ebrahimi V, Mazhary Z, Saadaie Jahromi B, Zuo T, Fard NA. The antiviral effects and underlying mechanisms of probiotics on viral infections. Microb Pathog 2025; 200:107377. [PMID: 39952625 DOI: 10.1016/j.micpath.2025.107377] [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: 08/27/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
In public health emergencies, viral diseases like influenza and COVID-19 have become a major concern. One of the proposed responses to this concern is the use of probiotics. Probiotics have a potent role in arming our bodies to combat viral infections. They affect the innate and adaptive immune systems in various ways. Accumulating studies has shown that probiotics can reduce the possibility of infection or the duration of respiratory symptoms by modulating the functions of the immune system. This review aims to summarize the impacts of probiotics on respiratory viral infections and their potential antiviral mechanisms. Therefore, we herein discussed probiotics in relation to lung immunity, distinct types of respiratory viral infections (VRIs), including influenza, rhinoviruses, respiratory syncytial virus, and upper respiratory viral infections, and lastly, probiotics and their effects on COVID-19. However, more studies are needed to explore the antiviral mechanisms of probiotics.
Collapse
Affiliation(s)
- Nahid Javanshir
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Valimohammad Ebrahimi
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Zakie Mazhary
- Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | | | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, China; Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China.
| | - Najaf Allahyari Fard
- Department of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| |
Collapse
|
4
|
Mi Y, Chen L, Liao N, Wan M. Mendelian randomization analysis revealed a gut microbiota-eye axis in acute anterior uveitis. Eye (Lond) 2025:10.1038/s41433-025-03715-3. [PMID: 39979613 DOI: 10.1038/s41433-025-03715-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Observational studies suggest that gut microbiome (GM) may contribute to acute anterior uveitis (AAU) development, but causality remains unclear. This study was conducted to test whether specific GM taxa were causally associated with AAU. METHODS The GM data were obtained from the DMP, which included 7738 individuals' faecal samples and an analysis of host genotype-taxa abundance associations. The AAU data were derived from the FinnGen Consortium (8624 cases and 473,095 controls). We primarily employed the inverse-variance weighted method, complemented by supplementary sensitivity analyses. RESULTS Higher abundance of Lachnospiraceae noname (OR = 0.86, 95% CI 0.81-0.91, P = 5.7 × 10-8), Alistipes finegoldii (OR = 0.87, 95% CI 0.78-0.96, P = 0.008), Erysipelotrichaceae (OR = 0.90, 95% CI 0.81-0.99, P = 0.037), Erysipelotrichia (OR = 0.90, 95% CI 0.81-0.99, P = 0.037), Erysipelotrichales (OR = 0.90, 95% CI 0.81-0.99, P = 0.037), and Bacteroides ovatus (OR = 0.93, 95% CI 0.87-1.00, P = 0.039) predicted a lower AAU risk. Conversely, higher abundance of Bifidobacterium catenulatum (OR = 1.06, 95% CI: 1.02-1.10, P = 0.005), Bacteroides coprocola (OR = 1.11, 95% CI: 1.02-1.21, P = 0.014), Parabacteroides unclassified (OR = 1.12, 95% CI 1.03-1.22, P = 0.010), and Prevotella (OR = 1.15, 95% CI: 1.01-1.29, P = 0.029) predicted a higher AAU risk. The results also showed a reverse causation from AAU to Bifidobacterium catenulatum (OR = 1.39, 95% CI: 1.03-1.86, P = 0.005). CONCLUSION This study suggests that specific GM is causally associated with AAU risk, warranting more mechanistic validation and clinical trials.
Collapse
Affiliation(s)
- Yuze Mi
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lu Chen
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Na Liao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Minghui Wan
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
5
|
Sharma A, Lee HJ. Antimicrobial Activity of Probiotic Bacteria Isolated from Plants: A Review. Foods 2025; 14:495. [PMID: 39942088 PMCID: PMC11817414 DOI: 10.3390/foods14030495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Lactic acid bacteria (LAB) constitute a heterogeneous group of bacteria isolated from fermented foods, animals, plants, and mammalian guts, with many health-promoting properties. Probiotics with antagonistic properties against human pathogens and foodborne bacteria have garnered significant attention from the scientific fraternity. A dedicated review focusing on plant-derived probiotic bacteria and their antagonistic properties has not been comprehensively reviewed. Thus, this review aimed at providing an overview of LAB isolates derived from several unconventional sources such as fruits, seeds, fruit pulp, leaves, roots, vegetables, grasses, and flowers and with their antibacterial, antifungal, and antiviral properties. This paper reviewed the antimicrobial properties of different genera, Lactobacillus, Leuconostoc, Weissella, Enterococcus, Pediococcus, Bacillus, and Fructobacillus, their postbiotics, and paraprobiotics. Several important mechanisms, including the secretion of bacteriocins, bacteriocin-like substances, reuterin, organic acids (lactic and acetic), peptides, exopolysaccharides, and hydrogen peroxide, have been attributed to their antimicrobial actions against pathogens. However, their precise mode of action is poorly understood; hence, further research should be conducted to reveal detailed mechanisms. Finally, the review discusses the summary and future implications. Given the significance, LAB and derived antimicrobial compounds can potentially be exploited in food preservation and safety or for medicinal applications after evaluating their safety.
Collapse
Affiliation(s)
- Anshul Sharma
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
- Institute for Ageing and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
- Institute for Ageing and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
6
|
González-Parra JA, Barrera-Conde M, Kossatz E, Veza E, de la Torre R, Busquets-Garcia A, Robledo P, Pizarro N. Microbiota and social behavior alterations in a mouse model of down syndrome: Modulation by a synbiotic treatment. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111200. [PMID: 39571716 DOI: 10.1016/j.pnpbp.2024.111200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Sex differences in the composition and functionality of gut microbiota are an emerging field of interest in neurodevelopmental disorders, as they may help in understanding the phenotypic disparities between males and females. This study aimed to characterize sex-related specific alterations in gut microbiota composition in a mouse model of Down syndrome (Ts65Dn mice, TS mice) through the sequencing of the PCR-amplified 16S ribosomal DNA fraction. Moreover, it intended to examine whether the modulation of gut microbiota by the administration of a synbiotic (SYN) treatment would be beneficial for the behavioral alterations observed in male and female TS mice. Our results show that male, but not female, TS mice exhibit alterations in beta diversity compared to their wild-type (WT) littermates. Sex-dependent differences are also observed in the relative abundance of the classes Bacilli and Clostridia. Administering the SYN effectively counteracts hypersociability in females, and normalizes the overall abundance of Bacilli, specifically by increasing Lactobacillaceae. On the contrary, it rescues emotional recognition deficits in male TS mice and increases the relative abundance of the families Lactobacillaceae, Streptococcaceae and Atopobiaceae. In addition, a metagenome KEGG analysis of differentially enriched pathways shows relevant changes in the cofactor biosynthesis and the amino acid synthesis categories. Finally, following SYN treatment, both male and female TS mice exhibit a robust increase in propionic acid levels compared to WT littermates. These findings suggest sex-specific mechanisms that could link gut microbiota composition with behavior in TS mice, and underscore the potential of targeted gut microbiota interventions to modulate social abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jose Antonio González-Parra
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, Hospital del Mar Research Institute, Barcelona, Spain
| | - Marta Barrera-Conde
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Elk Kossatz
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Emma Veza
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain; Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición (CIBERON), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Arnau Busquets-Garcia
- Cell-Type Mechanisms in Normal and Pathological Behavior Research Group, Neuroscience Programme, Hospital del Mar Research Institute, Barcelona, Spain.
| | - Patricia Robledo
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain; Department of Medicine and Life Sciences, University Pompeu Fabra, Barcelona, Spain.
| | - Nieves Pizarro
- Integrative Pharmacology and Systems Neuroscience, Neuroscience Research Program, Hospital del Mar Research Institute, Barcelona, Spain.
| |
Collapse
|
7
|
Vernon JJ. Modulation of the Human Microbiome: Probiotics, Prebiotics, and Microbial Transplants. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:277-294. [PMID: 40111698 DOI: 10.1007/978-3-031-79146-8_17] [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: 03/22/2025]
Abstract
The balance between health and disease is intrinsically linked to the interactions between microbial communities and the host. This complex environment of antagonism and synergy involves both prokaryotic and eukaryotic cells, whose collaborative metabolic pathways and immunomodulatory elements influence system homeostasis. As with the gut and other niches, the oral microbiome has the capacity to affect distal host sites. The ability to manipulate this environment holds the potential to impact local and systemic disease.With the increasing threat of antimicrobial resistance, novel approaches to reduce the burden of disease are essential. The use of probiotics and prebiotics is one such strategy. Probiotics introduce non-pathogenic bacteria into the environment to compete with pathogens for nutrients and attachment sites, or to produce metabolites that counteract disease aetiologies. Prebiotic compounds enhance the growth of health-associated organisms, offering additional benefits, whilst a conjunctive approach with probiotics potentially holds even greater promise. Though widely studied in the gastrointestinal context, their potential for treating oral diseases, such as dental caries and periodontitis, is less understood. Additionally, the use of microbial transplantations has demonstrated efficacy in other areas, reducing systemic inflammation and recolonising with commensal bacteria. Here we evaluate their use in the oral context and their modulatory impact on overall health.In this chapter, we discuss how pro- and prebiotic strategies seek to modulate both the oral and gut environments to promote oral health and prevent disease. We assess novel approaches for utilising health-associated microorganisms to combat oral disorders, either administered locally in the mouth or imparting influence through immune modulation via the oral-gut axis. By examining available clinical trial data, we aim to further understand the intricacies involved in this discipline. Furthermore, we consider the challenges facing the research community, including optimal candidate organism/compound selection and colonisation retention, as well as considerations for future research.
Collapse
Affiliation(s)
- Jon J Vernon
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK.
| |
Collapse
|
8
|
Salek S, Moazamian E, Mohammadi Bardbori A, Shamsdin SA. Anticancer effect of a combinatorial treatment of 5-fluorouracil and cell extract of some probiotic lactobacilli strains isolated from camel milk on colorectal cancer cells. Folia Microbiol (Praha) 2024:10.1007/s12223-024-01228-2. [PMID: 39702737 DOI: 10.1007/s12223-024-01228-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/16/2024] [Indexed: 12/21/2024]
Abstract
Colorectal cancer (CRC) has the highest mortality rate among cancer types, emphasizing the need for auxiliaries to 5-fluorouracil (5-FU) due to resistance and side effects. Metabolites produced by probiotic bacteria exhibit promising anticancer properties against CRC. In the current study, the anticancer effects of cell extract of three potential probiotic lactobacilli strains isolated from camel milk, Lactobacillus helveticus, Lactobacillus gallinarum, and Lactiplantibacillus plantarum, as well as that of the standard probiotic strain Lacticaseibacillus rhamnosus GG (LGG), on the human colon cancer cell line (HT-29) and the normal HEK293 cell line separately or in combination with 5-FU, were evaluated. This study isolated strains from camel milk and compared their probiotic properties to those of LGG. The cell viability, cell apoptosis, and Th17 cytokine production were assessed using the MTT assay, acridine orange/ethidium bromide (AO/EB) staining, and flow cytometry techniques, respectively. The cell extracts of lactobacilli strains combined with 5-FU reduced HT-29 cell viability effectively and increased cell apoptosis. Nevertheless, the cell extracts of lactobacilli strains combined with 5-FU controlled the cytotoxic impact of 5-FU on HEK-293 cell viability and reduced cell apoptosis. No significant differences were observed among the strains. Moreover, the cell extracts from the strains combined with 5-FU increased the levels of cytokines IFN-γ, TNF-α, and IL-17A, all of which contribute to immunity against tumors. The performance of the studied strains was similar to that of the standard probiotic strain (LGG). The investigation revealed that cell extracts from lactobacilli strains may serve as a promising complementary anticancer treatment.
Collapse
Affiliation(s)
- Sanaz Salek
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Elham Moazamian
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran.
| | - Afshin Mohammadi Bardbori
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedeh Azra Shamsdin
- Gasteroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
9
|
Xiong S, Zhang K, Wang J, Bai S, Zeng Q, Liu Y, Peng H, Xuan Y, Mu Y, Tang X, Ding X. Effects of xylo-oligosaccharide supplementation on the production performance, intestinal morphology, cecal short-chain fatty acid levels, and gut microbiota of laying hens. Poult Sci 2024; 103:104371. [PMID: 39405830 PMCID: PMC11525217 DOI: 10.1016/j.psj.2024.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024] Open
Abstract
This study investigated the effects of xylo-oligosaccharide supplementation on the production performance, intestinal morphology, cecal short-chain fatty acid levels, and gut microbiota of laying hens. A total of 800 Lohmann pink layers, each 48 wk old, were randomly divided into 5 dietary treatment groups, namely XOS at 0 (CON), 100 (XOS1), 200 (XOS2), 300 (XOS3) and 400 (X0S4) mg/kg. The experimental period was 24 wk. The results revealed that the egg production rate and the number of eggs laid by each layer between 1 to 12 wk increased as the XOS concentration increased (Plinear < 0.05). The sand-shell egg percentage decreased significantly from 1 to 12 wk in the XOS1, XOS2, and XOS3 groups (PANOVA < 0.05). Compared with the CON group, the 4 XOS dosage groups presented significant increases in the villus height and the ratio of villus height to crypt depth in the jejunum (PANOVA < 0.05), whereas a linear decrease in jejunal crypt depth (Plinear < 0.05) was noted. In addition, XOS supplementation significantly increased the concentrations of butyric acid and isovaleric acid in the caeca (PANOVA < 0.05). High-throughput sequencing analysis of bacterial 16S rRNA revealed that dietary XOS supplementation influenced the cecal microbiota. The alpha diversity analysis indicated that the richness of cecal bacteria was greater in the laying hens fed XOS. The modulation of the cecal microbiota composition upon the addition of XOS was characterized by an increased abundance of Firmicutes and Bifidobacteriaceae, and decreased abundance of Bacteroidetes. At the genus level, dietary XOS supplementation resulted in decreases in the abundances of Bacteroides and Rikenellaceae_RC9_gut_group and an increase in the abundance of Lactobacillus. In conclusion, dietary XOS supplementation improved the production performance of laying hens by increasing the production of short-chain fatty acids and improving their intestinal morphology, which was achieved mainly through changes in the composition of the intestinal microbiota. The recommended level of XOS in the diet of laying hens is 200 mg/kg.
Collapse
Affiliation(s)
- Siyu Xiong
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Keying Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Shiping Bai
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Qiufeng Zeng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yan Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Huanwei Peng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yue Xuan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Yadong Mu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China
| | - Xiaobing Tang
- Yibin Yatai Biotechnology Co., Ltd, Yibin 644000, China
| | - Xuemei Ding
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory of Animal Disease-resistant Nutrition, Ministry of Education, Sichuan Province, Chengdu 611130, China.
| |
Collapse
|
10
|
Kozakai T, Nakajima A, Miyazawa K, Sasaki Y, Odamaki T, Katoh T, Fukuma T, Xiao JZ, Suzuki T, Katayama T, Sakanaka M. An improved temperature-sensitive shuttle vector system for scarless gene deletion in human-gut-associated Bifidobacterium species. iScience 2024; 27:111080. [PMID: 39502284 PMCID: PMC11536034 DOI: 10.1016/j.isci.2024.111080] [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: 04/01/2024] [Revised: 08/27/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024] Open
Abstract
Bifidobacterium is a prevalent bacterial taxon in the human gut that comprises over 10 (sub)species. Previous studies suggest that these species use evolutionarily distinct strategies for symbiosis with their hosts. However, the underlying species-specific mechanisms remain unclear due to the lack of efficient gene knockout systems applicable across different species. Here, we developed improved temperature-sensitive shuttle vectors by introducing Ser139Trp into the replication protein RepB. We then used temperature-sensitive plasmids to construct a double-crossover-mediated scarless gene deletion system. The system was employed for targeted gene deletion in Bifidobacterium longum subsp. longum, B. longum subsp. infantis, Bifidobacterium breve, Bifidobacterium adolescentis, Bifidobacterium kashiwanohense, and Bifidobacterium pseudocatenulatum. Deletion of genes involved in capsular polysaccharide biosynthesis, aromatic lactic acid production, and sugar utilization resulted in the expected phenotypic changes in the respective (sub)species. The temperature-sensitive plasmids developed in this study will aid in deciphering the evolutionary traits of the human-gut-associated Bifidobacterium species.
Collapse
Affiliation(s)
- Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Keisuke Miyazawa
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Yuki Sasaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Takeshi Fukuma
- Faculty of Frontier Engineering, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
- WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Jin-zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
- Innovative Research Institute, R&D Division, Morinaga Milk Industry Co. Ltd., Zama, Kanagawa 252-8583, Japan
| | - Tohru Suzuki
- Faculty of Applied Biological Sciences, Gifu University, Gifu 501-1193, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
11
|
Pearce CS, Bukovsky D, Douchant K, Katoch A, Greenlaw J, Gale DJ, Nashed JY, Brien D, Kuhlmeier VA, Sabbagh MA, Blohm G, De Felice FG, Pare M, Cook DJ, Scott SH, Munoz DP, Sjaarda CP, Tusche A, Sheth PM, Winterborn A, Boehnke S, Gallivan JP. Changes in social environment impact primate gut microbiota composition. Anim Microbiome 2024; 6:66. [PMID: 39538341 PMCID: PMC11562706 DOI: 10.1186/s42523-024-00355-y] [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: 05/10/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The gut microbiota (GM) has proven to be essential for both physical health and mental wellbeing, yet the forces that ultimately shape its composition remain opaque. One critical force known to affect the GM is the social environment. Prior work in humans and free-ranging non-human primates has shown that cohabitation and frequent social interaction can lead to changes in GM composition. However, it is difficult to assess the direction of causation in these studies, and interpretations are complicated by the influence of uncontrolled but correlated factors, such as shared diet. RESULTS We performed a 15-month longitudinal investigation wherein we disentangled the impacts of diet and social living conditions on GM composition in a captive cohort of 13 male cynomolgus macaques. The animals were in single housing for the first 3 months of the study initially with a variable diet. After baseline data collection they were placed on a controlled diet for the remainder of the study. Following this diet shift the animals were moved to paired housing for 6 months, enabling enhanced social interaction, and then subsequently returned to single housing at the end of our study. This structured sequencing of diet and housing changes allowed us to assess their distinct impacts on GM composition. We found that the early dietary adjustments led to GM changes in both alpha and beta diversity, whereas changes in social living conditions only altered beta diversity. With respect to the latter, we found that two particular bacterial families - Lactobacillaceae and Clostridiaceae - demonstrated significant shifts in abundance during the transition from single housing to paired housing, which was distinct from the shifts we observed based on a change in diet. Conversely, we found that other bacteria previously associated with sociality were not altered based on changes in social living conditions but rather only by changes in diet. CONCLUSIONS Together, these findings decouple the influences that diet and social living have on GM composition and reconcile previous observations in the human and animal literatures. Moreover, the results indicate biological alterations of the gut that may, in part, mediate the relationship between sociality and wellbeing.
Collapse
Affiliation(s)
- Colleen S Pearce
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | | | - Katya Douchant
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Abhay Katoch
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Biology, Queen's University, Kingston, ON, Canada
| | - Jill Greenlaw
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Daniel J Gale
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Joseph Y Nashed
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Don Brien
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Valerie A Kuhlmeier
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Mark A Sabbagh
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Gunnar Blohm
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Martin Pare
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Douglas J Cook
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Neurosurgery, Queen's University, Kingston, ON, Canada
| | - Stephen H Scott
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Calvin P Sjaarda
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Anita Tusche
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Prameet M Sheth
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Andrew Winterborn
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Susan Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jason P Gallivan
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
- Department of Psychology, Queen's University, Kingston, ON, Canada.
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
12
|
He X, Zhang Z, Jiang H, Luo H, Gan Q, Wei K, Liu Y, Qin Y, Xiao M. Causal association of gut microbes and blood metabolites with acne identified through systematic mendelian randomization. Sci Rep 2024; 14:26816. [PMID: 39501024 PMCID: PMC11538280 DOI: 10.1038/s41598-024-78603-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: 02/03/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Acne is a prevalent inflammatory disease in dermatology, and its pathogenesis may be associated with inflammation, immunity, and other mechanisms. It commonly manifests in young individuals and frequently imposes a heavy economic, physical, and psychological burden on patients. Gut microbes and blood metabolites, as significant immune and inflammatory regulators in the body, have been hypothesized to form the "neurocutaneous axis." Nonetheless, the precise causal relationships among the gut microbes, circulating blood metabolites, and acne development have yet to be elucidated. This study employed bidirectional two-sample Mendelian randomization (MR) to probe the causal impacts of 412 distinct gut microbes and 249 blood metabolites on acne. Single nucleotide polymorphisms (SNPs), which are closely associated with gut microbes and blood metabolites, were utilized as instrumental variables. This approach was taken to discern whether these elements serve as pathogenic or protective factors in relation to acne. Furthermore, a mediation analysis encompassing gut microbes, blood metabolites, and acne was conducted to explore potential correlations between gut microbes and blood metabolites, as well as their cumulative effects on acne. This was done to substantiate the notion of causality. Bidirectional two-sample MR analysis revealed 8 gut bacteria, 6 bacterial metabolic abundance pathways determined by birdshot, and 8 blood metabolites significantly associated with acne. The mediation MR analysis revealed 2 potential causal relationships, namely, Bifidobacterium-DHA-Acne and Bifidobacterium-Degree of Unsaturation-Acne. This study identified gut microbes and blood metabolites that are causally associated with acne. A potential causal relationship between gut microbes and blood metabolites was obtained via mediation analysis. These insights pave the way for the identification of new targets and the formulation of innovative approaches for the prevention and treatment of acne.
Collapse
Affiliation(s)
- Xin He
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan Province, P R China
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Zhongyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Hengyu Jiang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Hui Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Qianrong Gan
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Kebo Wei
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Ying Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China
| | - Yuesi Qin
- Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan Province, P R China.
| | - Min Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, P. R. China.
| |
Collapse
|
13
|
Somers SE, Davidson GL, Mbandlwa P, McKeon CM, Stanton C, Ross RP, Quinn JL. Manipulating a host-native microbial strain compensates for low microbial diversity by increasing weight gain in a wild bird population. Proc Natl Acad Sci U S A 2024; 121:e2402352121. [PMID: 39401350 PMCID: PMC11513901 DOI: 10.1073/pnas.2402352121] [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: 02/02/2024] [Accepted: 08/25/2024] [Indexed: 10/30/2024] Open
Abstract
Empirical studies from laboratory systems and humans show that the gut microbiota is linked to host health. Similar evidence for effects on traits linked to fitness in nature is rare, not least because experimentally manipulating the gut microbiota is challenging. We isolated, characterized, and cultured a bacterial strain, Lactobacillus kimchicus APC4233, directly from a wild bird (the great tit Parus major) and provided it as a self-administered dietary supplement. We assessed the impact of the treatment on the host microbiota community, on weight, and tested whether the treatment affected a previous result linking microbiota alpha diversity to weight in nestlings. The treatment dramatically increased L. kimchicus abundance in the gut microbiota and increased alpha diversity. This effect was strongest in the youngest birds, validating earlier findings pointing to a brief developmental window when the gut microbiota are most sensitive. In time-lagged models, nestling weight was higher in the treatment birds suggesting L. kimchicus may have probiotic potential. There was also a positive time-lagged relationship between diversity and weight in control birds but not in the treatment birds, suggesting L. kimchicus helped birds compensate for low alpha diversity. We discuss why ecological context is likely key when predicting impacts of the microbiome. The manipulation of the gut microbiota with a host native strain in this wild population provides direct evidence for the role of the microbiota in the ecology and evolution of natural populations.
Collapse
Affiliation(s)
- Shane E. Somers
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
| | - Gabrielle L. Davidson
- School of Biological Sciences, University of East Anglia, NorwichNR4 7TU, United Kingdom
| | - Philiswa Mbandlwa
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - Caroline M. McKeon
- Environment and Marine Sciences, Agri-Food and Biosciences Institute, Northern IrelandBT9 5PX, United Kingdom
- Zoology Department, School of Natural Sciences, Trinity College Dublin, DublinD02 PN40, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, CorkP61 CK84, Ireland
| | - John L. Quinn
- School of Biological, Earth and Environmental Sciences, Distillery Fields, University College Cork, CorkT23 TK30, Ireland
- Environmental Research Institute, University College Cork, CorkT23 XE10, Ireland
| |
Collapse
|
14
|
Pérez-Jiménez GM, Alvarez-Villagomez CS, Martínez-Porchas M, Garibay-Valdez E, Sepúlveda-Quiroz CA, Méndez-Marín O, Martínez-García R, Jesús-Contreras R, Alvarez-González CA, De la Rosa-García SDC. The Indigenous Probiotic Lactococcus lactis PH3-05 Enhances the Growth, Digestive Physiology, and Gut Microbiota of the Tropical Gar ( Atractosteus tropicus) Larvae. Animals (Basel) 2024; 14:2663. [PMID: 39335253 PMCID: PMC11428600 DOI: 10.3390/ani14182663] [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: 08/10/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Probiotics in aquaculture hold promise for enhancing fish health and growth. Due to their increased specificity and affinity for their host, indigenous probiotics may offer isolated and potentially amplified benefits. This study investigated the effects of Lactococcus lactis PH3-05, previously isolated from adults of tropical gar (Atractosteus tropicus), on the growth, survival, digestive enzyme activity, intestinal morphology, expression of barrier and immune genes, and intestinal microbiota composition in the larvae of tropical gar. Larvae were fed with live L. lactis PH3-05 concentrations of 104, 106, and 108 CFU/g for 15 days alongside a control diet without probiotics. Higher concentrations of L. lactis PH3-05 (106 and 108 CFU/g) positively influenced larval growth, increasing hepatocyte area and enterocyte height. The 106 CFU/g dose significantly enhanced survival (46%) and digestive enzyme activity. Notably, the 108 CFU/g dose stimulated increased expression of muc-2 and il-10 genes, suggesting enhanced mucosal barrier function and anti-inflammatory response. Although L. lactis PH3-05 did not significantly change the diversity, structure, or Phylum level composition of intestinal microbiota, which was constituted by Proteobacteria, Bacteroidota, Chloroflexi, and Firmicutes, an increase in Lactobacillus abundance was observed in fish fed with 106 CFU/g, suggesting enhanced probiotic colonization. These results demonstrate that administering L. lactis PH3-05 at 106 CFU/g promotes growth, survival, and digestive health in A. tropicus larvae, establishing it as a promising indigenous probiotic candidate for aquaculture applications.
Collapse
Affiliation(s)
- Graciela María Pérez-Jiménez
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Carina Shianya Alvarez-Villagomez
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Marcel Martínez-Porchas
- Centro de Investigación en Alimentación y Desarrollo, A.C. Biología de Organismos Acuáticos, Hermosillo 83304, Sonora, Mexico; (M.M.-P.); (E.G.-V.)
| | - Estefanía Garibay-Valdez
- Centro de Investigación en Alimentación y Desarrollo, A.C. Biología de Organismos Acuáticos, Hermosillo 83304, Sonora, Mexico; (M.M.-P.); (E.G.-V.)
| | - César Antonio Sepúlveda-Quiroz
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
- Instituto Tecnológico de Villahermosa, Tecnológico Nacional de México, Carretera Villahermosa-Frontera, Km. 3.5, Ciudad Industrial, Villahermosa 86010, Tabasco, Mexico
| | - Otilio Méndez-Marín
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Rafael Martínez-García
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Ronald Jesús-Contreras
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Carlos Alfonso Alvarez-González
- Laboratorio de Fisiología en Recursos Acuáticos (LAFIRA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico; (G.M.P.-J.); (C.S.A.-V.); or (C.A.S.-Q.); (O.M.-M.); (R.M.-G.); (R.J.-C.)
| | - Susana del Carmen De la Rosa-García
- Laboratorio de Microbiología Aplicada (LABMIA), División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Carretera Villahermosa-Cárdenas Km. 0.5, Villahermosa 86039, Tabasco, Mexico
| |
Collapse
|
15
|
Rassmidatta K, Theapparat Y, Chanaksorn N, Carcano P, Adeyemi KD, Ruangpanit Y. Dietary Kluyveromyces marxianus hydrolysate alters humoral immunity, jejunal morphology, cecal microbiota and metabolic pathways in broiler chickens raised under a high stocking density. Poult Sci 2024; 103:103970. [PMID: 38970846 PMCID: PMC11264189 DOI: 10.1016/j.psj.2024.103970] [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: 04/02/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/08/2024] Open
Abstract
This study investigated the impact of dietary supplementation with hydrolyzed yeast (Kluyveromyces marxianus) on growth performance, humoral immunity, jejunal morphology, cecal microbiota and metabolic pathways in broilers raised at 45 kg/m2. A total of 1,176 mixed sex 1-day-old Ross 308 broilers were distributed into 42 pens and randomly assigned to either the control group, the control + 250 g hydrolyzed yeast (HY)/ton, 250HY group, or the control + 500 g HY/ton, 500HY group for 42 d. HY did not affect growth performance. However, HY reduced (P < 0.05) mortality at 25 to 35 d. Dietary HY lowered the heterophil/lymphocyte ratio and enhanced the villus height/crypt depth ratio and Newcastle disease titer (P < 0.05). Compared with HY250 and the control, HY500 upregulated (P < 0.05) IL-10. HY enhanced the α diversity, inferring the richness and evenness of the ceca microbiota. HY500 had greater β diversity than the control (P < 0.05). Six bacterial phyla, namely, Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, Verrucomicrobia, and Cyanobacteria, were found. The relative abundance of Firmicutes was greater in the HY500 treatment group than in the HY250 and control groups. HY decreased the abundance of Actinobacteria. HY supplementation altered (P < 0.05) the abundance of 8 higher-level taxa consisting of 2 classes (Bacilli and Clostridia), 1 order (Lactobacillales), 1 family (Streptococcaceae), and five genera (Streptococcus, Lachnospiraceae_uc, Akkermansiaceae, PACO01270_g, and LLKB_g). HY500 improved (P < 0.05) the abundance of Bacilli, Clostridia, Lactobacillales, Streptococcaceae, Streptococcus, PACO01270_g, and Lachnospiraceae_uc, while HY250 enhanced (P < 0.05) the abundance of Akkermansiaceae and LLKB_g. HY improved the abundance of Lactobacillus and Akkermansia spp. Minimal set of pathway analyses revealed that compared with the control, both HY250 and HY500 regulated 20 metabolic pathways. These findings suggest that dietary K. marxianus hydrolysate, especially HY500, improved humoral immunity and jejunal morphology and beneficially altered the composition and metabolic pathways of the cecal microbiota in broilers raised at 45 kg/m2.
Collapse
Affiliation(s)
- Konkawat Rassmidatta
- Department of Animal Science, Faculty of Agriculture at Kamphaeng Sean, Kasetsart University, Kamphang Saen Campus, Nakhon Pathom, 73140 Thailand
| | - Yongyuth Theapparat
- Center of Excellence in Functional Foods and Gastronomy, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai, Songkhla 90110, Thailand
| | | | | | - Kazeem D Adeyemi
- Department of Animal Production, Faculty of Agriculture, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Yuwares Ruangpanit
- Department of Animal Science, Faculty of Agriculture at Kamphaeng Sean, Kasetsart University, Kamphang Saen Campus, Nakhon Pathom, 73140 Thailand.
| |
Collapse
|
16
|
Adriani L, Mushawwir A, Mayasari N, Rosiyanti AS. Effect of Probiotic Consortium Administration in Improving Organ Function and Blood Biochemistry in Laying Chickens. Pak J Biol Sci 2024; 27:493-498. [PMID: 39530304 DOI: 10.3923/pjbs.2024.493.498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
<b>Background and Objective:</b> Probiotics have been known as a potential alternative to replace antibiotic growth promotors and have many benefits for poultry health. This research investigated that how administering a combination of probiotics affects the health and physiological parameters of laying chickens. This includes understanding if and how probiotics can enhance organ function and influence blood biochemistry profiles in these birds, therefore they are used to increase the production of laying hens. <b>Materials and Methods:</b> A Completely Randomized Design (CRD) with 56 hens was used. The treatments with two consortium yogurt B1 (<i>Bifidobacterium</i> spp. and <i>L. acidophilus</i>) and B2 (<i>L. bulgaricus</i>, <i>S. thermophilus</i>, <i>L. acidophilus</i> and <i>B. bifidum</i>) consisted of a control group that was not given the same treatment as the control group (T0), Group-1 was treated with WSPE probiotic B1 2% (T1), Group-2 was treated with WSPE probiotic B2 2% (T2), Group-3 was treated with 2% probiotic B1 powder (T3), Group-4 was treated with 3% probiotic B1 powder (T4), Group-5 were treated with 2% B2 probiotic powder (T5) and Group-6 were treated with 3% B2 probiotic powder (T6), data were analyzed using Analysis of Variance (ANOVA) and followed by Duncan's multiple range test. <b>Results:</b> The giving consortium probiotics to laying hens has a significant effect on uric acid levels as well as decreased SGOT, SGPT and creatinine levels also increasing total protein, albumin and globulin levels. <b>Conclusion:</b> The use of probiotics 2% increased organ function, namely an increase in total protein, albumin and globulin levels.
Collapse
|
17
|
Stastna M. The Role of Proteomics in Identification of Key Proteins of Bacterial Cells with Focus on Probiotic Bacteria. Int J Mol Sci 2024; 25:8564. [PMID: 39201251 PMCID: PMC11354107 DOI: 10.3390/ijms25168564] [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: 07/01/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/02/2024] Open
Abstract
Probiotics can affect human health, keep the balance between beneficial and pathogenic bacteria, and their colonizing abilities enable the enhancement of the epithelial barrier, preventing the invasion of pathogens. Health benefits of probiotics were related to allergy, depression, eczema, cancer, obesity, inflammatory diseases, viral infections, and immune regulation. Probiotic bacterial cells contain various proteins that function as effector molecules, and explaining their roles in probiotic actions is a key to developing efficient and targeted treatments for various disorders. Systematic proteomic studies of probiotic proteins (probioproteomics) can provide information about the type of proteins involved, their expression levels, and the pathological changes. Advanced proteomic methods with mass spectrometry instrumentation and bioinformatics can point out potential candidates of next-generation probiotics that are regulated under pharmaceutical frameworks. In addition, the application of proteomics with other omics methods creates a powerful tool that can expand our understanding about diverse probiotic functionality. In this review, proteomic strategies for identification/quantitation of the proteins in probiotic bacteria were overviewed. The types of probiotic proteins investigated by proteomics were described, such as intracellular proteins, surface proteins, secreted proteins, and the proteins of extracellular vesicles. Examples of pathological conditions in which probiotic bacteria played crucial roles were discussed.
Collapse
Affiliation(s)
- Miroslava Stastna
- Institute of Analytical Chemistry of the Czech Academy of Sciences, Veveri 97, 602 00 Brno, Czech Republic
| |
Collapse
|
18
|
Sakaryalı Uyar D, Üsküdar Güçlü A, Çelik E, Memiş Özgül B, Altay Koçak A, Başustaoğlu AC. Evaluation of probiotics' efficiency on cariogenic bacteria: randomized controlled clinical study. BMC Oral Health 2024; 24:886. [PMID: 39095860 PMCID: PMC11297621 DOI: 10.1186/s12903-024-04659-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Probiotics are live beneficial bacteria to human health and their efficiency on oral health is still being investigated. The purpose of this study was to evaluate the level of Streptococcus mutans and Lactobacillus species with and without the use of probiotics for six-months after the treatment of all dental caries under general anesthesia. METHODS Fifty-eight pediatric patients without any systemic diseases, whose dental treatments were completed under general anesthesia (GA), were included in the study. The patients were recruited in two-groups; Group A: Patients started using probiotics after GA and Group B: Patients did not use probiotics after GA. Saliva samples were taken from all patients on the day before GA (T0), at one-month (T1), three-month (T2) and six-month (T3) follow-up after GA. The counts of cariogenic bacteria were determined by the analysis of saliva samples using real-time polymerase chain reaction. Statistical significance level was accepted as p < 0.05. RESULTS There was statistically significant difference between Group A and B for T0, T1, T2 and T3 regarding S. mutans (p = 0.001, p = 0.04, p = 0.04, p = 0.03; p < 0.05). However, there was no statistically significant difference between groups regarding Lactobacillus species (p ≥ 0.05). CONCLUSIONS Probiotic use and treatment of all caries significantly reduced the level of S. mutans but not Lactobacillus species. Furthermore, S. mutans decreased after cessation of probiotics, but it was not statistically significant. TRIAL REGISTRATION Study was registered as "Effects of Probiotics on Streptococcus mutans and Lactobacillus species" with the registration number of NCT05859646 (16/05/2023) at https://www. CLINICALTRIALS gov Protocol Registration and Results System.
Collapse
Affiliation(s)
- Didem Sakaryalı Uyar
- Department of Pediatric Dentistry, Faculty of Dentistry, Başkent University, Ankara, Türkiye.
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye.
| | - Aylin Üsküdar Güçlü
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| | - Ekin Çelik
- Department of Medical Biology, Faculty of Medicine, Kırşehir Ahi Evran University, Kırşehir, Türkiye
| | - Betül Memiş Özgül
- Department of Pediatric Dentistry, Faculty of Dentistry, Lokman Hekim University, Ankara, Türkiye
| | - Aylin Altay Koçak
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| | - Ahmet Celal Başustaoğlu
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| |
Collapse
|
19
|
Li C, Wang ZX, Xiao H, Wu FG. Intestinal Delivery of Probiotics: Materials, Strategies, and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310174. [PMID: 38245861 DOI: 10.1002/adma.202310174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/04/2024] [Indexed: 01/22/2024]
Abstract
Probiotics with diverse and crucial properties and functions have attracted broad interest from many researchers, who adopt intestinal delivery of probiotics to modulate the gut microbiota. However, the major problems faced for the therapeutic applications of probiotics are the viability and colonization of probiotics during their processing, oral intake, and subsequent delivery to the gut. The challenges of simple oral delivery (stability, controllability, targeting, etc.) have greatly limited the use of probiotics in clinical therapies. Nanotechnology can endow the probiotics to be delivered to the intestine with improved survival rate and increased resistance to the adverse environment. Additionally, the progress in synthetic biology has created new opportunities for efficiently and purposefully designing and manipulating the probiotics. In this article, a brief overview of the types of probiotics for intestinal delivery, the current progress of different probiotic encapsulation strategies, including the chemical, physical, and genetic strategies and their combinations, and the emerging single-cell encapsulation strategies using nanocoating methods, is presented. The action mechanisms of probiotics that are responsible for eliciting beneficial effects are also briefly discussed. Finally, the therapeutic applications of engineered probiotics are discussed, and the future trends toward developing engineered probiotics with advanced features and improved health benefits are proposed.
Collapse
Affiliation(s)
- Chengcheng Li
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Zi-Xi Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
20
|
Singh S, Koo OK. A Comprehensive Review Exploring the Protective Role of Specific Commensal Gut Bacteria against Salmonella. Pathogens 2024; 13:642. [PMID: 39204243 PMCID: PMC11356920 DOI: 10.3390/pathogens13080642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Gut microbiota is a diverse community of microorganisms that constantly work to protect the gut against pathogens. Salmonella stands out as a notorious foodborne pathogen that interacts with gut microbes, causing an imbalance in the overall composition of microbiota and leading to dysbiosis. This review focuses on the interactions between Salmonella and the key commensal bacteria such as E. coli, Lactobacillus, Clostridium, Akkermansia, and Bacteroides. The review highlights the role of these gut bacteria and their synergy in combating Salmonella through several mechanistic interactions. These include the production of siderophores, which compete with Salmonella for essential iron; the synthesis of short-chain fatty acids (SCFAs), which exert antimicrobial effects and modulate the gut environment; the secretion of bacteriocins, which directly inhibit Salmonella growth; and the modulation of cytokine responses, which influences the host's immune reaction to infection. While much research has explored Salmonella, this review aims to better understand how specific gut bacteria engage with the pathogen, revealing distinct defense mechanisms tailored to each species and how their synergy may lead to enhanced protection against Salmonella. Furthermore, the combination of these commensal bacteria could offer promising avenues for bacteria-mediated therapy during Salmonella-induced gut infections in the future.
Collapse
Affiliation(s)
| | - Ok Kyung Koo
- Department of Food Science & Technology, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
21
|
Tantibhadrasapa A, Li S, Buddhasiri S, Sukjoi C, Mongkolkarvin P, Boonpan P, Wongpalee SP, Paenkaew P, Sutheeworapong S, Nakphaichit M, Nitisinprasert S, Hsieh MH, Thiennimitr P. Probiotic Limosilactobacillus reuteri KUB-AC5 decreases urothelial cell invasion and enhances macrophage killing of uropathogenic Escherichia coli in vitro study. Front Cell Infect Microbiol 2024; 14:1401462. [PMID: 39091675 PMCID: PMC11291381 DOI: 10.3389/fcimb.2024.1401462] [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: 03/15/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Bacterial urinary tract infections (UTI) are among the most common infectious diseases worldwide. The rise of multidrug-resistant (MDR) uropathogenic Escherichia coli (UPEC) UTI cases is a significant threat to healthcare systems. Several probiotic bacteria have been proposed as an alternative to combat MDR UTI. Lactic acid bacteria in the genus Limosilactobacillus are some of the most studied and used probiotics. However, strain-specific effects play a critical role in probiotic properties. L. reuteri KUB-AC5 (AC5), isolated from the chicken gut, confers antimicrobial and immunobiotic effects against some human pathogens. However, the antibacterial and immune modulatory effects of AC5 on UPEC have never been explored. Methods Here, we investigated both the direct and indirect effects of AC5 against UPEC isolates (UTI89, CFT073, and clinical MDR UPEC AT31) in vitro. Using a spot-on lawn, agar-well diffusion, and competitive growth assays, we found that viable AC5 cells and cell-free components of this probiotic significantly reduced the UPEC growth of all strains tested. The human bladder epithelial cell line UM-UC-3 was used to assess the adhesion and pathogen-attachment inhibition properties of AC5 on UPEC. Results and discussion Our data showed that AC5 can attach to UM-UC-3 and decrease UPEC attachment in a dose-dependent manner. Pretreatment of UPEC-infected murine macrophage RAW264.7 cells with viable AC5 (multiplicity of infection, MOI = 1) for 24 hours enhanced macrophage-killing activity and increased proinflammatory (Nos2, Il6, and Tnfa) and anti-inflammatory (Il10) gene expression. These findings indicate the gut-derived AC5 probiotic could be a potential urogenital probiotic against MDR UTI.
Collapse
Affiliation(s)
| | - Songbo Li
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Key Laboratory of Tumor Immunopathology, Youjiang Medical University for Nationalities, Baise, China
| | - Songphon Buddhasiri
- Research Center for Veterinary Biosciences and Veterinary Public Health, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chutikarn Sukjoi
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Panupon Mongkolkarvin
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattarapon Boonpan
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Somsakul Pop Wongpalee
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Prasobsook Paenkaew
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sawannee Sutheeworapong
- Pilot Plant Development and Training Institute (PDTI), King Mongkut’s University of Technology Thonburi (KMUTT), Bangkok, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
- Specialized Research Unit: Probiotics and Prebiotics for Health, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
- Specialized Research Unit: Probiotics and Prebiotics for Health, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Michael H. Hsieh
- Department of Urology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Department of Pediatrics, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
- Department of Microbiology, Immunology, and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Parameth Thiennimitr
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
22
|
Zhang Y, Liu Y, Jiao S, Wang Y, Sa R, Zhao F, Xie J. Short-term supplementation with uncoated and encapsulated Enterococcus faecium affected growth performance, gut microbiome and intestinal barrier integrity in broiler chickens. Poult Sci 2024; 103:103808. [PMID: 38761463 PMCID: PMC11133978 DOI: 10.1016/j.psj.2024.103808] [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: 02/05/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024] Open
Abstract
Enterococcus faecium (E. faecium) is an alternative to antibiotics, while the probiotic effect of short-term application in mature broiler chickens remains unclear. In the current study, 48 Arbor Acres male broilers were chosen to investigate the effects of E. faecium on growth performance, the gut microbiome and intestinal health during the finishing period. Forty-eight birds were randomly allocated to 4 treatment groups that were fed a corn-soybean meal basal diet (Con), a basal diet supplemented with 1 g/kg amoxicillin (ABX), 5×106 CFU/g encapsulated E. faecium (cEF), or 5×106 CFU/g uncoated E. faecium (EF) from d 33 to 42. The results showed that 10 d of antibiotic treatment decreased the growth performance of the broilers (P < 0.05). The feed conversion ratio of the cEF and EF groups were lower than that of the Con group by 0.13 and 0.07, respectively (P > 0.05). The abundance of viable ileal and cecal E. faecium in the cEF group was greater than that in the EF group (P < 0.05), and both groups were markedly greater than those in the Con and ABX groups (P < 0.05). The ABX treatment decreased the Shannon and Chao1 indices of the cecal microbiota, while the dietary E. faecium treatment resulted in significant differences in the β diversity of the ileal and cecal microbiota (P < 0.05). Mantel correlation revealed that the ileal microbiota at the genus level was significantly correlated with the growth performance of broilers, with Lactobacillus, Bacillus and Escherichia-Shigella showing positive and strong correlations (P < 0.05). In the ileum, the crypt depth was lower in the cEF group than in the Con group, but the villi height-to-crypt depth ratio was greater in the cEF group than in the other groups (P = 0.037). However, the expression of the ZO-2 and Occludin genes was downregulated in the E. faecium-fed birds (P < 0.05). In the cecum, the acetate, butyrate and total SCFA levels were greater in the EF group (P < 0.05), while the propionate, isobutyrate and isovalerate levels were lower in the ABX group (P < 0.05). In summary, 10 d of dietary supplementation with E. faecium markedly increased colonization in mature broilers and potentially improved growth performance by modulating the ileal microbiota. Encapsulation techniques could enable a slow release of E. faecium in the intestine, thereby reducing the negative impacts of rapid expansion of E. faecium on the intestinal epithelium.
Collapse
Affiliation(s)
- Ying Zhang
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Youyou Liu
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Songjun Jiao
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuming Wang
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Renna Sa
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Feng Zhao
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jingjing Xie
- The State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
23
|
Du S, Song Z, Cen Y, Fan J, Li P, Si H, Hu D. Susceptibility and cecal microbiota alteration to Eimeria-infection in Yellow-feathered broilers, Arbor Acres broilers and Lohmann pink layers. Poult Sci 2024; 103:103824. [PMID: 38772089 PMCID: PMC11131079 DOI: 10.1016/j.psj.2024.103824] [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: 02/03/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/23/2024] Open
Abstract
Coccidiosis, which is caused by Eimeria species, results in huge economic losses to the poultry industry. Arbor Acres (AA) broilers and yellow-feathered broilers are the dominant broilers in northern and southern China, respectively. However, their susceptibility to coccidiosis has not been fully compared. In this study, the susceptibility of yellow-feathered broilers, AA broilers and Lohmann pink layers to E. tenella was evaluated based on mortality rate, relative body weight gain rate, intestinal lesion score, oocyst output, anticoccidial index (ACI), and cecum weight and length. The yellow-feathered broilers were shown to produce significantly fewer oocysts with higher intestinal lesion score compared to AA broilers, which had the highest growth rates and ACI scores. Subsequently, changes in the cecal microbiota of the 3 chicken lines before and after high-dose infection (1 × 104 oocysts) with E. tenella were determined by 16S rRNA sequencing. The results showed that composition of the microbiota changed dramatically after infection. The abundance of Firmicutes and Bacteroidetes in the infected chickens decreased, and Proteobacteria increased significantly among the different chicken lines. At the genus level, Escherichia increased significantly in all 3 groups of infected chickens, but Lactobacillus decreased to 0% in the infected yellow-feathered broilers. The results of the study indicate that the susceptibility to E. tenella varies among the 3 chicken lines, and that changes in intestinal microbiota by E. tenella-infection among the different chicken lines had a similar trend, but to different degrees. This study provides basic knowledge of the susceptibility in the 3 chicken lines, which can be helpful for the control and prevention of coccidiosis.
Collapse
Affiliation(s)
- Shiqi Du
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Zhixuan Song
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Yucan Cen
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Jingzhi Fan
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Peiyao Li
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, China
| | - Dandan Hu
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, China; Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning, 530004, China; Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, 530004, China.
| |
Collapse
|
24
|
Charkhian H, Soleimannezhadbari E, Bodaqlouei A, Lotfollahi L, Lotfi H, Yousefi N, Shojadel E, Gholinejad Z. Assessment of bacteriocin production by clinical Pseudomonas aeruginosa isolates and their potential as therapeutic agents. Microb Cell Fact 2024; 23:175. [PMID: 38872163 PMCID: PMC11170890 DOI: 10.1186/s12934-024-02450-w] [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: 04/06/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Bacterial infections and the rising antimicrobial resistance pose a significant threat to public health. Pseudomonas aeruginosa produces bacteriocins like pyocins, especially S-type pyocins, which are promising for biological applications. This research focuses on clinical P. aeruginosa isolates to assess their bacteriocin production, inhibitory spectrum, chemical structure, antibacterial agents, and preservative potential. METHODS The identification of P. aeruginosa was conducted through both phenotypic and molecular approaches. The inhibitory spectrum and antibacterial potential of the isolates were assessed. The kinetics of antibacterial peptide production were investigated, and the activity of bacteriocin was quantified in arbitrary units (AU ml-1). Physico-chemical characterization of the antibacterial peptides was performed. Molecular weight estimation was carried out using SDS-PAGE. qRT-PCR analysis was employed to validate the expression of the selected candidate gene. RESULT The antibacterial activity of P. aeruginosa was attributed to the secretion of bacteriocin compounds, which belong to the S-type pyocin family. The use of mitomycin C led to a significant 65.74% increase in pyocin production by these isolates. These S-type pyocins exhibited the ability to inhibit the growth of both Gram-negative (P. mirabilis and P. vulgaris) and Gram-positive (S. aureus, S. epidermidis, E. hirae, S. pyogenes, and S. mutans) bacteria. The molecular weight of S-type pyocin was 66 kDa, and its gene expression was confirmed through qRT-PCR. CONCLUSION These findings suggest that S-type pyocin hold significant potential as therapeutic agents against pathogenic strains. The Physico-chemical resistance of S-type pyocin underscores its potential for broad applications in the pharmaceutical, hygiene, and food industries.
Collapse
Affiliation(s)
- Hamed Charkhian
- Young Researchers Club, Urmia Branch, Islamic Azad University, Urmia, Iran
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Soleimannezhadbari
- Young Researchers Club, Urmia Branch, Islamic Azad University, Urmia, Iran
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Amin Bodaqlouei
- Department of Pharmaceutical and Biomolecular Science, Faculty of Pharmaceutical Science, University of Milan, Milan, Italy
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Lida Lotfollahi
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nesa Yousefi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Ehsan Shojadel
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zafar Gholinejad
- Department of Medical Laboratory Science, Urmia Branch, Islamic Azad University, Urmia, Iran
| |
Collapse
|
25
|
Moreno-Muñoz JA, Ojeda JD, López JJ. A Probiotic Bacterium with Activity against the Most Frequent Bacteria and Viruses Causing Pediatric Diarrhea: Bifidobacterium longum subsp. infantis CECT 7210 ( B. infantis IM1 ®). Microorganisms 2024; 12:1183. [PMID: 38930565 PMCID: PMC11206103 DOI: 10.3390/microorganisms12061183] [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: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The second leading cause of death in children under five years old is diarrheal disease. Probiotics, specifically bifidobacteria, have been associated with a reduction in the number of diarrhea episodes and their severity in babies. In this paper, we summarize the preclinical and clinical evidence of the efficacy of B. longum subsp. infantis IM1® against various gastrointestinal pathogens using in vitro models, animal models, and clinical studies carried out in our laboratory. The preclinical data demonstrate that IM1® effectively inhibits rotavirus replication (by up to 36.05%) in MA-104 and HT-29 cells and from infection (up to 48.50%) through the production of an 11-amino-acid peptide. IM1® displays the capability to displace pathogens from enterocytes, particularly Cronobacter sakazakii and Salmonella enterica, and to reduce the adhesion to the HT29 cells of C. sakazakii and Shigella sonnei. In animal models, the IM1® strain exhibits in vivo protection against rotavirus and improves the clinical symptomatology of bacterial gastroenteritis. A clinical study involving infants under 3 months of age revealed that IM1® reduced episodes of diarrhea, proving to be safe, well tolerated, and associated with a lower prevalence of constipation. B. infantis IM1® emerges as an effective probiotic, diminishing episodes of diarrhea caused by gastrointestinal pathogens.
Collapse
Affiliation(s)
- José Antonio Moreno-Muñoz
- Laboratorios Ordesa S.L., Parc Científic de Barcelona, C/Baldiri Reixac 15-21, 08028 Barcelona, Spain; (J.D.O.); (J.J.L.)
| | | | | |
Collapse
|
26
|
Zwolschen JW, Vos AP, Ariëns RMC, Schols HA. In vitro batch fermentation of (un)saturated homogalacturonan oligosaccharides. Carbohydr Polym 2024; 329:121789. [PMID: 38286556 DOI: 10.1016/j.carbpol.2024.121789] [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: 10/23/2023] [Revised: 12/14/2023] [Accepted: 01/04/2024] [Indexed: 01/31/2024]
Abstract
Pectin, predominantly present within plant cell walls, is a dietary fiber that potentially induces distinct health effects depending on its molecular structure. Such structure-dependent health effects of pectin-derived galacturonic acid oligosaccharides (GalA-OS) are yet largely unknown. This study describes the influence of methyl-esterification and ∆4,5-unsaturation of GalA-OS through defined sets of GalA-OS made from pectin using defined pectinases, on the fermentability by individual fecal inocula. The metabolite production, OS utilization, quantity and size, methyl-esterification and saturation of remaining GalA-OS were monitored during the fermentation of GalA-OS. Fermentation of all GalA-OS predominantly induced the production of acetate, butyrate and propionate. Metabolization of unsaturated GalA-OS (uGalA-OS) significantly increased butyrate formation compared to saturated GalA-OS (satGalA-OS), while satGalA-OS significantly increased propionate formation. Absence of methyl-esters within GalA-OS improved substrate metabolization during the first 18 h of fermentation (99 %) compared to their esterified analogues (51 %). Furthermore, HPAEC and HILIC-LC-MS revealed accumulation of specific methyl-esterified GalA-OS, confirming that methyl-esterification delays fermentation. Fermentation of structurally distinct GalA-OS results in donor specific microbiota composition with uGalA-OS specifically stimulating the butyrate-producer Clostridium Butyricum. This study concludes that GalA-OS fermentation induces highly structure-dependent changes in the gut microbiota, further expanding their potential use as prebiotics.
Collapse
Affiliation(s)
- J W Zwolschen
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - A P Vos
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - R M C Ariëns
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - H A Schols
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands.
| |
Collapse
|
27
|
Salek S, Moazamian E, Mohammadi Bardbori A, Shamsdin SA. The anticancer effect of potential probiotic L. fermentum and L. plantarum in combination with 5-fluorouracil on colorectal cancer cells. World J Microbiol Biotechnol 2024; 40:139. [PMID: 38514489 DOI: 10.1007/s11274-024-03929-9] [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: 10/25/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024]
Abstract
5-Fluorouracil (5-FU) is an effective chemotherapy drug in the treatment of colorectal cancer (CRC). However, auxiliary or alternative therapies must be sought due to its resistance and potential side effects. Certain probiotic metabolites exhibit anticancer properties. In this study evaluated the anticancer and potential therapeutic activities of cell extracts potential probiotic strains, Limosilactobacillus fermentum and Lactiplantibacillus plantarum isolated from the mule milk and the standard probiotic strain Lacticaseibacillus rhamnosus GG (LGG) against the human colon cancer cell line (HT-29) and the normal cell line (HEK-293) alone or in combination with 5-FU. In this study, L. plantarum and L. fermentum, which were isolated from mule milk, were identified using biochemical and molecular methods. Their probiotic properties were investigated in vitro and compared with the standard probiotic strain of the species L. rhamnosus GG. The MTT assay, acridine orange/ethidium bromide (AO/EB) fluorescent staining, and flow cytometry were employed to measure the viability of cell lines, cell apoptosis, and production rates of Th17 cytokines, respectively. The results demonstrated that the combination of lactobacilli cell extracts and 5-FU decreased cell viability and induced apoptosis in HT-29 cells. Furthermore, this combination protected HEK-293 cells from the cytotoxic effects of 5-FU, enhancing their viability and reducing apoptosis. Moreover, the combination treatment led to an increase in the levels of IL-17A, IFN-γ, and TNF-α, which can enhance anti-tumor immunity. In conclusion, the cell extracts of the lactobacilli strains probably can act as a potential complementary anticancer therapy.
Collapse
Affiliation(s)
- Sanaz Salek
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Elham Moazamian
- Department of Microbiology, College of Sciences, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran.
| | - Afshin Mohammadi Bardbori
- Department of Toxicology and Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyedeh Azra Shamsdin
- Gasteroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Niu D, Feng N, Xi S, Xu J, Su Y. Genomics-based analysis of four porcine-derived lactic acid bacteria strains and their evaluation as potential probiotics. Mol Genet Genomics 2024; 299:24. [PMID: 38438804 DOI: 10.1007/s00438-024-02101-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/16/2023] [Indexed: 03/06/2024]
Abstract
The search for probiotics and exploration of their functions are crucial for livestock farming. Recently, porcine-derived lactic acid bacteria (LAB) have shown great potential as probiotics. However, research on the evaluation of porcine-derived LAB as potential probiotics through genomics-based analysis is relatively limited. The present study analyzed four porcine-derived LAB strains (Lactobacillus johnsonii L16, Latilactobacillus curvatus ZHA1, Ligilactobacillus salivarius ZSA5 and Ligilactobacillus animalis ZSB1) using genomic techniques and combined with in vitro tests to evaluate their potential as probiotics. The genome sizes of the four strains ranged from 1,897,301 bp to 2,318,470 bp with the GC contents from 33.03 to 41.97%. Pan-genomic analysis and collinearity analysis indicated differences among the genomes of four strains. Carbohydrate active enzymes analysis revealed that L. johnsonii L16 encoded more carbohydrate active enzymes than other strains. KEGG pathway analysis and in vitro tests confirmed that L. johnsonii L16 could utilize a wide range of carbohydrates and had good utilization capacity for each carbohydrate. The four strains had genes related to acid tolerance and were tolerant to low pH, with L. johnsonii L16 showing the greatest tolerance. The four strains contained genes related to bile salt tolerance and were able to tolerate 0.1% bile salt. Four strains had antioxidant related genes and exhibited antioxidant activity in in vitro tests. They contained the genes linked with organic acid biosynthesis and exhibited antibacterial activity against enterotoxigenic Escherichia coli K88 (ETEC K88) and Salmonella 6,7:c:1,5, wherein, L. johnsonii L16 and L. salivarius ZSA5 had gene clusters encoding bacteriocin. Results suggest that genome analysis combined with in vitro tests is an effective approach for evaluating different strains as probiotics. The findings of this study indicate that L. johnsonii L16 has the potential as a probiotic strain among the four strains and provide theoretical basis for the development of probiotics in swine production.
Collapse
Affiliation(s)
- Dekai Niu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang Road, Nanjing, 210095, China
| | - Ni Feng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang Road, Nanjing, 210095, China
| | - Siteng Xi
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang Road, Nanjing, 210095, China
| | - Jianjian Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang Road, Nanjing, 210095, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang Road, Nanjing, 210095, China.
| |
Collapse
|
29
|
Yehuala GA, Shibeshi NT, Kim SH, Park MK. Characterization of Autochthonous Lactic Acid Bacteria Isolated from a Traditional Ethiopian Beverage, Tella. Foods 2024; 13:575. [PMID: 38397552 PMCID: PMC10888401 DOI: 10.3390/foods13040575] [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: 01/11/2024] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
This study aimed to isolate lactic acid bacteria (LAB) from a traditional Ethiopian fermented product, Tella, and evaluate their functional properties. Of forty-three isolates, seven LAB were screened and identified as Pediococcus pentosaceus, Latilactobacillus curvatus, Leuconostoc mesenteroides, and Lactiplantibacillus plantarum species. The isolates were tested for their alcohol tolerance, acid and bile resistance, auto-aggregation, co-aggregation, hydrophobicity, antibacterial activity, and antibiotic susceptibility. LAB isolates, specifically P. pentosaceus TAA01, L. mesenteroides TDB22, and L. plantarum TDM41, showed a higher degree of alcohol tolerance in 8% and 10% (w/v) ethanol concentrations. Additionally, these three isolates displayed survival rates >85% in both acidic pH and bile environments. Among the isolates, L. plantarum TDM41 demonstrated the highest auto-aggregation, co-aggregation, and hydrophobicity with (44.9 ± 1.7)%, (41.4 ± 0.2)%, and (52.1 ± 0.1)% values, respectively. The cell-free supernatant of the isolates exhibited antibacterial activity against foodborne pathogens of Escherichia coli, Salmonella Enteritidis, and Staphylococcus aureus. Each isolate exhibited various levels of resistance and susceptibility to seven antibiotics and resistance was observed against four of the antibiotics tested. After performing a principal component analysis, Pediococcus pentosaceus TAA01, L. mesenteroides TDB22, and L. plantarum TDM41 were selected as the most promising ethanol-tolerant probiotic isolates.
Collapse
Affiliation(s)
- Gashaw Assefa Yehuala
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; (G.A.Y.); (S.-H.K.)
- College of Biological and Chemical Engineering, Addis Ababa Science and Technology University, Addis Ababa P.O. Box 16417, Ethiopia
- School of Chemical and Bio-Engineering, Addis Ababa Institute of Technology, Addis Ababa University, Addis Ababa P.O. Box 385, Ethiopia;
| | - Nurelegne Tefera Shibeshi
- School of Chemical and Bio-Engineering, Addis Ababa Institute of Technology, Addis Ababa University, Addis Ababa P.O. Box 385, Ethiopia;
| | - Su-Hyeon Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; (G.A.Y.); (S.-H.K.)
- Food and Bio-Industry Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mi-Kyung Park
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; (G.A.Y.); (S.-H.K.)
- Food and Bio-Industry Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
30
|
Lin M, Yanjun C. Research progress on the mechanism of probiotics regulating cow milk allergy in early childhood and its application in hypoallergenic infant formula. Front Nutr 2024; 11:1254979. [PMID: 38419849 PMCID: PMC10900986 DOI: 10.3389/fnut.2024.1254979] [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: 07/08/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Some infants and young children suffer from cow's milk allergy (CMA), and have always mainly used hypoallergenic infant formula as a substitute for breast milk, but some of these formulas can still cause allergic reactions. In recent years, it has been found that probiotic nutritional interventions can regulate CMA in children. Scientific and reasonable application of probiotics to hypoallergenic infant formula is the key research direction in the future. This paper discusses the mechanism and clinical symptoms of CMA in children. This review critically ex- amines the issue of how probiotics use intestinal flora as the main vector to combine with the immune system to exert physiological functions to intervene CMA in children, with a particular focus on four mechanisms: promoting the early establishment of intestinal microecological balance, regulating the body's immunity and alleviating allergic response, enhancing the intestinal mucosal barrier function, and destroying allergen epitopes. Additionally, it overviews the development process of hypoallergenic infant formula and the research progress of probiotics in hypoallergenic infant formula. The article also offers suggestions and outlines potential future research directions and ideas in this field.
Collapse
Affiliation(s)
| | - Cong Yanjun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, College of Food and Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
31
|
Saha MR, Dey P. Pharmacological benefits of Acacia against metabolic diseases: intestinal-level bioactivities and favorable modulation of gut microbiota. Arch Physiol Biochem 2024; 130:70-86. [PMID: 34411504 DOI: 10.1080/13813455.2021.1966475] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022]
Abstract
CONTEXT Obesity-associated chronic metabolic disease is a leading contributor to mortality globally. Plants belonging to the genera Acacia are routinely used for the treatment of diverse metabolic diseases under different ethnomedicinal practices around the globe. OBJECTIVE The current review centres around the pharmacological evidence of intestinal-level mechanisms for metabolic health benefits by Acacia spp. RESULTS Acacia spp. increase the proportions of gut commensals (Bifidobacterium and Lactobacillus) and reduces the population of opportunistic pathobionts (Escherichia coli and Clostridium). Acacia gum that is rich in fibre, can also be a source of prebiotics to improve gut health. The intestinal-level anti-inflammatory activities of Acacia are likely to contribute to improvements in gut barrier function that would prevent gut-to-systemic endotoxin translocation and limit "low-grade" inflammation associated with metabolic diseases. CONCLUSION This comprehensive review for the first time has emphasised the intestinal-level benefits of Acacia spp. which could be instrumental in limiting the burden of metabolic disease.
Collapse
Affiliation(s)
- Manas Ranjan Saha
- Department of Life Science, Vidyasagar Primary Teachers Training Institute (B.Ed.), Malda, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
32
|
Zhang Z, Yin B, Liu F, Zhou W, Wang M, Chang Z, Zhou J, Yue M, Chen J, Feng Z. Effect of the initial pH of the culture medium on the nutrient consumption pattern of Bifidobacterium animalis subsp. lactis Bb12 and the improvement of acid resistance by purine and pyrimidine compounds. J Appl Microbiol 2024; 135:lxae022. [PMID: 38299790 DOI: 10.1093/jambio/lxae022] [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: 08/14/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/02/2024]
Abstract
AIMS During fermentation, the accumulation of acidic products can induce media acidification, which restrains the growth of Bifidobacterium animalis subsp. lactis Bb12 (Bb12). This study investigated the nutrient consumption patterns of Bb12 under acid stress and effects of specific nutrients on the acid resistance of Bb12. METHODS AND RESULTS Bb12 was cultured in chemically defined medium (CDM) at different initial pH values. Nutrient consumption patterns were analyzed in CDM at pH 5.3, 5.7, and 6.7. The patterns varied with pH: Asp + Asn had the highest consumption rate at pH 5.3 and 5.7, while Ala was predominant at pH 6.7. Regardless of the pH levels (5.3, 5.7, or 6.7), ascorbic acid, adenine, and Fe2+ were vitamins, nucleobases, and metal ions with the highest consumption rates, respectively. Nutrients whose consumption rates exceeded 50% were added individually in CDM at pH 5.3, 5.7, and 6.7. It was demonstrated that only some of them could promote the growth of Bb12. Mixed nutrients that could promote the growth of Bb12 were added to three different CDM. In CDM at pH 5.3, 5.7, and 6.7, it was found that the viable cell count of Bb12 was the highest after adding mixed nutrients, which were 8.87, 9.02, and 9.10 log CFU ml-1, respectively. CONCLUSIONS The findings suggest that the initial pH of the culture medium affects the nutrient consumption patterns of Bb12. Specific nutrients can enhance the growth of Bb12 under acidic conditions and increase its acid resistance.
Collapse
Affiliation(s)
- Zongcai Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Boxing Yin
- Yangzhou Yangda Kangyuan Dairy Co., Ltd, No. 88, Dingxing Road, Guangling District, Yangzhou 225004, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Wei Zhou
- Yangzhou Yangda Kangyuan Dairy Co., Ltd, No. 88, Dingxing Road, Guangling District, Yangzhou 225004, China
| | - Mengrui Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Ziqing Chang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Junping Zhou
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Mingzhe Yue
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Junxia Chen
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, No. 600, Changjiang Road, Harbin 150030, China
| | - Zhen Feng
- Yangzhou Yangda Kangyuan Dairy Co., Ltd, No. 88, Dingxing Road, Guangling District, Yangzhou 225004, China
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning 571533, China
| |
Collapse
|
33
|
Maigoro AY, Lee JH, Kim H, Frunze O, Kwon HW. Gut Microbiota of Apis mellifera at Selected Ontogenetic Stages and Their Immunogenic Potential during Summer. Pathogens 2024; 13:122. [PMID: 38392860 PMCID: PMC10893431 DOI: 10.3390/pathogens13020122] [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: 01/10/2024] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Honeybees (Apis mellifera) are pollinating agents of economic importance. The role of the gut microbiome in honeybee health has become increasingly evident due to its relationship with immune function, growth, and development. Although their dynamics at various developmental stages have been documented, their dynamics during the era of colony collapse disorder and immunogenic potential, which are connected to the antagonistic immune response against pathogens, need to be elucidated. Using 16S rRNA gene Illumina sequencing, the results indicated changes in the gut microbiota with the developmental stage. The bacterial diversity of fifth stage larva was significantly different among the other age groups, in which Fructobacillus, Escherichia-Shigella, Bombella, and Tyzzerella were unique bacteria. In addition, the diversity of the worker bee microbiome was distinct from that of the younger microbiome. Lactobacillus and Gilliamella remained conserved throughout the developmental stages, while Bifidobacterium colonized only worker bees. Using an in silico approach, the production potential of lipopolysaccharide-endotoxin was predicted. Forager bees tend to have a higher abundance rate of Gram-negative bacteria. Our results revealed the evolutionary importance of some microbiome from the larval stage to the adult stage, providing insight into the potential dynamics of disease response and susceptibility. This finding provides a theoretical foundation for furthering the understanding of the function of the gut microbiota at various developmental stages related to probiotic development and immunogenic potential.
Collapse
Affiliation(s)
- Abdulkadir Yusif Maigoro
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea (H.K.)
| | - Jeong-Hyeon Lee
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea;
| | - Hyunjee Kim
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea (H.K.)
| | - Olga Frunze
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea (H.K.)
| | - Hyung-Wook Kwon
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012, Republic of Korea (H.K.)
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea;
| |
Collapse
|
34
|
Prieto-Santiago V, Aguiló-Aguayo I, Ortiz-Solà J, Anguera M, Abadias M. Selection of a Probiotic for Its Potential for Developing a Synbiotic Peach and Grape Juice. Foods 2024; 13:350. [PMID: 38275717 PMCID: PMC10814886 DOI: 10.3390/foods13020350] [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: 12/15/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Due to recent interest in the potential of probiotics as health promoters and the impact of health and environmental concerns on eating habits, non-dairy probiotic food products are required. This study aimed to evaluate the viability of different probiotic microorganisms in peach and grape juice (PGJ) with or without the prebiotic inulin and their antimicrobial activity against the foodborne pathogen Listeria monocytogenes and the juice spoilage microorganism Saccharomyces cerevisiae. Firstly, the viability of seven probiotic strains was studied in PGJ with an initial concentration of 107 CFU/mL for 21 days at 4 °C and for 3 days at 37 °C. In parallel, the physicochemical effect, the antimicrobial effect and the lactic acid production in PGJ were evaluated. Secondly, the probiotic with the best viability results was selected to study its antimicrobial effect against L. monocytogenes and S. cerevisiae, as well as ethanol and acetaldehyde production by the latter. L. casei showed the highest viability and grew in both refrigerated and fermentation conditions (1 log), produced the greatest lactic acid (5.12 g/L) and demonstrated in vitro anti-Listeria activity. Although the addition of the prebiotic did not improve the viability, lactic acid production or anti-Listeria activity of the probiotics, under the conditions studied, the prebiotic potential of inulin, support the design of a synbiotic juice. Finally, although none of the probiotic, fermentation products, or postbiotics showed any antimicrobial activity against L. monocytogenes or S. cerevisiae, the addition of L. casei to the PGJ significantly reduced the production of S. cerevisiae metabolite ethanol (29%) and acetaldehyde (50%). L. casei might be a suitable probiotic to deliver a safe and functional PGJ, although further research should be carried out to determine the effect of the probiotic and fermentation on the nutritional profile of PGJ.
Collapse
Affiliation(s)
| | | | | | | | - Maribel Abadias
- Institute of Agrifood Research and Technology (IRTA), Postharvest Program, Edifici Fruitcentre, Parc Agrobiotech Lleida, Parc de Gardeny, 25003 Lleida, Spain; (V.P.-S.); (I.A.-A.); (J.O.-S.); (M.A.)
| |
Collapse
|
35
|
Marzhoseyni Z, Shaghaghi Z, Alvandi M, Shirvani M. Investigating the Influence of Gut Microbiota-related Metabolites in Gastrointestinal Cancer. Curr Cancer Drug Targets 2024; 24:612-628. [PMID: 38213140 DOI: 10.2174/0115680096274860231111210214] [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: 07/30/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 01/13/2024]
Abstract
Gastrointestinal (GI) cancer is a major health concern due to its prevalence, impact on well-being, high mortality rate, economic burden, and potential for prevention and early detection. GI cancer research has made remarkable strides in understanding biology, risk factors, and treatment options. An emerging area of research is the gut microbiome's role in GI cancer development and treatment response. The gut microbiome, vital for digestion, metabolism, and immune function, is increasingly linked to GI cancers. Dysbiosis and alterations in gut microbe composition may contribute to cancer development. Scientists study how specific bacteria or microbial metabolites influence cancer progression and treatment response. Modulating the gut microbiota shows promise in enhancing treatment efficacy and preventing GI cancers. Gut microbiota dysbiosis can impact GI cancer through inflammation, metabolite production, genotoxicity, and immune modulation. Microbes produce metabolites like short-chain fatty acids, bile acids, and secondary metabolites. These affect host cells, influencing processes like cell proliferation, apoptosis, DNA damage, and immune regulation, all implicated in cancer development. This review explores the latest research on gut microbiota metabolites and their molecular mechanisms in GI cancers. The hope is that this attempt will help in conducting other relevant research to unravel the precise mechanism involved, identify microbial signatures associated with GI cancer, and develop targets.
Collapse
Affiliation(s)
- Zeynab Marzhoseyni
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Iran, Sari, Iran
| | - Zahra Shaghaghi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maria Shirvani
- Infectious Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
36
|
Gu Q, Yan J, Lou Y, Zhang Z, Li Y, Zhu Z, Liu M, Wu D, Liang Y, Pu J, Zhao X, Xiao H, Li P. Bacteriocins: Curial guardians of gastrointestinal tract. Compr Rev Food Sci Food Saf 2024; 23:e13292. [PMID: 38284593 DOI: 10.1111/1541-4337.13292] [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: 08/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024]
Abstract
The human gastrointestinal (GI) tract microbiome secretes various metabolites that play pivotal roles in maintaining host physiological balance and influencing disease progression. Among these metabolites, bacteriocins-small, heat-stable peptides synthesized by ribosomes-are notably prevalent in the GI region. Their multifaceted benefits have garnered significant interest in the scientific community. This review comprehensively explores the methods for mining bacteriocins (traditional separation and purification, bioinformatics, and artificial intelligence), their effects on the stomach and intestines, and their complex bioactive mechanisms. These mechanisms include flora regulation, biological barrier restoration, and intervention in epithelial cell pathways. By detailing each well-documented bacteriocin, we reveal the diverse ways in which bacteriocins interact with the GI environment. Moreover, the future research direction is prospected. By further studying the function and interaction of intestinal bacteriocins, we can discover new pharmacological targets and develop drugs targeting intestinal bacteriocins to regulate and improve human health. It provides innovative ideas and infinite possibilities for further exploration, development, and utilization of bacteriocins. The inevitable fact is that the continuously exploration of bacteriocins is sure to bring the promising future for demic GI health understanding and interference strategy.
Collapse
Affiliation(s)
- Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Yan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yeqing Lou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zihao Zhang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yonglu Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zichun Zhu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Manman Liu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Danli Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ying Liang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Pu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodan Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
37
|
Zhang C, Li G, Lu T, Liu L, Sui Y, Bai R, Li L, Sun B. The Interaction of Microbiome and Pancreas in Acute Pancreatitis. Biomolecules 2023; 14:59. [PMID: 38254659 PMCID: PMC10813032 DOI: 10.3390/biom14010059] [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: 11/29/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Acute pancreatitis (AP) is a common acute abdomen disease characterized by the pathological activation of digestive enzymes and the self-digestion of pancreatic acinar cells. Secondary infection and sepsis are independent prognosticators for AP progression and increased mortality. Accumulating anatomical and epidemiological evidence suggests that the dysbiosis of gut microbiota affects the etiology and severity of AP through intestinal barrier disruption, local or systemic inflammatory response, bacterial translocation, and the regulatory role of microbial metabolites in AP patients and animal models. Recent studies discussing the interactions between gut microbiota and the pancreas have opened new scopes for AP, and new therapeutic interventions that target the bacteria community have received substantial attention. This review concentrates on the alterations of gut microbiota and its roles in modulating gut-pancreas axis in AP. The potential therapies of targeting microbes as well as the major challenges of applying those interventions are explored. We expect to understand the roles of microbes in AP diagnosis and treatment.
Collapse
Affiliation(s)
- Can Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Guanqun Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Tianqi Lu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Liwei Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Yuhang Sui
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Rui Bai
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; (C.Z.)
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin 150001, China
| |
Collapse
|
38
|
Abramov VM, Kosarev IV, Machulin AV, Deryusheva EI, Priputnevich TV, Panin AN, Chikileva IO, Abashina TN, Manoyan AM, Akhmetzyanova AA, Blumenkrants DA, Ivanova OE, Papazyan TT, Nikonov IN, Suzina NE, Melnikov VG, Khlebnikov VS, Sakulin VK, Samoilenko VA, Gordeev AB, Sukhikh GT, Uversky VN, Karlyshev AV. Anti- Salmonella Defence and Intestinal Homeostatic Maintenance In Vitro of a Consortium Containing Limosilactobacillus fermentum 3872 and Ligilactobacillus salivarius 7247 Strains in Human, Porcine, and Chicken Enterocytes. Antibiotics (Basel) 2023; 13:30. [PMID: 38247590 PMCID: PMC10812507 DOI: 10.3390/antibiotics13010030] [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: 11/07/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Limosilactobacillus fermentum strain 3872 (LF3872) was originally isolated from the breast milk of a healthy woman during lactation and the breastfeeding of a child. Ligilactobacillus salivarius strain 7247 (LS7247) was isolated at the same time from the intestines and reproductive system of a healthy woman. The genomes of these strains contain genes responsible for the production of peptidoglycan-degrading enzymes and factors that increase the permeability of the outer membrane of Gram-negative pathogens. In this work, the anti-Salmonella and intestinal homeostatic features of the LF3872 and LS7247 consortium were studied. A multi-drug resistant (MDR) strain of Salmonella enteritidis (SE) was used in the experiments. The consortium effectively inhibited the adhesion of SE to intact and activated human, porcine, and chicken enterocytes and reduced invasion. The consortium had a bactericidal effect on SE in 6 h of co-culturing. A gene expression analysis of SE showed that the cell-free supernatant (CFS) of the consortium inhibited the expression of virulence genes critical for the colonization of human and animal enterocytes. The CFS stimulated the production of an intestinal homeostatic factor-intestinal alkaline phosphatase (IAP)-in Caco-2 and HT-29 enterocytes. The consortium decreased the production of pro-inflammatory cytokines IL-8, TNF-α, and IL-1β, and TLR4 mRNA expression in human and animal enterocytes. It stimulated the expression of TLR9 in human and porcine enterocytes and stimulated the expression of TLR21 in chicken enterocytes. The consortium also protected the intestinal barrier functions through the increase of transepithelial electrical resistance (TEER) and the inhibition of paracellular permeability in the monolayers of human and animal enterocytes. The results obtained suggest that a LF3872 and LS7247 consortium can be used as an innovative feed additive to reduce the spread of MDR SE among the population and farm animals.
Collapse
Affiliation(s)
- Vyacheslav M. Abramov
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Igor V. Kosarev
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Andrey V. Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Evgenia I. Deryusheva
- Institute for Biological Instrumentation, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Tatiana V. Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Alexander N. Panin
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Irina O. Chikileva
- Blokhin National Research Center of Oncology, Ministry of Health RF, 115478 Moscow, Russia
| | - Tatiana N. Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Ashot M. Manoyan
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Anna A. Akhmetzyanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Dmitriy A. Blumenkrants
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | - Olga E. Ivanova
- Federal Service for Veterinary and Phytosanitary Surveillance (Rosselkhoznadzor) Federal State Budgetary Institution “The Russian State Center for Animal Feed and Drug Standardization and Quality” (FGBU VGNKI), 123022 Moscow, Russia
| | | | - Ilia N. Nikonov
- Federal State Educational Institution of Higher Professional Education, Moscow State Academy of Veterinary Medicine and Biotechnology Named after K.I. Skryabin, 109472 Moscow, Russia;
| | - Nataliya E. Suzina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Vyacheslav G. Melnikov
- Gabrichevsky Research Institute for Epidemiology and Microbiology, 125212 Moscow, Russia
| | | | - Vadim K. Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Russia; (V.S.K.); (V.K.S.)
| | - Vladimir A. Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center “Pushchino Scientific Center for Biological Research of Russian Academy of Science”, Russian Academy of Science, 142290 Pushchino, Russia
| | - Alexey B. Gordeev
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Gennady T. Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Ministry of Health, 117997 Moscow, Russia (A.B.G.)
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Andrey V. Karlyshev
- Department of Biomolecular Sciences, School of Life Sciences, Chemistry and Pharmacy, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
39
|
Dos Santos VHB, de Azevedo Ximenes ECP, de Souza RAF, da Silva RPC, da Conceição Silva M, de Andrade LVM, de Souza Oliveira VM, de Melo-Júnior MR, Costa VMA, de Barros Lorena VM, de Araújo HDA, de Lima Aires A, de Azevedo Albuquerque MCP. Effects of the probiotic Bacillus cereus GM on experimental schistosomiasis mansoni. Parasitol Res 2023; 123:72. [PMID: 38148420 DOI: 10.1007/s00436-023-08090-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/09/2023] [Indexed: 12/28/2023]
Abstract
Probiotics contribute to the integrity of the intestinal mucosa and preventing dysbiosis caused by opportunistic pathogens, such as intestinal helminths. Bacillus cereus GM obtained from Biovicerin® was cultured to obtain spores for in vivo evaluation on experimental schistosomiasis. The assay was performed for 90 days, where all animals were infected with 50 cercariae of Schistosoma mansoni on the 15th day. Three experimental groups were formed, as follows: G1-saline solution from the 1st until the 90th day; G2-B. cereus GM (105 spores in 300 μL of sterile saline) from the 1st until the 90th day; and G3-B. cereus GM 35th day (onset of oviposition) until the 90th day. G2 showed a significant reduction of 43.4% of total worms, 48.8% of female worms and 42.5% of eggs in the liver tissue. In G3, the reduction was 25.2%, 29.1%, and 44% of the total number of worms, female worms, and eggs in the liver tissue, respectively. G2 and G3 showed a 25% (p < 0.001) and 22% (p < 0.001) reduction in AST levels, respectively, but ALT levels did not change. ALP levels were reduced by 23% (p < 0.001) in the G2 group, but not in the G3. The average volume of granulomas reduced (p < 0.0001) 65.2% and 46.3% in the liver tissue and 83.0% and 53.2% in the intestine, respectively, in groups G2 and G3. Th1 profile cytokine (IFN-γ, TNF-α, and IL-6) and IL-17 were significantly increased (p < 0.001) stimulated with B. cereus GM in groups G2 and G3. IL-4 showed significant values when the stimulus was mediated by ConA. By modulating the immune response, B. cereus GM reduced the burden of worms, improved some markers of liver function, and reduced the granulomatous inflammatory reaction in mice infected with S. mansoni, especially when administered before infection.
Collapse
Affiliation(s)
- Victor Hugo Barbosa Dos Santos
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Eulália Camelo Pessoa de Azevedo Ximenes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil
- Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Renan Andrade Fernandes de Souza
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | | | | | - Valdenia Maria de Souza Oliveira
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Vlaudia Maria Assis Costa
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Departamento de Patologia, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Hallysson Douglas Andrade de Araújo
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Laboratório de Biotecnologia e Fármacos e Laboratório de Tecnologia de Biomateriais - Centro Acadêmico de Vitória de Santo Antão, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - André de Lima Aires
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil
- Centro de Ciências Médicas, Área Acadêmica de Medicina Tropical, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Mônica Camelo Pessoa de Azevedo Albuquerque
- Instituto Keizo Asami (iLIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil.
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, PE, Brazil.
- Centro de Ciências Médicas, Área Acadêmica de Medicina Tropical, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
40
|
Ferdiansyah MK, Kang HS, Kim GY, Park B, Kularathna RMRE, Abraha HB, Kim KP. Purine nucleosidase (PNase) activity, probiotics potential, and food applicability of a newly-isolated Levilactobacillus brevis LAB42. FOOD SCI TECHNOL INT 2023:10820132231219859. [PMID: 38115801 DOI: 10.1177/10820132231219859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Hyperuricemia, a condition characterized by elevated levels of uric acid in the blood, is known as a risk factor for gout disease. In this study, we isolated a total of 72 MRS-grown colonies and evaluated their purine nucleosidase (PNase) activity. Among the isolated bacteria, Levilactobacillus (L.) brevis LAB42 displayed the highest PNase activity. Our findings also indicate that PNase activity can vary among lactic acid bacterial strains and during different growth phases. Based on the kinetics study, LAB42 consistently exhibits the highest PNase activity. Due to its ability to attach to Caco-2 cells and its resistance to acidic environments and bile exposure, L. brevis LAB42 was chosen for further studies and showed that with the right combination of additives, it has the potential to be an appropriate starter for milk fermentation.
Collapse
Affiliation(s)
- Mokhammad Khoiron Ferdiansyah
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Hai-Seong Kang
- Division of Food Microbiology, National Institute of Food and Drug Safety Evaluation, Cheongju, South Korea
| | - Ga Yeong Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Beomseok Park
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Ramesha M R E Kularathna
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Haftom Baraki Abraha
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| | - Kwang-Pyo Kim
- Department of Food Science and Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
- Department of Agricultural Convergence Technology, College of Agriculture and Life Sciences, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
41
|
Quan Z, Zhang X, Wang S, Meng Y. Causal analysis of the gut microbiota in differentiated thyroid carcinoma: a two-sample Mendelian randomization study. Front Genet 2023; 14:1299930. [PMID: 38155712 PMCID: PMC10753834 DOI: 10.3389/fgene.2023.1299930] [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: 09/23/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023] Open
Abstract
Objective: Numerous studies have highlighted an association between the gut microbiota (GM) and thyroid tumors. Employing Mendelian randomization methodology, we seek to elucidate the causal link between the gut microbiota and thyroid neoplasms. Methods: We procured data from the Mibiogen database encompassing 211 distinct gut microbiota taxa, alongside extensive genome-wide association studies (GWAS) summary data for differentiated thyroid carcinoma (DTC). Our principal analytical approach involved the application of the Inverse-Variance Weighted method (IVW) within the framework of Mendelian randomization. Simultaneously, we conducted sensitivity analyses to assess result heterogeneity, horizontal pleiotropy, and outcome stability. Results: IVW analysis revealed a dual role of the GM in thyroid carcinoma. The phylum Actinobacteria (OR, 0.249 [95% CI, 0.121-0.515]; p < 0.001) was associated with a decreased risk of DTC. Conversely, the genus Ruminiclostridium9 (OR, 11.276 [95% CI, 4.406-28.860]; p < 0.001), class Mollicutes (OR, 5.902 [95% CI, 1.768-19.699]; p = 0.004), genus RuminococcaceaeUCG004 (OR, 3.831 [95% CI, 1.516-9.683]; p = 0.005), genus Paraprevotella (OR, 3.536 [95% CI, 1.330-9.401]; p = 0.011), and phylum Tenericutes (OR, 5.902 [95% CI, 1.768-19.699]; p = 0.004) were associated with an increased risk of DTC. Conclusion: Our findings underscore that the presence of genus Ruminiclostridium9, class Mollicutes, genus RuminococcaceaeUCG004, genus Paraprevotella, and phylum Tenericutes is associated with an elevated risk of DTC, whereas the presence of the phylum Actinobacteria is linked to a decreased risk. These discoveries enhance our comprehension of the relationship between the GM and DTC.
Collapse
Affiliation(s)
- Zheng Quan
- Department of Oncology Surgery, The Affiliated Hospital of Northwest University, Xi’an, China
| | - Xiaoyu Zhang
- Department of Intensive Care Unit, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shilong Wang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yong Meng
- Department of Oncology Surgery, The Affiliated Hospital of Northwest University, Xi’an, China
| |
Collapse
|
42
|
Kaur S, Sharma P, Mayer MJ, Neuert S, Narbad A, Kaur S. Beneficial effects of GABA-producing potential probiotic Limosilactobacillus fermentum L18 of human origin on intestinal permeability and human gut microbiota. Microb Cell Fact 2023; 22:256. [PMID: 38087304 PMCID: PMC10717626 DOI: 10.1186/s12934-023-02264-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) is a non-protein amino acid with neuroinhibitory, antidiabetic, and antihypertensive properties and is used as a drug for treating anxiety and depression. Some strains of lactobacilli are known to produce GABA and strengthen the gut barrier function which play an important role in ameliorating the effects caused by the pathogen on the gut barrier. The probiotic bacteria are also known to modulate the human fecal microbiota, however, the role of GABA-producing strains on the gut epithelium permeability and gut microbiota is not known. RESULTS In this study, we report the production of high levels of GABA by potential probiotic bacterium Limosilactobacillus fermentum L18 for the first time. The kinetics of the production of GABA by L18 showed that the maximum production of GABA in the culture supernatant (CS) occurred at 24 h, whereas in fermented milk it took 48 h of fermentation. The effect of L18 on the restoration of lipopolysaccharide (LPS)-disrupted intestinal cell membrane permeability in Caco-2 monolayers showed that it significantly restored the transepithelial electrical resistance (TEER) values, by significantly increasing the levels of junction proteins, occludin and E-cadherin in L18 and LPS-treated Caco-2 cells as compared to only LPS-treated cells. The effect of GABA-secreting L18 on the metataxonome of human stool samples from healthy individuals was investigated by a batch fermentor that mimics the conditions of the human colon. Although, no differences were observed in the α and β diversities of the L18-treated and untreated samples at 24 h, the relative abundances of bacterial families Lactobacillaceae and Bifidobacteriaceae increased in the L18-treated group, but both decreased in the untreated groups. On the other hand, the relative abundance of Enterobacteriaceae decreased in the L18 samples but it increased in the untreated samples. CONCLUSION These results indicate that Li. fermentum L18 is a promising GABA-secreting strain that strengthens the gut epithelial barrier by increasing junction protein concentrations and positively modulating the gut microbiota. It has the potential to be used as a psychobiotic or for the production of functional foods for the management of anxiety-related illnesses.
Collapse
Affiliation(s)
- Sumanpreet Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Medical Laboratory Sciences, Lovely Professional University, Jalandhar, India
| | - Preeti Sharma
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
| | - Melinda J Mayer
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Saskia Neuert
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- East Genomics Laboratory Hub, Cambridge University Hospitals Genomic Laboratory, Hills Road, Cambridge, UK
| | - Arjan Narbad
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India.
| |
Collapse
|
43
|
Shao Y, Wu X, Yu Z, Li M, Sheng T, Wang Z, Tu J, Song X, Qi K. Gut Microbiome Analysis and Screening of Lactic Acid Bacteria with Probiotic Potential in Anhui Swine. Animals (Basel) 2023; 13:3812. [PMID: 38136849 PMCID: PMC10741066 DOI: 10.3390/ani13243812] [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: 10/08/2023] [Revised: 11/09/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
With the widespread promotion of the green feeding concept of "substitution and resistance", there is a pressing need for alternative products in feed and breeding industries. Employing lactic acid bacteria represents one of the most promising antimicrobial strategies to combat infections caused by pathogenic bacteria. As such, we analyzed the intestinal tract of Anhui local pig breeds, including LiuBai Pig, YueHei Pig, and HuoShou Pig, to determine the composition and diversity of intestinal microbiota using 16S rRNA. Further, the functionality of the pigs' intestinal microbiota was studied through metagenomic sequencing. This study revealed that lactic acid bacteria were the primary contributors to the functional composition, as determined through a species functional contribution analysis. More specifically, the functional contribution of lactic acid bacteria in the HuoShou Pig group was higher than that of the LiuBai Pig and YueHei Pig. Subsequently, the intestinal contents of the HuoShou Pig group were selected for the screening of the dominant lactic acid bacteria strains. Out of eight strains of lactic acid bacteria, the acid-production capacity, growth curve, and tolerance to a simulated intestinal environment were assessed. Additional assessments included surface hydrophobicity, the self-aggregation capability, co-agglutination of lactic acid bacteria with pathogenic bacteria, and an in vitro bacteriostatic activity assay. Lactobacillus johnsonii L5 and Lactobacillus reuteri L8 were identified as having a strong overall performance. These findings serve as a theoretical basis for the further development of pig-derived probiotics, thereby promoting the application of lactic acid bacteria to livestock production.
Collapse
Affiliation(s)
- Ying Shao
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiaoyan Wu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhaorong Yu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Min Li
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tingting Sheng
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhenyu Wang
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jian Tu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiangjun Song
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kezong Qi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (X.W.); (Z.Y.); (M.L.); (T.S.); (Z.W.); (J.T.)
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
44
|
He X, Zhang W, Feng P, Mai Z, Gong X, Zhang G. Role of Surface Coverage of Sessile Probiotics in Their Interplay with Pathogen Bacteria Investigated by Digital Holographic Microscopy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:17308-17317. [PMID: 37974298 DOI: 10.1021/acs.langmuir.3c02436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
The adhesion of probiotics plays an important role in the gastrointestinal tract. Understanding the effect of the coverage of colonized probiotics on enteric pathogens is critical for the design of effective probiotic therapies. In the present work, we have investigated the adaptive behaviors of the intestinal pathogenic bacteria Enterobacter sakazakii (ES) near the surfaces coated with a probiotic─Lactobacillus rhamnosus GG (LGG) as a function of surface coverage ratio (CRLGG) by using a home-setup digital holographic microscopy. It shows that ES cells can adaptively sense LGG within a distance of 4.2 μm, even at CRLGG values as low as 0.05%. The growth inhibition of ES cells slightly varies with CRLGG, but the near-surface acceleration and accumulation of ES cells have much dependence on CRLGG. As CRLGG increases from 0.05 to 24.6%, the percentage of actively swimming ES, the motion bias, the acceleration, and the interplay duration do not linearly vary with CRLGG. Instead, each of them shows an extreme at CRLGG of 13.4%, corresponding to the chemotaxis behaviors of ES cells induced by diffusing stimuli (organic acids, bacteriocins, etc.) released from LGG, which showed an extreme concentration gradient at CRLGG = 13.4% by simulations. Our study clearly demonstrates that surface coverage of sessile probiotics profoundly influences their interplay with pathogen bacteria, which should be taken into account in designing probiotic therapies.
Collapse
Affiliation(s)
- Xintong He
- Faculty of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| | - Weixiong Zhang
- Faculty of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| | - Pu Feng
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou 510641, P. R. China
| | - Zhihui Mai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, P. R. China
| | - Xiangjun Gong
- Faculty of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
- Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates (South China University of Technology), Guangzhou 510640, P. R. China
| | - Guangzhao Zhang
- Faculty of Materials Science and Engineering, South China University of Technology, Guangzhou 510640, P. R. China
| |
Collapse
|
45
|
Itani K, Marcussen C, Rocha SDC, Kathiresan P, Mydland LT, Press CM, Xie Z, Tauson AH, Øverland M. Effect of Cyberlindnera jadinii yeast on growth performance, nutrient digestibility, and gut health of broiler chickens from 1 to 34 d of age. Poult Sci 2023; 102:103127. [PMID: 37837676 PMCID: PMC10585334 DOI: 10.1016/j.psj.2023.103127] [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: 05/15/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/16/2023] Open
Abstract
The effect of dietary graded levels of Cyberlindnera jadinii yeast (C. jadinii) on growth performance, nutrient digestibility, and gut health of broilers was evaluated from 1 to 34 d of age. A total of 360 male broiler chicks were randomly allocated to 1 of 4 dietary treatments (6 replicate pens each) consisting of a wheat-soybean meal-based pelleted diet (Control or CJ0), and 3 diets in which 10% (CJ10), 20% (CJ20), and 30% (CJ30) of the crude protein were supplied by C. jadinii, by gradually replacing protein-rich ingredients. Body weight and feed intake were measured at d 1, 11, 22, and 32. Pellet temperature, durability, and hardness increased linearly (P < 0.05) with C. jadinii inclusion, with highest (P < 0.05) values for CJ30. Up until d 22, feed conversion ratio (FCR) was similar between treatments (P = 0.169). Overall, increasing C. jadinii inclusion linearly increased (P = 0.047) feed intake but had no effect on weight gain or mortality. FCR increased (P < 0.05) linearly with increasing C. jadinii inclusion but only birds fed CJ30 had a significantly poorer FCR compared to the Control. Ileal digestibility was not affected by C. jadinii inclusion, however, there was a significant linear decrease in crude protein and phosphorus, and a tendency for a decrease in fat digestibility. Apparent metabolizable energy (AME) decreased (P < 0.001) quadratically with increasing C. jadinii and was significantly lower in CJ30 compared to the Control. Ileal concentrations of volatile fatty acids (VFAs) were not affected by C. jadinii inclusion, but butyric acid and total VFAs were linearly and quadratically increased and were significantly higher in cecal digesta of birds fed CJ20 and CJ30. Increasing C. jadinii inclusion was associated with an increase (P < 0.05) in the relative abundance of lactobacillus in the ileum and cecum. In conclusion, C. jadinii yeast can supply up to 20% of the total dietary protein without negatively affecting performance, digestibility, or gut health of broilers. The potential confounding role of feed processing and C. jadinii cell wall components on broiler performance is discussed.
Collapse
Affiliation(s)
- Khaled Itani
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway
| | - Caroline Marcussen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegärdsvej 3, 1870 Frederiksberg C, Denmark; Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlagevej 16, 1870 Frederiksberg C, Denmark
| | - Sérgio D C Rocha
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway
| | - Purushothaman Kathiresan
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway
| | - Liv Torunn Mydland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway
| | - Charles McLean Press
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway
| | - Zhuqing Xie
- Department of Food Science, Faculty of Science, University of Copenhagen, Rolighedsvej 26,1958 Frederiksberg C, Denmark
| | - Anne-Helene Tauson
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway; Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Gronnegärdsvej 3, 1870 Frederiksberg C, Denmark
| | - Margareth Øverland
- Department of Animal and Aquacultural Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, P.O. Box 5003, 1432 Aas, Norway.
| |
Collapse
|
46
|
Jesser KJ, Trueba G, Konstantinidis KT, Levy K. Why are so many enteric pathogen infections asymptomatic? Pathogen and gut microbiome characteristics associated with diarrhea symptoms and carriage of diarrheagenic E. coli in northern Ecuador. Gut Microbes 2023; 15:2281010. [PMID: 37992406 PMCID: PMC10730187 DOI: 10.1080/19490976.2023.2281010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/05/2023] [Indexed: 11/24/2023] Open
Abstract
A high proportion of enteric infections, including those caused by diarrheagenic Escherichia coli (DEC), are asymptomatic for diarrhea. The factors responsible for the development of diarrhea symptoms, or lack thereof, remain unclear. Here, we used DEC isolate genome and whole stool microbiome data from a case-control study of diarrhea in Ecuador to examine factors associated with diarrhea symptoms accompanying DEC carriage. We investigated i) pathogen abundance, ii) gut microbiome characteristics, and iii) strain-level pathogen characteristics from DEC infections with diarrhea symptoms (symptomatic infections) and without diarrhea symptoms (asymptomatic infections). We also included data from individuals with and without diarrhea who were not infected with DEC (uninfected cases and controls). i) E. coli relative abundance in the gut microbiome was highly variable, but higher on-average in individuals with symptomatic compared to asymptomatic DEC infections. Similarly, the number and relative abundances of virulence genes in the gut were higher in symptomatic than asymptomatic DEC infections. ii) Measures of microbiome diversity were similar regardless of diarrhea symptoms or DEC carriage. Proteobacterial families that have been described as pathobionts were enriched in symptomatic infections and uninfected cases, whereas potentially beneficial taxa, including the Bacteroidaceae and Bifidobacteriaceae, were more abundant in individuals without diarrhea. An analysis of high-level gene functions recovered in metagenomes revealed that genes that were differentially abundant by diarrhea and DEC infection status were more abundant in symptomatic than asymptomatic DEC infections. iii) DEC isolates from symptomatic versus asymptomatic individuals showed no significant differences in virulence or accessory gene content, and there was no phylogenetic signal associated with diarrhea symptoms. Together, these data suggest signals that distinguish symptomatic from asymptomatic DEC infections. In particular, the abundance of E. coli, the virulence gene content of the gut microbiome, and the taxa present in the gut microbiome have an apparent role.
Collapse
Affiliation(s)
- Kelsey J Jesser
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Gabriel Trueba
- Instituto de Microbiología, Universidad San Francisco de Quito, Quito, Ecuador
| | - Konstantinos T. Konstantinidis
- School of Civil and Environmental Engineering and School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Karen Levy
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
47
|
Ye Q, Lao L, Zhang A, Qin Y, Zong M, Pan D, Yang H, Wu Z. Multifunctional properties of the transmembrane LPxTG-motif protein derived from Limosilactobacillus reuteri SH-23. J Dairy Sci 2023; 106:8207-8220. [PMID: 37641365 DOI: 10.3168/jds.2023-23440] [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: 03/02/2023] [Accepted: 05/16/2023] [Indexed: 08/31/2023]
Abstract
The LPxTG-motif protein is an important transmembrane protein with high hydrophilicity and stability, as evidenced by its stress tolerance and adhesion ability. In this study, a novel LPxTG-motif protein with esterase activity (LEP) was expressed, and the multifunctional properties such as adhesion properties and esterase activity were also investigated. When cocultured with Limosilactobacillus reuteri SH-23, the adhesion ability of L. reuteri SH-23 to HT-29 cells was improved, and this adhesion was further found relating to the potential target protein Pyruvate kinase M1/2 (PKM) of HT-29 cells. In addition, as a multifunctional protein, LEP can promote the hydrolysis of bovine milk lipids with its esterase activity, and the activity was enhanced in the presence of Zn2+ and Mn2+ at pH 7. Furthermore, the polyunsaturated fatty acids (PUFA) such as linoleic acid and eicosapentaenoic acid were found to increase during the hydrolyzing process. These unique properties of LEP provide a comprehensive understanding of the adhesion function and PUFA releasing properties of the multifunctional protein derived from L. reuteri SH-23 and shed light on the beneficial effect of this Lactobacillus strain on the colonization of the gastrointestinal tract.
Collapse
Affiliation(s)
- Qianwen Ye
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Lifeng Lao
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Ao Zhang
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Yiman Qin
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Manli Zong
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China
| | - Hua Yang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315211, Zhejiang, P. R. China.
| | - Zhen Wu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, Zhejiang, P. R. China.
| |
Collapse
|
48
|
Lee BH, Chen YZ, Shen TL, Pan TM, Hsu WH. Proteomic characterization of extracellular vesicles derived from lactic acid bacteria. Food Chem 2023; 427:136685. [PMID: 37356267 DOI: 10.1016/j.foodchem.2023.136685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/08/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
Lactobacillus species confer health benefits by their metabolites, secreted molecules, and population numbers. Extracellular vesicles (EVs) are nano-sized particles released from cells and mediate intercellular communications. EVs-encapsulated cargos are a crucial key to decide involved biological function. However, little is known about the composition of EVs, leaving mechanisms by which Lactobacillus-derived EVs affect recipient cells remaining unresolved. This study examined the composition of EV proteins from Lactobacillus species by using liquid chromatography coupled with tandem mass spectrometry, including L. plantarum, L. fermentum, and L. gasseri. The major proteins of EVs are associated with biological processes such as catalytic activity, gluco-neogenesis, cell wall organization, and glycolytic processes. Motif enrichment analysis revealed that EVs from L. plantarum and L. fermentum contained proteins with serine-rich motif. This is the first study to report the composition and comparison of EV proteins from Lactobacillus species, providing important information of EVs in functional food products development.
Collapse
Affiliation(s)
- Bao-Hong Lee
- Department of Horticulture, National Chiayi University, Chiayi 60004, Taiwan
| | - You-Zuo Chen
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan
| | - Tang-Long Shen
- Department of Plant Pathology and Microbiology, National Taiwan University, Taipei 106319, Taiwan; Center for Biotechnology, National Taiwan University, Taipei 106319, Taiwan
| | - Tzu-Ming Pan
- Department of Research and Development Division, SunWay Biotech Co., Ltd., Taipei 114067, Taiwan; Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei 106319, Taiwan
| | - Wei-Hsuan Hsu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan.
| |
Collapse
|
49
|
Kim S, Lee S, Kim TY, Lee SH, Seo SU, Kweon MN. Newly isolated Lactobacillus paracasei strain modulates lung immunity and improves the capacity to cope with influenza virus infection. MICROBIOME 2023; 11:260. [PMID: 37996951 PMCID: PMC10666316 DOI: 10.1186/s40168-023-01687-8] [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: 05/23/2023] [Accepted: 10/01/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND The modulation of immune responses by probiotics is crucial for local and systemic immunity. Recent studies have suggested a correlation between gut microbiota and lung immunity, known as the gut-lung axis. However, the evidence and mechanisms underlying this axis remain elusive. RESULTS In this study, we screened various Lactobacillus (L.) strains for their ability to augment type I interferon (IFN-I) signaling using an IFN-α/β reporter cell line. We identified L. paracasei (MI29) from the feces of healthy volunteers, which showed enhanced IFN-I signaling in vitro. Oral administration of the MI29 strain to wild-type B6 mice for 2 weeks resulted in increased expression of IFN-stimulated genes and pro-inflammatory cytokines in the lungs. We found that MI29-treated mice had significantly increased numbers of CD11c+PDCA-1+ plasmacytoid dendritic cells and Ly6Chi monocytes in the lungs compared with control groups. Pre-treatment with MI29 for 2 weeks resulted in less weight loss and lower viral loads in the lung after a sub-lethal dose of influenza virus infection. Interestingly, IFNAR1-/- mice did not show enhanced viral resistance in response to oral MI29 administration. Furthermore, metabolic profiles of MI29-treated mice revealed changes in fatty acid metabolism, with MI29-derived fatty acids contributing to host defense in a Gpr40/120-dependent manner. CONCLUSIONS These findings suggest that the newly isolated MI29 strain can activate host defense immunity and prevent infections caused by the influenza virus through the gut-lung axis. Video Abstract.
Collapse
Affiliation(s)
- Seungil Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sohyeon Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Tae-Young Kim
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Su-Hyun Lee
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea
| | - Sang-Uk Seo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, Republic of Korea.
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Deng Y, Pan J, Yang X, Yang S, Chi H, Yang X, Qu X, Sun S, You L, Hou C. Dual roles of nanocrystalline cellulose extracted from jute ( Corchorus olitorius L.) leaves in resisting antibiotics and protecting probiotics. NANOSCALE ADVANCES 2023; 5:6435-6448. [PMID: 38024324 PMCID: PMC10662138 DOI: 10.1039/d3na00345k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 08/14/2023] [Indexed: 12/01/2023]
Abstract
Antibiotics can cure diseases caused by bacterial infections, but their widespread use can have some side effects, such as probiotic reduction. There is an urgent need for such agents that can not only alleviate the damage caused by antibiotics, but also maintain the balance of the gut microbiota. In this study, we first characterized the nanocrystalline cellulose (NCC) extracted from plant jute (Corchorus olitorius L.) leaves. Next, we evaluated the protective effect of jute NCC and cellulose on human model gut bacteria (Lacticaseibacillus rhamnosus and Escherichia coli) under antibiotic stress by measuring bacterial growth and colony forming units. We found that NCC is more effective than cellulose in adsorbing antibiotics and defending the gut bacteria E. coli. Interestingly, the low-dose jute NCC clearly maintained the balance of key gut bacteria like Snodgrassella alvi and Lactobacillus Firm-4 in bees treated with tetracycline and reduced the toxicity caused by antibiotics. It also showed a more significant protective effect on human gut bacteria, especially L. rhamnosus, than cellulose. This study first demonstrated that low-dose NCC performed satisfactorily as a specific probiotic to mitigate the adverse effects of antibiotics on gut bacteria.
Collapse
Affiliation(s)
- Yanchun Deng
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Jiangpeng Pan
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Xiai Yang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Sa Yang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences Beijing 100093 P. R. China
- Graduate School of Chinese Academy of Agricultural Sciences Beijing 100081 P. R. China
| | - Haiyang Chi
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Xiushi Yang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Xiaoxin Qu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Shitao Sun
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| | - Linfeng You
- Department of Food and Biotechnology Engineering, Chongqing Technology and Business University Chongqing 400067 P. R. China
| | - Chunsheng Hou
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences Changsha 410205 P. R. China
| |
Collapse
|