1
|
Abdullahi AM, Zhao S, Xu Y. Efficacy of probiotic supplementation in preventing necrotizing enterocolitis in preterm infants: a systematic review and meta-analysis. J Matern Fetal Neonatal Med 2025; 38:2485215. [PMID: 40204632 DOI: 10.1080/14767058.2025.2485215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/27/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a severe condition in preterm infants, involving intestinal inflammation and bacterial invasion, leading to high morbidity and mortality. Probiotics may reduce NEC by promoting beneficial gut bacteria, but the role of Bifidobacterium bifidum G001 (BBG001) is not well understood. This meta-analysis evaluates the effectiveness of BBG001 versus placebo in preventing NEC, late-onset sepsis, and all-cause mortality in preterm infants. METHODS Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a systematic review and meta-analysis of randomized controlled trials (RCTs) from 2014 to 2024. Databases searched included PubMed, Cochrane Central Register, MEDLINE, and EMBASE. Inclusion criteria encompassed RCTs involving preterm infants (<37 weeks gestation and/or <2500 g birth weight) receiving enteral probiotics, including BBG001, compared to placebo. Outcomes assessed were incidence of NEC (≥stage II), late-onset sepsis, and all-cause mortality. Data extraction and quality assessment were independently performed. RESULTS From 430 identified records, nine studies involving 7180 preterm infants met the inclusion criteria. BBG001 significantly reduced the risk of NEC stage >2 (OR = 0.66; 95% CI [0.47, 0.94], p = .02), mortality and morbidity (OR = 0.74; 95% CI [0.62, 0.89], p = .001), and sepsis (OR = 0.71; 95% CI [0.53, 0.93], p = .01). It also lowered the odds of periventricular leukomalacia (OR = 0.61; 95% CI [0.47, 0.78], p < .0001) and intraventricular hemorrhage (OR = 0.48; 95% CI [0.33, 0.69], p < .0001). Additionally, BBG001 showed a protective effect against infection-related outcomes, reducing the odds of death attributed to infection (OR = 0.80; 95% CI: 0.65, 0.99; p = .04). CONCLUSIONS Probiotic supplementation with BBG001 appears effective in reducing NEC incidence, bloodstream infections, and infection-related deaths in preterm infants. Despite promising results, variability in study designs necessitates further large-scale RCTs to confirm these findings and establish BBG001's role in neonatal care.
Collapse
Affiliation(s)
| | - Shuangkui Zhao
- Department of NICU, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanping Xu
- Department of NICU, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
2
|
Sabino YNV, Paiva AD, Fonseca BR, Medeiros JD, Machado ABF. Deciphering probiotic potential: a comprehensive guide to probiogenomic analyses. Future Microbiol 2025:1-12. [PMID: 40227157 DOI: 10.1080/17460913.2025.2492472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
In recent years, the study of probiotics has advanced significantly, driven by growing interest in their potential health benefits and applications in the food and pharmaceutical industries. Probiotics are claimed to enhance gut health, modulate immune responses, improve digestion, synthesize beneficial compounds for the host, and even impact mental health through the gut-brain axis. However, traditional in vitro methods for identifying probiotics have limitations, such as low reproducibility in phenotypic screening, limited capacity to discover new strains, restricted evaluation of safety, and inefficiencies in fully understanding the biological properties responsible for health-promoting effects. Advancements in genomic analysis technology have provided a cost-effective approach to further explore probiotic strains and enhance understanding of the molecular mechanisms driving their beneficial effects in hosts. Here, we describe a comprehensive workflow for probiogenomic analysis aimed at establishing a gold-standard pipeline for screening probiotic potential based on genome sequencing. This pipeline encompasses steps from acquiring genomes to screening for safety-related features, genomic plasticity, and probiotic markers through whole-genome sequencing. In addition, this study outlines the respective methodological approaches and provides the most comprehensive database documented to date, comprising 243 genes potentially associated with probiotic function.
Collapse
Affiliation(s)
- Yasmin Neves Vieira Sabino
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Aline Dias Paiva
- Department of Microbiology, Immunology and Parasitology, Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Bárbara Ribeiro Fonseca
- Department of Parasitology, Microbiology and Immunology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Julliane Dutra Medeiros
- Department of Biology, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | |
Collapse
|
3
|
Esam RM, Hafez RS, Khafaga NIM, Ahmed LI, Soliman TN, Fahim KM. Novel utilization of micro-encapsulated Lactobacillus acidophilus and bacterial /yeast combination enhanced the AFM 1 reduction in spiked yoghurt. Int J Food Microbiol 2025; 436:111205. [PMID: 40239292 DOI: 10.1016/j.ijfoodmicro.2025.111205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Aflatoxin M1 (AFM1) is a significant public health hazard threatening dairy food safety and the dairy industry. Therefore, the present study evaluated the effectiveness of five probiotic (viable) and parabiotic (non-viable: heat and acid-treated) strains (Bifidobacterium bifidum, Lactobacillus acidophilus, Bacillus subtilis, Lactiplantibacillus plantarum, and Saccharomyces cerevisiae) in reducing AFM1 in yoghurt over two weeks. It also explored the ability of the micro-encapsulated L. acidophilus and the viable and non-viable new bacterial/yeast combinations (L. acidophilus- B. bifidum- S. cerevisiae) as promising and new strategies to eliminate and control AFM1 in the dairy plant. All the studied strains reduced AFM1 efficiently in pro and parabiotic yoghurt compared to the control yoghurt (without fortification) (P < 0.05), with the highest efficacy in L. acidophilus. Furthermore, the bacterial/yeast combination scored a better AFM1 reduction percentage than the single treatments, with a binding percentage of 90 % in acid-treated co-culture. However, the innovative application of the encapsulated L. acidophilus with chitosan-CaCl2-alginate (Alg/CaCl2/CH) and chitosan‑sodium tripolyphosphate (CH/TPP) was considered the best treatment as they achieved fast and significant AFM1 reduction percentages of 68 and 81 %, respectively, from the first day of storage. In conclusion, these findings provided a safe and effective solution for AFM1 control in the dairy industry. Additionally, the effective reduction percentages obtained by parabiotics open the door for extensive application in non-fermented dairy foods.
Collapse
Affiliation(s)
| | - Ragaa Shehata Hafez
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | | | - Lamiaa Ibrahim Ahmed
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Tarek Nour Soliman
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Karima Mogahed Fahim
- Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| |
Collapse
|
4
|
Yarlagadda T, Carey A, Bryan E, Huygens F, Yarlagadda P, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. The response of nasal epithelial cells exposed to novel Lactobacillus and alpha-haemolytic Streptococcus isolated from the upper respiratory tract of children. J Appl Microbiol 2025; 136:lxaf071. [PMID: 40107866 DOI: 10.1093/jambio/lxaf071] [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: 07/11/2024] [Revised: 01/27/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
AIMS To investigate the response of primary nasal epithelial cells (NECs) to novel alpha haemolytic Streptococcus and lactobacilli strains, isolated from the upper respiratory tract of children. METHODS AND RESULTS Submerged cultures of NECs from healthy adult donors were exposed to either novel strains; Lactobacillus rhamnosus D3189, D3160, Streptococcus salivarius D3837; or commercially available probiotic strains L. rhamnosus LB21, S. salivarius K12; or a pathogenic strain (S. pneumoniae 49619). Cytotoxicity (measured through lactate dehydrogenase release) and cytokine release were quantified 24 hours post-exposure. Exposure to novel and commercially available strains did not induce the production of IFN-β, IFN-λ1/3, IL-1β, IL-6, IL-8, or TNF-α production or the release of LDH. Conversely, the pathogenic strain S. pneumoniae 49 619 significantly elevated the expression of IL-1β, IL-8, TNF-α, and LDH in NECs. CONCLUSIONS The findings within this study highlight the non-pathogenic nature of these novel strains and support further investigation of the potential to develop nasally administered probiotics.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Flavia Huygens
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Prasad Yarlagadda
- School of Engineering, University of Southern Queensland, Queensland 4300, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Queensland 4000, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| |
Collapse
|
5
|
Hou S, Li R, Zhang Y, Liang P, Yang H, He H, Wang L, Sun Y, Jin T, Liu Z, Xie J. Supplementation of mixed Lactobacillus alleviates metabolic impairment, inflammation, and dysbiosis of the gut microbiota in an obese mouse model. Front Nutr 2025; 12:1554996. [PMID: 40206949 PMCID: PMC11978641 DOI: 10.3389/fnut.2025.1554996] [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: 01/08/2025] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction Obesity is a complex metabolic disease, which is often accompanied with impaired glucose and lipid metabolism and chronic inflammation. Probiotics have been considered as a strategy for treating obesity, while the genus of Lactobacillus is the most commonly tested and approved probiotics. Some multi-strain probiotics were proven to produce synergistic effects on treating obesity as compared to mono-strain ones. Methods The purpose of this study was to investigate the anti-obesity effect of a new probiotic formation contained Lactobacillus plantarum L14, Lactobacillus paracasei L9, Lactobacillus rhamnosus GG, and Lactobacillus sakei X-MRS-2, designated as L-PPRS. Multi-strain probiotics L-PPRS was shown to have a better antiadipogenic effect than mono-strain probiotics in 3T3-L1 cell. Subsequently, L-PPRS was orally supplemented to a high-fat diet (HFD) induced obese mouse model for two kinds of treatment course, a short-term (8 weeks) one and a long-term (12 weeks) one. Results We found that intervention of L-PPRS not only significantly inhibited weight gain in HFD-fed mice, but also improved glucose tolerance, insulin sensitivity and reduced serum lipid levels. Furthermore, L-PPRS intervention reduced fat accumulation in the adipose tissue and the liver, and ameliorated the antioxidant capacity of liver in HFD-fed mice. L-PPRS intervention modulated the expression of lipid-metabolic genes, and exhibited excellent anti-inflammatory effect. In addition, L-PPRS intervention restored the dysbiosis of gut microbiota via reducing the Firmicutes/ Bacteroidetes ratio, and increasing the abundance of beneficial intestinal bacteria. In conclusion, this study proved that L-PPRS could effectively prevent the development of obesity and its associated abnormalities, and the long-term supplementation of L-PPRS provided a more profound benefit than the short-term. Discussion This study highlights the potential of L-PPRS as an effective anti-obesity strategy.
Collapse
Affiliation(s)
- Shulin Hou
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Ruining Li
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yunyun Zhang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Ping Liang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Haishan Yang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Huili He
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Yaojun Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Tianru Jin
- Department of Physiology, Temerty Faculty of Medicine, Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Zhizhen Liu
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jun Xie
- Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
6
|
Patra S, Pradhan B, Roychowdhury A. Complete genome sequence, metabolic profiling and functional studies reveal Ligilactobacillus salivarius LS-ARS2 is a promising biofilm-forming probiotic with significant antioxidant, antibacterial, and antibiofilm potential. Front Microbiol 2025; 16:1535388. [PMID: 40182284 PMCID: PMC11965632 DOI: 10.3389/fmicb.2025.1535388] [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: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Background Probiotics restore microbial balance and prevent gut-inflammation. Therefore, finding out novel probiotic strains is a demand. As gut-microbe, benefits of Ligilactobacillus salivarius (LS) are established. However, strain-specific detailed studies are limited. Here, we illustrate probiotic attributes of novel LS-ARS2 for its potential application as food-supplement and/or therapeutic to improve gut-health. Methods Whole genome sequencing (WGS) and phylogenetic analysis confirm the strain as LS. To establish probiotic properties, acid-bile tolerance, auto-aggregation, cell-surface-hydrophobicity, biofilm-formation, and adhesion-assays are performed. To ensure safety attributes, antibiotic-susceptibility, hemolytic, DNase, trypan-blue, and MTT assays are done. ABTS, DPPH, superoxide, hydroxyl free radical scavenging assays are used to determine anti-oxidant potential. Antibacterial assays, including co-culture assay with pathogen and pathogenic biofilm-inhibition assays, are performed to explore antibacterial efficacy. To characterize metabolic-profile of LS-ARS2-derived cell-free-supernatant (CFS), HRMS analysis are carried out. Consequently, WGS-analyses predict potential molecular associations related to functional outcomes. Results We find LS-ARS2 a remarkable fast-growing strain that shows acid and bile tolerance (>60% survival rate), indicating promising gut-sustainability. High auto-aggregation capacity (>80%), robust cell-surface hydrophobicity (>85%), and adhesion efficacy to Caco-2 cells illustrate significant potential of LS-ARS2 for gut colonization. Fascinatingly, LS-ARS2 is able to form biofilm within 24 h (p < 0.0001), rare among LS strains, indicating the potential of the strain for efficient stay in the gut. The strain ensures safety attributes. LS-ARS2-WGS analysis recognizes probiotic-specific determinants, predicts genomic stability, identifies orthologous-clusters for diverse functions, and predicts metabolites and bacteriocins. HRMS-studies with LS-ARS2-CFS further validate the presence of diverse beneficial metabolites with antimicrobial and immunomodulatory potential. LS-ARS2 shows significant antioxidant properties in ABTS (>60%), DPPH (>10 U/mL), superoxide (>70%), and hydroxyl free radical scavenging assays (>70%). Further, LS-ARS2 shows antimicrobial activities against Gram-positive Methicillin-resistant Staphylococcus aureus (MRSA) and Gram-negative multidrug-resistant clinical strains enterotoxigenic Escherichia coli, Vibrio cholerae, and Shigella flexneri. Anti-Salmonella effect of LS-ARS2 is prominent (p < 0.0001). Most interestingly, LS-ARS2-CFS inhibits MRSA-biofilm (p < 0.0001), again rare among LS strains. Conclusion LS-ARS2 is a novel, fast-growing, biofilm-forming probiotic with significant antioxidant, antibacterial, and anti-biofilm potentials, suggesting the promising applications of LS-ARS2 for combating pathogenic biofilms and improving gut-health. However, further in vivo studies would facilitate their potential applications.
Collapse
Affiliation(s)
- Sinjini Patra
- Biochemistry and Cell Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Biswaranjan Pradhan
- S. K. Dash Center of Excellence of Biosciences and Engineering & Technology (SKBET), Indian Institute of Technology Bhubaneswar, Odisha, India
| | - Anasuya Roychowdhury
- Biochemistry and Cell Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha, India
| |
Collapse
|
7
|
Li F, Armet AM, Korpela K, Liu J, Quevedo RM, Asnicar F, Seethaler B, Rusnak TBS, Cole JL, Zhang Z, Zhao S, Wang X, Gagnon A, Deehan EC, Mota JF, Bakal JA, Greiner R, Knights D, Segata N, Bischoff SC, Mereu L, Haqq AM, Field CJ, Li L, Prado CM, Walter J. Cardiometabolic benefits of a non-industrialized-type diet are linked to gut microbiome modulation. Cell 2025; 188:1226-1247.e18. [PMID: 39855197 DOI: 10.1016/j.cell.2024.12.034] [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: 05/29/2024] [Revised: 10/24/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
Industrialization adversely affects the gut microbiome and predisposes individuals to chronic non-communicable diseases. We tested a microbiome restoration strategy comprising a diet that recapitulated key characteristics of non-industrialized dietary patterns (restore diet) and a bacterium rarely found in industrialized microbiomes (Limosilactobacillus reuteri) in a randomized controlled feeding trial in healthy Canadian adults. The restore diet, despite reducing gut microbiome diversity, enhanced the persistence of L. reuteri strain from rural Papua New Guinea (PB-W1) and redressed several microbiome features altered by industrialization. The diet also beneficially altered microbiota-derived plasma metabolites implicated in the etiology of chronic non-communicable diseases. Considerable cardiometabolic benefits were observed independently of L. reuteri administration, several of which could be accurately predicted by baseline and diet-responsive microbiome features. The findings suggest that a dietary intervention targeted toward restoring the gut microbiome can improve host-microbiome interactions that likely underpin chronic pathologies, which can guide dietary recommendations and the development of therapeutic and nutritional strategies.
Collapse
Affiliation(s)
- Fuyong Li
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Animal Science and Technology, College of Animal Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Anissa M Armet
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Katri Korpela
- Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, Helsinki 00014, Uusimaa, Finland
| | - Junhong Liu
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Rodrigo Margain Quevedo
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Benjamin Seethaler
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Tianna B S Rusnak
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Janis L Cole
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Zhihong Zhang
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Shuang Zhao
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Xiaohang Wang
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada
| | - Adele Gagnon
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Edward C Deehan
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68588, USA
| | - João F Mota
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; Faculty of Nutrition, Federal University of Goiás, Goiânia, Goiás 74605-080, Brazil
| | - Jeffrey A Bakal
- Division of General Internal Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Russell Greiner
- Department of Computing Science, University of Alberta, Edmonton, AB T6G 2R3, Canada; Alberta Machine Intelligence Institute, Edmonton, AB T5J 3B1, Canada
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento 38123, Trentino, Italy
| | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Baden-Württemberg, Germany
| | - Laurie Mereu
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Andrea M Haqq
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Catherine J Field
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Liang Li
- The Metabolomics Innovation Centre, Edmonton, AB T6G 2E9, Canada; Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Carla M Prado
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jens Walter
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, AB T6G 2E1, Canada; APC Microbiome Ireland, University College Cork, Cork T12 YT20, Munster, Ireland; School of Microbiology, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Medicine, University College Cork, Cork T12 YT20, Munster, Ireland; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| |
Collapse
|
8
|
Kenneth MJ, Wu CC, Fang CY, Hsu TK, Lin IC, Huang SW, Chiu YC, Hsu BM. Exploring the Impact of Chemotherapy on the Emergence of Antibiotic Resistance in the Gut Microbiota of Colorectal Cancer Patients. Antibiotics (Basel) 2025; 14:264. [PMID: 40149075 PMCID: PMC11939702 DOI: 10.3390/antibiotics14030264] [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: 01/13/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
With nearly half of colorectal cancer (CRC) patients diagnosed at advanced stages where surgery alone is insufficient, chemotherapy remains a cornerstone for this cancer treatment. To prevent infections and improve outcomes, antibiotics are often co-administered. However, chemotherapeutic interactions with the gut microbiota cause significant non-selective toxicity, affecting not only tumor and normal epithelial cells but also the gut microbiota. This toxicity triggers the bacterial SOS response and loss of microbial diversity, leading to bacterial mutations and dysbiosis. Consequently, pathogenic overgrowth and systemic infections increase, necessitating broad-spectrum antibiotics intervention. This review underscores how prolonged antibiotic use during chemotherapy, combined with chemotherapy-induced bacterial mutations, creates selective pressures that drive de novo antimicrobial resistance (AMR), allowing resistant bacteria to dominate the gut. This compromises the treatment efficacy and elevates the mortality risk. Restoring gut microbial diversity may mitigate chemotherapy-induced toxicity and improve therapeutic outcomes, and emerging strategies, such as fecal microbiota transplantation (FMT), probiotics, and prebiotics, show considerable promise. Given the global threat posed by antibiotic resistance to cancer treatment, prioritizing antimicrobial stewardship is essential for optimizing antibiotic use and preventing resistance in CRC patients undergoing chemotherapy. Future research should aim to minimize chemotherapy's impact on the gut microbiota and develop targeted interventions to restore microbial diversity affected during chemotherapy.
Collapse
Affiliation(s)
- Mutebi John Kenneth
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
- Doctoral Program in Science, Technology, Environment and Mathematics, National Chung Cheng University, Chiayi 621, Taiwan
| | - Chin-Chia Wu
- Division of Colorectal Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 622, Taiwan
- College of Medicine, Tzu Chi University, Hualien 970, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 970, Taiwan
| | - Chuan-Yin Fang
- Division of Colon and Rectal Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 600, Taiwan
| | - Tsui-Kang Hsu
- Department of Ophthalmology, Cheng Hsin General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - I-Ching Lin
- Department of Family Medicine, Asia University Hospital, Taichung 413, Taiwan
- Department of Kinesiology, Health and Leisure, Chienkuo Technology University, Changhua 500, Taiwan
| | - Shih-Wei Huang
- Center for Environmental Toxin and Emerging Contaminant Research, Cheng Shiu University, Kaohsiung 833, Taiwan
| | - Yi-Chou Chiu
- General Surgery, Surgical Department, Cheng Hsin General Hospital, Taipei 112, Taiwan
| | - Bing-Mu Hsu
- Department of Earth and Environmental Sciences, National Chung Cheng University, Chiayi 621, Taiwan
| |
Collapse
|
9
|
Xi Z, Fenglin X, Yun Z, Chunrong L. Efficacy of probiotics in the treatment of allergic diseases: a meta-analysis. Front Nutr 2025; 12:1502390. [PMID: 40104820 PMCID: PMC11913692 DOI: 10.3389/fnut.2025.1502390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Background information Allergic diseases are an increasingly serious health issue worldwide, affecting not only the physiological health of patients but also significantly reducing their quality of life, thereby imposing a substantial economic burden on families and society. According to data from the World Health Organization, the incidence of allergic diseases has risen markedly over the past few decades, particularly among children and adolescents, making it a significant public health challenge. Although several clinical studies have explored the effects of probiotics in the treatment of food-induced allergies and allergic diseases, the results have been inconsistent. Some studies indicate positive effects, while others fail to demonstrate their efficacy. Therefore, a systematic evaluation of the effectiveness of probiotics in allergic diseases is particularly important. Some studies indicate that patients with food allergies may also experience respiratory symptoms, and certain foods may be associated with the onset or exacerbation of allergic rhinitis and asthma. Diseases such as allergic rhinitis, asthma, and atopic dermatitis are characterized by inappropriate immune responses to typically harmless environmental allergens. The incidence of these diseases is continuously rising in urban populations, prompting researchers to extensively explore novel therapeutic strategies that can effectively modulate immune responses. Objective The aim of this study is to systematically assess the effectiveness of probiotics in the treatment of allergic diseases. By integrating the results of existing clinical studies, we hope to provide a clearer scientific basis for the treatment of allergic diseases. Methods We conducted a comprehensive literature search in databases such as PubMed for articles published before the end of 2023 that evaluated the effectiveness of probiotics in treating allergic diseases. Inclusion criteria focused on studies reporting binary outcomes related to the efficacy of probiotics in patients with food allergies, asthma, allergic rhinitis, or atopic dermatitis. Data were collected using Excel software, and the Review Manager software was used to analyze the collected binary variable data. The effectiveness of probiotics was assessed by calculating the risk ratio (RR) and its 95% confidence interval (CI). Heterogeneity among studies was evaluated using the I2 statistic, and publication bias was assessed through funnel plots. Results The analysis of the aggregated binary data indicates that probiotics significantly improve clinical outcomes in patients with allergic diseases. Additional analysis using continuous variables (scores) further demonstrates the effectiveness of probiotics in alleviating allergic diseases. Subgroup analyses show that probiotics are effective in treating various common conditions, with two specific probiotics strains being particularly effective for allergic diseases. Conclusion We included literature involving pediatric patients with common allergic diseases, Probiotics can help treat allergic diseases by modulating immune mechanisms, but allergic diseases are typically caused by multiple factors and individual variations, however, allergic diseases are typically caused by multiple factors and individual variations, so they should not be used as the sole treatment method.This meta-analysis provides evidence supporting the effectiveness of probiotics in various allergic diseases. The findings suggest that probiotics can serve as a beneficial adjunctive therapy for the treatment of these conditions. Systematic review registration https://clinicaltrials.gov/, CRD42024586317.
Collapse
Affiliation(s)
- Zhang Xi
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xing Fenglin
- Chengdu Qingyang District Maternal and Child Health Hospital, Chengdu, China
| | - Zhao Yun
- Chengdu Children's Specialized Hospital, Chengdu, China
| | - Li Chunrong
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
10
|
Ji M, Rong X, Wu Y, Li H, Zhao X, Zhao Y, Guo X, Cao G, Yang Y, Li B. Effects of Fermented Liquid Feed with Compound Probiotics on Growth Performance, Meat Quality, and Fecal Microbiota of Growing Pigs. Animals (Basel) 2025; 15:733. [PMID: 40076016 PMCID: PMC11899608 DOI: 10.3390/ani15050733] [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: 01/10/2025] [Revised: 03/01/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Feed fermentation enhances feed nutrition and animal health, but its impact on the gut microbiota of young pigs remains unclear. This study aimed to evaluate the effects of a probiotic fermented feed, which includes Lactobacillus plantarum, Pediococcus pentosaceus, Bacillus subtilis, and Bacillus coagulans, on the growth performance, meat quality, and intestinal microbiota of growing pigs. We randomly assigned 24 Duroc × Landrace × Landrace pigs to two groups: a control (Ctrl) group and the fermented liquid feed (FLF) group, with three replicate pens per group and four pigs per pen. Results indicated that the FLF group experienced a significant decrease in anti-nutritional factors like α-conglycinin and β-conglycinin. In addition, the average daily gain of pigs in the FLF group increased significantly, while the feed conversion ratio and shear force decreased. HE staining showed that the FLF group had notably enhanced villus height in the jejunum and ileum. 16S rRNA sequencing revealed a marked increase in the relative abundance of certain microbes in the FLF group, which were predominantly associated with carbohydrate and amino acid metabolism. These results indicated that compound probiotic FLF can elevate feed quality, enhance the growth performance of growing pigs, and ameliorate the structure of the gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yang Yang
- Key Laboratory of Animal Science, College of Animal Science, Shanxi Agricultural University, No.1, Mingxian South Road, Taigu, Jinzhong 030801, China; (M.J.); (Y.W.); (Y.Z.); (X.G.); (G.C.)
| | - Bugao Li
- Key Laboratory of Animal Science, College of Animal Science, Shanxi Agricultural University, No.1, Mingxian South Road, Taigu, Jinzhong 030801, China; (M.J.); (Y.W.); (Y.Z.); (X.G.); (G.C.)
| |
Collapse
|
11
|
Tarik M, Ramakrishnan L, Bhatia N, Roy A, Kandasamy D, Chandran DS, Singh A, Kalaivani M, Neelamraju J, Madempudi RS. Supplementation With Bacillus clausii UBBC-07 Enhances Circulating Essential Amino Acids in Young Adults: A Double-Blind, Randomized, Controlled Trial. Cureus 2025; 17:e81119. [PMID: 40276429 PMCID: PMC12020262 DOI: 10.7759/cureus.81119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Background and aim Probiotics have been linked to improved gastrointestinal health and essential nutrient absorption. This study aimed to assess the impact of Bacillus clausii(Shouchella clausii) UBBC-07 plus whey protein supplementation on the bioavailability of circulating essential amino acids (EAAs) in physically active young adults. Methods In this double-blind, randomized, controlled trial, 70 physically active male participants (21.46±3.19 years) were instructed to ingest either a probiotic supplement containing two billion colony-forming unit (CFU) Bacillus clausii UBBC-07 + 20 g of whey protein or a control supplement containing placebo + 20 g of whey protein once daily for 60 days. All the participants followed a supervised exercise protocol. The circulating amino acid levels were determined using a high-performance liquid chromatography with fluorescence detection (HPLC-FLD) assay and compared using the student's t-test and a repeated measures analysis of variance (ANOVA). Results After 60 days, a significant improvement in the probiotic group was observed compared to the control group in terms of total levels of circulating EAAs (mean change: 258 pmol/μl, 95% CI: 161.5-354.4 vs. 76.4 pmol/μl, 95% CI: 16.5-136.4; p=0.002) and branched-chain amino acids or BCAAs (mean change: 144.2 pmol/μl, 95% CI: 89-199.3 vs. 37.5 pmol/μl, 95% CI: 7.3-67.8; p=0.001) as well as levels of isoleucine (p=0.003), leucine (p>0.001), and valine (p=0.001). Total plasma free amino acids (PFAAs) were also increased in the probiotic group (p<0.001). The improvement in the one-repetition maximum (RM) leg press was higher in the probiotic group as compared to the control group (mean change: 20.46 kg, 95% CI: 14.73-26.19 vs. 14.09 kg, 95% CI: 8.44, 19.73; p=0.045). A trend towards improvement in deadlift and vertical jump was also observed in the former group. No probiotic-mediated gastrointestinal upsets and respiratory symptoms or any other adverse events were observed. Conclusion A significant improvement in circulating EAA levels in the probiotic group suggests an enhancement of protein absorption with Bacillus clausii UBBC-07 supplementation. The effect of BCAAs, which enhance muscle strength, is evident in the significant improvement in leg press and a trend towards improvement in deadlift and vertical jump in the probiotic group. This has positive implications for individuals involved in sports activities.
Collapse
Affiliation(s)
- Mohamad Tarik
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Nidhi Bhatia
- Department of Orthopaedics, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Atanu Roy
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | | | - Dinu S Chandran
- Department of Physiology, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | - Mani Kalaivani
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, New Delhi, IND
| | | | - Ratna S Madempudi
- Centre for Research and Development, Unique Biotech Limited, Hyderabad, IND
| |
Collapse
|
12
|
Alshaikh BN, Ting J, Lee S, Lemyre B, Wong J, Afifi J, Beltempo M, Shah PS. Effectiveness and Risks of Probiotics in Preterm Infants. Pediatrics 2025; 155:e2024069102. [PMID: 39933567 DOI: 10.1542/peds.2024-069102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/21/2024] [Indexed: 02/13/2025] Open
Abstract
OBJECTIVE To evaluate the effectiveness and risks of probiotics among infants born before 34 weeks' gestation and with a birth weight less than 1000 g. METHODS A population-based retrospective cohort study of infants born before 34 weeks' gestation and admitted to 33 Canadian Neonatal Network (CNN) units between January 1, 2016, and December 31, 2022. We excluded infants who were moribund on admission, died within the first 2 days, were admitted to CNN sites more than 2 days after birth, had major congenital anomalies, or never received enteral feeds. Logistic regression, propensity score-matched, and inverse probability of treatment weighting analyses were applied. RESULTS Among 32 667 eligible infants born before 34 weeks' gestation, 18 793 (57.5%) (median [IQR] gestational age, 29 [27-31] weeks) received probiotics, and 13 874 (42.5%) (median [IQR] gestational age, 31 [29-33] weeks) did not receive probiotics. In these infants, probiotics were associated with decreased mortality rates (adjusted odds ratio [aOR], 0.62; 98.3% CI, 0.53-0.73) but not decreased rates of necrotizing enterocolitis (NEC) (aOR, 0.92; 98.3% CI, 0.78-1.09) or late-onset sepsis (aOR, 0.90; 98.3% CI, 0.80-1.01). In 7401 infants with a birth weight less than 1000 g, probiotics were associated with decreased mortality rates (aOR, 0.58; 98.3% CI, 0.47-0.71) but not decreased NEC (aOR, 0.90; 98.3% CI. 0.71-1.13) or late-onset sepsis rates (aOR, 1.01; 98.3% CI, 0.86-1.18). Probiotic sepsis occurred in 27 (1.4/1000) infants born before 34 weeks' gestation and 20 (4/1000) infants with a birth weight less than 1000 g. Three infants with probiotic sepsis died, with probiotic sepsis deemed a possible cause in 2 cases. CONCLUSION Probiotics used in Canadian neonatal units were associated with decreased mortality in infants born before 34 weeks' gestation and with a birth weight less than 1000 g with limited effects on NEC and late-onset sepsis. Probiotic sepsis was rare.
Collapse
Affiliation(s)
- Belal N Alshaikh
- Neonatal Gastroenterology and Nutrition Program, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joseph Ting
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Seungwoo Lee
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Brigitte Lemyre
- Department of Pediatrics, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan Wong
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jehier Afifi
- Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marc Beltempo
- Division of Neonatology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Prakesh S Shah
- Department of Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Sun T, Song B, Li B. Gut microbiota and atrial cardiomyopathy. Front Cardiovasc Med 2025; 12:1541278. [PMID: 39968343 PMCID: PMC11832500 DOI: 10.3389/fcvm.2025.1541278] [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: 12/10/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Atrial cardiomyopathy is a multifaceted heart disease characterized by structural and functional abnormalities of the atria and is closely associated with atrial fibrillation and its complications. Its etiology involves a number of factors, including genetic, infectious, immunologic, and metabolic factors. Recent research has highlighted the critical role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, and this is consistent with the gut-heart axis having major implications for cardiac health. The aim of this work is to bridge the knowledge gap regarding the interactions between the gut microbiota and atrial cardiomyopathy, with a particular focus on elucidating the mechanisms by which gut dysbiosis may induce atrial remodeling and dysfunction. This article provides an overview of the role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, including changes in the composition of the gut microbiota and the effects of its metabolites. We also discuss how diet and exercise affect atrial cardiomyopathy by influencing the gut microbiota, as well as possible future therapeutic approaches targeting the gut-heart axis. A healthy gut microbiota can prevent disease, but ecological dysbiosis can lead to a variety of symptoms, including the induction of heart disease. We focus on the pathophysiological aspects of atrial cardiomyopathy, the impact of gut microbiota dysbiosis on atrial structure and function, and therapeutic strategies exploring modulation of the microbiota for the treatment of atrial cardiomyopathy. Finally, we discuss the role of gut microbiota in the treatment of atrial cardiomyopathy, including fecal microbiota transplantation and oral probiotics or prebiotics. Our study highlights the importance of gut microbiota homeostasis for cardiovascular health and suggests that targeted interventions on the gut microbiota may pave the way for innovative preventive and therapeutic strategies targeting atrial cardiomyopathy.
Collapse
Affiliation(s)
- Tingting Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Beibei Song
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Bo Li
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
14
|
D’Amico V, Cavaliere M, Ivone M, Lacassia C, Celano G, Vacca M, la Forgia FM, Fontana S, De Angelis M, Denora N, Lopedota AA. Microencapsulation of Probiotics for Enhanced Stability and Health Benefits in Dairy Functional Foods: A Focus on Pasta Filata Cheese. Pharmaceutics 2025; 17:185. [PMID: 40006552 PMCID: PMC11859715 DOI: 10.3390/pharmaceutics17020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Probiotics provide significant health benefits, but their viability is often compromised during production, storage, and passage through the gastrointestinal tract. These challenges hinder their effective incorporation into functional applications, particularly in dairy functional foods, in which factors such as acidity, oxygen exposure, and storage conditions negatively impact cell survival. The focus was on functional dairy foods, particularly on pasta filata cheeses. Indeed, the use of probiotics in pasta filata cheeses presents significant challenges due to the specific manufacturing processes, which encompass the application of high temperatures and other harsh conditions. These factors can adversely affect the viability and availability of probiotic microorganisms. However, microencapsulation has emerged as a promising solution, offering a protective barrier that enhances probiotic stability, improves survival rates, and facilitates targeted release in the gastrointestinal environment. This review examines the pivotal role of microencapsulation in stabilising probiotics for functional applications, emphasising its relevance in high-value food systems. Functional applications, including foods designed to offer essential nutritional benefits and promote host health, play a crucial role in disease prevention and immune system support, reducing the risk of infections and other physiological impairments. Key microencapsulation technologies are analysed, focusing on their benefits, limitations, and challenges related to scalability and industrial implementation. Additionally, this review discusses strategies to optimise formulations, ensure the sensory quality of final products, and explore future opportunities for expanding innovative applications that align with growing consumer demand for health-promoting solutions.
Collapse
Affiliation(s)
- Vita D’Amico
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 4, E. Orabona Street, 70125 Bari, Italy; (V.D.); (M.I.); (C.L.); (N.D.)
| | - Mariasimona Cavaliere
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 165/A, G. Amendola Street, 70126 Bari, Italy; (M.C.); (G.C.); (M.V.); (M.D.A.)
| | - Marianna Ivone
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 4, E. Orabona Street, 70125 Bari, Italy; (V.D.); (M.I.); (C.L.); (N.D.)
| | - Chiara Lacassia
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 4, E. Orabona Street, 70125 Bari, Italy; (V.D.); (M.I.); (C.L.); (N.D.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 165/A, G. Amendola Street, 70126 Bari, Italy; (M.C.); (G.C.); (M.V.); (M.D.A.)
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 165/A, G. Amendola Street, 70126 Bari, Italy; (M.C.); (G.C.); (M.V.); (M.D.A.)
| | - Flavia Maria la Forgia
- Centro Studi e Ricerche “Dr. S. Fontana 1900–1982”, Farmalabor s.r.l., 47, Piano S. Giovanni Street, 76012 Canosa di Puglia, Italy; (F.M.l.F.); (S.F.)
| | - Sergio Fontana
- Centro Studi e Ricerche “Dr. S. Fontana 1900–1982”, Farmalabor s.r.l., 47, Piano S. Giovanni Street, 76012 Canosa di Puglia, Italy; (F.M.l.F.); (S.F.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 165/A, G. Amendola Street, 70126 Bari, Italy; (M.C.); (G.C.); (M.V.); (M.D.A.)
| | - Nunzio Denora
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 4, E. Orabona Street, 70125 Bari, Italy; (V.D.); (M.I.); (C.L.); (N.D.)
| | - Angela Assunta Lopedota
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, 4, E. Orabona Street, 70125 Bari, Italy; (V.D.); (M.I.); (C.L.); (N.D.)
| |
Collapse
|
15
|
Chen A, Gong Y, Wu S, Du Y, Liu Z, Jiang Y, Li J, Miao YB. Navigating a challenging path: precision disease treatment with tailored oral nano-armor-probiotics. J Nanobiotechnology 2025; 23:72. [PMID: 39893419 PMCID: PMC11786591 DOI: 10.1186/s12951-025-03141-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Oral probiotics have significant potential for preventing and treating many diseases. Yet, their efficacy is often hindered by challenges related to survival and colonization within the gastrointestinal tract. Nanoparticles emerge as a transformative solution, offering robust protection and enhancing the stability and bioavailability of these probiotics. This review explores the innovative application of nanoparticle-armored engineered probiotics for precise disease treatment, specifically addressing the physiological barriers associated with oral administration. A comprehensive evaluation of various nano-armor probiotics and encapsulation methods is provided, carefully analyzing their respective merits and limitations, alongside strategies to enhance probiotic survival and achieve targeted delivery and colonization within the gastrointestinal tract. Furthermore, the review explores the potential clinical applications of nano-armored probiotics in precision therapeutics, critically addressing safety and regulatory considerations, and proposing the innovative concept of 'probiotic intestinal colonization with nano armor' for brain-targeted therapies. Ultimately, this review aspires to guide the advancement of nano-armored probiotic therapies, driving progress in precision medicine and paving the way for groundbreaking treatment modalities.
Collapse
Affiliation(s)
- Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Shaoquan Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ye Du
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Jiahong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| |
Collapse
|
16
|
Todor SB, Ichim C. Microbiome Modulation in Pediatric Leukemia: Impact on Graft-Versus-Host Disease and Treatment Outcomes: A Narrative Review. CHILDREN (BASEL, SWITZERLAND) 2025; 12:166. [PMID: 40003268 PMCID: PMC11854176 DOI: 10.3390/children12020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025]
Abstract
The gut microbiome significantly influences the outcomes of pediatric leukemia, particularly in patients undergoing hematopoietic stem cell transplantation (HSCT). Dysbiosis, caused by chemotherapy, antibiotics, and immune system changes, contributes to complications such as graft-versus-host disease (GVHD), gastrointestinal issues, and infections. Various microbiome-related interventions, including prebiotics, probiotics, postbiotics, and fecal microbiota transplantation (FMT), have shown potential in mitigating these complications. Specific microbial signatures have been linked to GVHD risk, and interventions like inulin, Lactobacillus, and SCFAs (short-chain fatty acids), particularly butyrate, may help modulate the immune system and improve outcomes. FMT, while showing promising results in restoring microbial balance and alleviating GVHD, still requires careful monitoring due to potential risks in immunocompromised patients. Despite positive findings, more research is needed to optimize microbiome-based therapies and ensure their safety and efficacy in pediatric leukemia care.
Collapse
Affiliation(s)
- Samuel Bogdan Todor
- Faculty of Medicine, University Lucian Blaga of Sibiu, 550024 Sibiu, Romania;
| | | |
Collapse
|
17
|
Farmakioti I, Stylianopoulou E, Siskos N, Karagianni E, Kandylas D, Vasileiou AR, Fragkiskatou F, Somalou P, Tsaroucha A, Ypsilantis P, Panas P, Kourkoutas Y, Skavdis G, Grigoriou ME. Enhancing Gut Microbiome and Metabolic Health in Mice Through Administration of Presumptive Probiotic Strain Lactiplantibacillus pentosus PE11. Nutrients 2025; 17:442. [PMID: 39940300 PMCID: PMC11820638 DOI: 10.3390/nu17030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025] Open
Abstract
Background: Over the past decade, probiotics have gained increasing recognition for their health benefits to the host. While most research has focused on the therapeutic effects of probiotics in the treatment of various diseases, recent years have seen a shift towards exploring their role in enhancing and supporting overall health. Methods: In this work, we have studied the effects of a novel potential probiotic strain, Lactiplantibacillus pentosus PE11, in healthy mice following a six-week dietary intervention. The assessment included monitoring the general health of the animals, biochemical analyses, profiling of the gut and fecal microbial communities, and gene expression analysis. Results: Our results showed that the administration of Lactiplantibacillus pentosus PE11 led to changes in the composition of the fecal microbiome, specifically an increase in the Firmicutes/Bacteroidetes ratio and in the relative abundance of the Lachnospiraceae, Ruminococcaceae, and Rikenellaceae families. Reduced Tnf expression and elevated Zo1 expression were also observed in the cecum, pointing to anti-inflammatory properties and improved intestinal barrier integrity. Additionally, a significant reduction in triglycerides and alanine aminotransferase levels-within physiological ranges-was observed, along with a trend toward decreased total cholesterol levels. Conclusions: These findings suggest that in healthy mice, Lactiplantibacillus pentosus PE11 has the potential to positively influence gut microbiome structure and metabolism, thereby supporting improved overall health.
Collapse
Affiliation(s)
- Ioanna Farmakioti
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Electra Stylianopoulou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Nikistratos Siskos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Evangelia Karagianni
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Dionysios Kandylas
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Andreas Rafail Vasileiou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Fragkiski Fragkiskatou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Paraskevi Somalou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Alexandra Tsaroucha
- Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.T.); (P.Y.)
| | - Petros Ypsilantis
- Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.T.); (P.Y.)
| | | | - Yiannis Kourkoutas
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - George Skavdis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Maria E. Grigoriou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| |
Collapse
|
18
|
Naeem M, Bourassa D. Probiotics in Poultry: Unlocking Productivity Through Microbiome Modulation and Gut Health. Microorganisms 2025; 13:257. [PMID: 40005624 PMCID: PMC11857632 DOI: 10.3390/microorganisms13020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This review explores the role of probiotics in improving productivity and gut health in poultry through microbiome modulation, particularly during early life. Gut health is pivotal to poultry performance, influencing nutrient absorption, immune function, and disease resistance. Early-life interventions target the microbiome to shape long-term health and productivity. Probiotics, live microorganisms providing health benefits, improve gut health through the competitive exclusion of pathogens, immune modulation, antimicrobial compound production, and enhancing gut barrier integrity. Applying probiotics improves growth performance, feed conversion efficiency, body weight gain, and carcass quality by promoting lean muscle growth and reducing fat deposition. For laying hens, probiotics enhance egg production and quality. These benefits are linked to better nutrient utilization, a well-balanced microbiome, and reduced gastrointestinal disorders. However, the efficacy of probiotics depends on strain specificity, dosage, and administration methods. Factors like environmental conditions, storage stability, and interactions with other feed additives also influence their effectiveness. Despite these challenges, advancements in microbiome research and probiotic technologies, such as precision probiotics and synbiotics, provide promising solutions. Future research should focus on optimizing formulations, understanding host-microbiome interactions, and leveraging new technologies for targeted microbiome management.
Collapse
Affiliation(s)
- Muhammad Naeem
- Department of Poultry Science, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
19
|
Seo S, Jeong DW, Sul S, Lee JH. Specificity of Amino Acid Profiles Produced in Soybean Fermentations by Three Bacillus spp. J Microbiol Biotechnol 2024; 35:e2411038. [PMID: 39848677 PMCID: PMC11813359 DOI: 10.4014/jmb.2411.11038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 01/25/2025]
Abstract
We compared the salt tolerance and proteolytic activity of 120 strains of each of Bacillus subtilis, Bacillus velezensis, and Bacillus licheniformis. Most B. subtilis strains exhibited growth in 12% (w/v) NaCl and showed proteolytic activity in 10% or 11% NaCl. The majority of B. velezensis strains grew in 14% NaCl and showed proteolytic activity in 12% or 13% NaCl. Most B. licheniformis strains grew in 14% NaCl and exhibited proteolytic activity in 5%-7% NaCl. We selected nine representative strains of each species based on their proteolytic activities and analyzed the free amino acid (FAA) profiles produced by culture of the bacteria on soybean. Statistical analyses of the 22 FAAs quantified in the cultures revealed clustering of FAA production profiles at the species level. The FAA production profiles of B. subtilis and B. velezensis were similar, and both differed from that of B. licheniformis. These trends persisted in cultures containing 7% NaCl. These results suggest that FAA production profiles are characteristic of each Bacillus species. Specifically, in soybean cultures compared with uninoculated soybeans, B. subtilis increased the amounts of leucine and phenylalanine; B. velezensis increased the amounts of leucine, phenylalanine, and tyrosine; and B. licheniformis increased the amounts of alanine, glutamic acid, tyrosine, and ornithine, and dramatically decreased the amount of arginine. The proteolytic activity of B. velezensis strains correlated with the quantity of FAAs in their soybean cultures. Considering its salt tolerance and proteolytic activity, B. velezensis showed high potential for contributing to the ripening of high-salt fermented soybean foods. Our results regarding the specific production of amino acids at the species level and correlations between proteolytic activities and produced amino acid quantities will facilitate the determination and selection of target strains for functional Bacillus-fermented foods.
Collapse
Affiliation(s)
- Sumin Seo
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| | - Do-Won Jeong
- Department of Food and Nutrition, Dongduk Women’s University, Seoul 02748, Republic of Korea
| | - Sooyoung Sul
- Division of Sports Science, Kyonggi University, Suwon 16227, Republic of Korea
| | - Jong-Hoon Lee
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
20
|
Kovačec E, Kraigher B, Podnar E, Lories B, Steenackers H, Mandic‐Mulec I. Bacillus subtilis Intraspecies Interactions Shape Probiotic Activity Against Salmonella Typhimurium. Microb Biotechnol 2024; 17:e70065. [PMID: 39718437 PMCID: PMC11667775 DOI: 10.1111/1751-7915.70065] [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: 05/17/2024] [Revised: 09/23/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
Commercial probiotics are often formulated as multi-strain cocktails, but the effects of social interactions, particularly between strains of a species, are often neglected, despite their potential to contribute to higher-order interactions where these interactions could affect those with a third party. In this study, we investigated the probiotic potential of a collection of Bacillus subtilis strains against Salmonella Typhimurium in single-strain and mixed cultures. The results indicate a promising probiotic potential of B. subtilis as 38 out of 39 strains significantly inhibited the growth of S. Typhimurium. Next, we tested the effect of mixing B. subtilis strains that differ in their inhibitory potency against S. Typhimurium. The results show that strong inhibition by one strain can be significantly reduced by mixing with a less effective strain. Moreover, mixing similarly effective strains mostly resulted in a decreased growth inhibition of the pathogen. Additionally, we found a group of highly aggressive strains, which completely eliminated other B. subtilis strains in the two-strain mixtures. Overall, this work shows that intraspecies interactions between B. subtilis strains can significantly alter the probiotic effect against S. Typhimurium, which is of great importance for future research on the development of multi-strain probiotics.
Collapse
Affiliation(s)
- Eva Kovačec
- Department of Microbiology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Barbara Kraigher
- Department of Microbiology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Eli Podnar
- Department of Microbiology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| | - Bram Lories
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG)KU LeuvenLeuvenBelgium
| | - Hans Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG)KU LeuvenLeuvenBelgium
| | - Ines Mandic‐Mulec
- Department of Microbiology, Biotechnical FacultyUniversity of LjubljanaLjubljanaSlovenia
| |
Collapse
|
21
|
Almabruk BA, Bajafar AA, Mohamed AN, Al-Zahrani SA, Albishi NM, Aljarwan R, Aljaser RA, Alghamdi LI, Almutairi TS, Alsolami AS, Alghamdi JK. Efficacy of Probiotics in the Management of Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e75954. [PMID: 39830577 PMCID: PMC11741147 DOI: 10.7759/cureus.75954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Irritable bowel syndrome (IBS) significantly impacts quality of life. Probiotics offer relief by modulating gut microbiota, but variability in outcomes necessitates a systematic evaluation of their efficacy. This study aims to evaluate the efficacy of probiotics in improving symptoms of IBS through a systematic review and meta-analysis. A comprehensive search of PubMed and Google Scholar identified studies published between 2014 and 2018. Inclusion criteria focused on randomized controlled trials evaluating probiotics in adult IBS patients diagnosed using standardized criteria. Statistical analysis utilized random effects models to account for heterogeneity, with subgroup analysis performed for IBS subtypes. This review included 23 studies involving 3,288 participants. Probiotics significantly reduced abdominal pain (mean difference = -1.66, 95% CI = -2.39 to -0.93, p < 0.0001) and bloating (mean difference = -2.13, 95% CI = -3.96 to -0.30, p = 0.0224). Improvement in stool habits was significant (mean difference = -1.52, 95% CI = -2.15 to -0.88, p < 0.0001), particularly in diarrhea-predominant IBS. Quality of life improved significantly, with a mean increase of 8.77 points (95% CI = 0.91 to 16.64, p = 0.028). Adverse events were mild and infrequent. However, heterogeneity was high (I² > 90%), reflecting variability in study protocols. Probiotics are effective in reducing IBS symptoms and improving quality of life, mainly in diarrhea-predominant IBS. More research should be conducted that focuses on standardized, long-term trials to refine treatment strategies.
Collapse
Affiliation(s)
| | - Ali A Bajafar
- General Practice, King Abdullah Medical Complex, Jeddah, SAU
| | - Ahmed N Mohamed
- Critical Care Medicine, SEHA Salma Rehabilitation Hospital, Abu Dhabi, ARE
| | - Saleh A Al-Zahrani
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Naif M Albishi
- Nursing, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | - Rafeef Aljarwan
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, SAU
| | | | | | | | - Almas S Alsolami
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
| | - Joud K Alghamdi
- Faculty of Medicine, King Abdulaziz University Hospital, Jeddah, SAU
| |
Collapse
|
22
|
Amar Y, Grube J, Köberle M, Schaubeck M, Biedermann T, Volz T. Bifidobacterium breve DSM 32583 and Limosilactobacillus fermentum CECT5716 postbiotics attenuate S. aureus and IL-33-induced Th2 responses. Microbiol Res 2024; 289:127913. [PMID: 39316930 DOI: 10.1016/j.micres.2024.127913] [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: 07/12/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 09/26/2024]
Abstract
Over the past decades, the prevalence of allergic diseases noticeably increased in industrialized countries. The Th2 immune response plays a central role in these pathologies and its modulation using pro-/postbiotics constitutes a promising approach to prevent or alleviate disease symptoms. The aim of this in vitro study, was to investigate the ability of human milk-derived Bifidobacterium breve DSM 32583 (Bb) and Limosilactobacillus fermentum CECT5716 (Lf), to modulate the Th2 induced responses. To this end, Th2 cells were generated by co-culturing of human naïve Th cells with monocyte-derived dendritic cells (moDCs) either stimulated with Staphylococcus aureus or IL-33. The immunomodulatory effects of pro-/postbiotic preparations of Bb and Lf on moDCs and Th2 cells were evaluated in terms of maturation markers expression and cytokines production. Remarkably, the tested strains induced the anti-inflammatory cytokine IL-10 in moDCs, in a strain-, dose- and viability-dependent manner with no significant upregulation of IL-12p70 nor CD83, CD86 or HLA-DR. Interestingly, Bb and Lf postbiotics were able to dampen the Th2/Th1 response induced upon S. aureus- or IL-33 stimulation. They were also able to synergistically induce IL-10 in moDCs and T cells, upon co-stimulation with LPS. Finally, we observed that live probiotics triggered a mild Th1 response that was attenuated in the presence of galacto-oligosaccharides. Altogether, Bb and Lf pro-/postbiotics exhibited remarkable immune regulatory effects on both moDCs and Th2 cells. Therefore, further in vivo studies should be considered to validate these findings and assess their ability to prevent allergy or alleviate its symptoms in affected patients.
Collapse
Affiliation(s)
- Yacine Amar
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany.
| | - Jana Grube
- HiPP GmbH & Co. Vertrieb KG, Pfaffenhofen (Ilm) 85276, Germany
| | - Martin Köberle
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | | | - Tilo Biedermann
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| | - Thomas Volz
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich 80802, Germany
| |
Collapse
|
23
|
Lungaro L, Malfa P, Manza F, Costanzini A, Valentini G, Squarzanti DF, Viciani E, Velichevskaya A, Castagnetti A, Barbalinardo M, Gentili D, Cariani A, Ghisellini S, Caputo F, De Giorgio R, Caio G. Clinical Efficacy of Probiotics for Allergic Rhinitis: Results of an Exploratory Randomized Controlled Trial. Nutrients 2024; 16:4173. [PMID: 39683566 PMCID: PMC11644003 DOI: 10.3390/nu16234173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Allergic Rhinitis (AR) is an atopic disease affecting the upper airways of predisposed subjects exposed to aeroallergens. This study evaluates the effects of a mix of specific probiotics (L. acidophilus PBS066, L. rhamnosus LRH020, B. breve BB077, and B. longum subsp. longum BLG240) on symptoms and fecal microbiota modulation in subjects with AR. Methods: Probiotic effects were evaluated at the beginning (T0), at four and eight weeks of treatment (T1 and T2, respectively), and after four weeks of follow-up from the end of treatment (T3) (n = 19) compared to the placebo group (n = 22). AR symptoms and quality of life were evaluated by the mini rhinitis quality of life questionnaire (MiniRQLQ) at each time point. Allergic immune response and fecal microbiota compositions were assessed at T0, T2, and T3. The study was registered on Clinical-Trial.gov (NCT05344352). Results: The probiotic group showed significant improvement in the MiniRQLQ score at T1, T2, and T3 vs. T0 (p < 0.01, p < 0.05, p < 0.01, respectively). At T2, the probiotic group showed an increase in Dorea, which can be negatively associated with allergic diseases, and Fusicatenibacter, an intestinal bacterial genus with anti-inflammatory properties (p-value FDR-corrected = 0.0074 and 0.013, respectively). Conversely, at T3 the placebo group showed an increase in Bacteroides and Ruminococcaceae unassigned, (p-value FDR-corrected = 0.033 and 0.023, respectively) which can be associated with allergies, while the probiotic group showed a significative increase in the Prevotella/Bacteroides ratio (p-value FDR-corrected = 0.023). Conclusions: This probiotic formulation improves symptoms and quality of life in subjects with AR, promoting a shift towards anti-inflammatory and anti-allergic bacterial species in the intestinal microbiota.
Collapse
Affiliation(s)
- Lisa Lungaro
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | | | - Francesca Manza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Guido Valentini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | | | - Elisa Viciani
- Wellmicro Srl, 40138 Bologna, Italy; (E.V.); (A.V.); (A.C.)
| | | | | | - Marianna Barbalinardo
- Institute for the Study of Nanostructured Materials (CNR-ISMN), National Research Council, 40129 Bologna, Italy; (M.B.); (D.G.)
| | - Denis Gentili
- Institute for the Study of Nanostructured Materials (CNR-ISMN), National Research Council, 40129 Bologna, Italy; (M.B.); (D.G.)
| | - Alessio Cariani
- Analysis Laboratory, St. Anna Hospital, 44121 Ferrara, Italy; (A.C.); (S.G.)
| | - Sara Ghisellini
- Analysis Laboratory, St. Anna Hospital, 44121 Ferrara, Italy; (A.C.); (S.G.)
| | - Fabio Caputo
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
- Complex Operational Unit of Internal Medicine, S.S. Annunziata Hospital, Cento, 44121 Ferrara, Italy
| | - Giacomo Caio
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.M.); (A.C.); (G.V.); (F.C.); (R.D.G.)
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital-Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
24
|
Li Z, Li M, Fang X, Yu D, Hu X. Dietary Lactobacillus johnsonii-derived extracellular vesicles ameliorate acute colitis by regulating gut microbiota and maintaining intestinal barrier homeostasis. Food Funct 2024; 15:11757-11779. [PMID: 39545264 DOI: 10.1039/d4fo04194a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disease with intricate pathogenesis, and clinical treatment is still not ideal. The imbalance of gut microbiota is associated with IBD progression. Various probiotics have been used as functional foods for the prevention and treatment of IBD, but the specific mechanism is still not fully understood. Lactobacillus johnsonii (L. johnsonii) is a potential anti-inflammatory bacterium, and compared to other probiotic Lactobacillus species, its colonization in the gut of colitis patients is significantly reduced. In this study, we first found that dietary L. johnsonii exerts strong anti-inflammatory and antioxidant effects in colitis mice, and this beneficial effect is directly related to its derived extracellular vesicles (LJ-EVs). Further experimental results indicate that LJ-EVs effectively prevented colitis symptoms and modulated gut microbiota and metabolic pathways. Meanwhile, we have studied for the first time the protective effect of LJ-EVs on the intestinal barrier from the perspective of reducing oxidative stress. We found that LJ-EVs can be directly taken up by intestinal epithelial cells and activate the Nrf2/HO-1 antioxidant signaling pathway, reducing endotoxin damage to cells and maintaining intestinal barrier homeostasis, which cascades to alleviate intestinal inflammation response. This study reveals the mechanism of L. johnsonii in treating colitis and provides a new approach for the development of oral LJ-EVs for the treatment of colitis.
Collapse
Affiliation(s)
- Zhiguo Li
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, P. R. China.
| | - Mengyu Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, P. R. China
| | - Xin Hu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, P. R. China.
| |
Collapse
|
25
|
Qian X, Li Q, Zhu H, Chen Y, Lin G, Zhang H, Chen W, Wang G, Tian P. Bifidobacteria with indole-3-lactic acid-producing capacity exhibit psychobiotic potential via reducing neuroinflammation. Cell Rep Med 2024; 5:101798. [PMID: 39471819 PMCID: PMC11604549 DOI: 10.1016/j.xcrm.2024.101798] [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: 02/28/2024] [Revised: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 11/01/2024]
Abstract
The escalating global prevalence of depression demands effective therapeutic strategies, with psychobiotics emerging as a promising solution. However, the molecular mechanisms governing the neurobehavioral impact of psychobiotics remain elusive. This study reveals a significant reduction in hippocampal indole-3-lactic acid (ILA) levels in depressed mice, which is ameliorated by the psychobiotic Bifidobacterium breve. In both human subjects and mice, the ILA increase in the circulatory system results from bifidobacteria supplementation. Further investigation identifies the key aromatic lactate dehydrogenase (Aldh) gene and pathway in bifidobacteria responsible for ILA production. Importantly, the antidepressant effects are nullified in the Aldh mutants compared to the wild-type strain. At the bifidobacteria species level, those with Aldh exhibit heightened antidepressant effects. Finally, this study emphasizes the antidepressant efficacy of psychobiotic-derived ILA, potentially mediated by aryl hydrocarbon receptor (AhR) signaling activation to alleviate neuroinflammation. This study unveils the molecular and genetic foundations of psychobiotics' antidepressant effects, offering insights for microbial therapies targeting mood disorders.
Collapse
Affiliation(s)
- Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qing Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Guopeng Lin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
26
|
Wu Y, Gao D, Pan Y, Dong Y, Bai Z, Gu S. Modulation of Serum Metabolic Profiles by Bifidobacterium breve BBr60 in Obesity: A Randomized Controlled Trial. Foods 2024; 13:3655. [PMID: 39594072 PMCID: PMC11594036 DOI: 10.3390/foods13223655] [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: 10/20/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity, a prevalent metabolic disorder in youth, leads to complications and economic strain. Gut dysbiosis significantly contributes to obesity and metabolic issues. Bifidobacterium breve, a probiotic strain, may help regulate gut dysbiosis and benefit obese individuals. However, more research is needed on its effect on serum metabolism. A total of 75 overweight or obese young adults (aged 19-45) participated in the current study, and were randomly divided into probiotic and placebo groups using a random number table. Both groups received dietary guidance and underwent twelve weeks of intervention with either oral Bifidobacterium breve BBr60 (BBr60) or a placebo. After the intervention, collection and analysis of fasting serum samples were conducted using mass spectrometry coupled with liquid chromatography. Analyses of associations were conducted in order to determine the correlations between key serum metabolites and clinical obesity indicators, aiming to understand the influence of BBr60. Due to 10 participants dropping out for personal reasons, the study included 32 and 33 participants in the placebo and the BBr60 groups, respectively. The BBr60 intervention significantly regulated 134 serum metabolites and influenced crucial metabolic pathways in obesity management (p < 0.05), including ascorbate and aldarate metabolism for oxidative stress reduction, cholesterol metabolism for lipid regulation, parathyroid hormone synthesis, secretion and action for the endocrine system, oxidative phosphorylation for enhanced energy efficiency, and glycolysis/gluconeogenesis for glucose metabolism. Analysis showed a positive relationship between fasting blood glucose (FBG), aspartate aminotransferase (AST), total protein (TP), and the content of 5-Methyl DL-glutamate (p < 0.05). Similarly, body mass index (BMI), weight, and body fat percentage (BFP) were positively linked to serum metabolites (1-Hydroxycyclohexyl) acetic acid, and 5-Oxooctanoic acid (p < 0.05). Significant associations of AST levels with key serum metabolites in cholesterol metabolism pathways further suggest BBr60's potential to improve liver function and overall metabolic health in overweight or obese individuals. These findings support BBr60's effectiveness in modulating serum metabolic profiles and suggest it may improve liver function and BMI in overweight or obese individuals by regulating key serum metabolites.
Collapse
Affiliation(s)
- Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Y.W.); (D.G.); (Y.P.)
- Henan Engineering Research Center of Food Microbiology, Luoyang 471000, China
| | - Dejiao Gao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Y.W.); (D.G.); (Y.P.)
| | - Yujia Pan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Y.W.); (D.G.); (Y.P.)
| | - Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Zhouya Bai
- Henan Engineering Research Center of Food Material, Henan University of Science and Technology, Luoyang 471023, China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Y.W.); (D.G.); (Y.P.)
- Henan Engineering Research Center of Food Microbiology, Luoyang 471000, China
| |
Collapse
|
27
|
Martinez Guevara D, Vidal Cañas S, Palacios I, Gómez A, Estrada M, Gallego J, Liscano Y. Effectiveness of Probiotics, Prebiotics, and Synbiotics in Managing Insulin Resistance and Hormonal Imbalance in Women with Polycystic Ovary Syndrome (PCOS): A Systematic Review of Randomized Clinical Trials. Nutrients 2024; 16:3916. [PMID: 39599701 PMCID: PMC11597640 DOI: 10.3390/nu16223916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent research suggests that probiotics and synbiotics may improve these parameters by modulating the gut microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL, Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed with PCOS according to the Rotterdam criteria were included. Data on participants, interventions, and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR, fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal. Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous populations, and short intervention durations limit the generalization of the findings. Larger, longer, multicenter trials with diverse populations and standardized methodologies are needed to confirm the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve clinical management and quality of life for affected women, but additional evidence is required to support widespread use.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 5183000, Colombia; (D.M.G.); (S.V.C.); (I.P.); (A.G.); (M.E.); (J.G.)
| |
Collapse
|
28
|
Whelan K, Alexander M, Gaiani C, Lunken G, Holmes A, Staudacher HM, Theis S, Marco ML. Design and reporting of prebiotic and probiotic clinical trials in the context of diet and the gut microbiome. Nat Microbiol 2024; 9:2785-2794. [PMID: 39478082 DOI: 10.1038/s41564-024-01831-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 09/12/2024] [Indexed: 11/02/2024]
Abstract
Diet is a major determinant of the gastrointestinal microbiome composition and function, yet our understanding of how it impacts the efficacy of prebiotics and probiotics is limited. Here we examine current evidence of dietary influence on prebiotic and probiotic efficacy in human studies, including potential mechanisms. We propose that habitual diet be included as a variable in prebiotic and probiotic intervention studies. This recommendation is based on the potential mechanisms via which diet can affect study outcomes, either directly or through the gut microbiome. We consider the challenges and opportunities of dietary assessment in this context. Lastly, we provide recommendations for the design, conduct and reporting of human clinical trials of prebiotics and probiotics (and other biotic interventions) to account for any effect of diet and nutrition.
Collapse
Affiliation(s)
- Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, UK.
| | - Margaret Alexander
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Claire Gaiani
- Laboratoire d'Ingenierie des Biomolecules, Université de Lorraine, Nancy, France
- Institut Universitaire de France, Paris, France
| | - Genelle Lunken
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Andrew Holmes
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Heidi M Staudacher
- Food and Mood Centre, IMPACT Institute, Deakin University, Melbourne, Victoria, Australia
| | | | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
29
|
Soleimanpour S, Abavisani M, Khoshrou A, Sahebkar A. Probiotics for autism spectrum disorder: An updated systematic review and meta-analysis of effects on symptoms. J Psychiatr Res 2024; 179:92-104. [PMID: 39265200 DOI: 10.1016/j.jpsychires.2024.09.009] [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: 03/26/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Recent researches highlighted the significant role of the gut-brain axis and gut microbiota in autism spectrum disorder (ASD), a neurobehavioral developmental disorder characterized by a variety of neuropsychiatric and gastrointestinal symptoms, suggesting that alterations in the gut microbiota may correlate with the severity of ASD symptoms. Therefore, this study was designed to conduct a comprehensive systematic review and meta-analysis of the effectiveness of probiotic interventions in ameliorating behavioral symptoms in individuals with ASD. METHODS This study adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. A comprehensive literature search was performed across multiple databases including the Cochrane Library, PubMed, Web of Science, and Google Scholar up until June 2024. Inclusion criteria encompassed published randomized clinical trials (RCTs), focusing on probiotic interventions and evaluating outcomes related to ASD behavior symptoms. The study utilized Cochrane's Risk of Bias 2 for bias assessment and applied random effect models with inverse variance method for statistical analysis, also addressing publication bias and conducting subgroup analyses through Begg's and Egger's tests to explore the effects of various factors on the outcomes. RESULTS Our meta-analysis, which looked at eight studies with a total of 318 samples from ASD patients aged 1.5-20 years, showed that the probiotic intervention group had significantly better behavioral symptoms compared to the control group. This was shown by a pooled standardized mean difference (SMD) of -0.38 (95% CI: 0.58 to -0.18, p < 0.01). Subgroup analyses revealed significant findings across a variety of factors: studies conducted in the European region showed a notable improvement with an SMD of -0.44 (95%CI: 0.72 to -0.15); interventions lasting longer than three months exhibited a significant improvement with an SMD of -0.43 (95%CI: 0.65 to -0.21); and studies focusing on both participants under and greater than 10 years found significant benefits with an SMDs of -0.37 and -0.40, respectively (95%CI: 0.65 to -0.09, and 95%CI: 0.69 to -0.11, respectively). Moreover, both multi-strain probiotics and single-strain interventions showed an overall significant improvement with a SMD of -0.53 (95%CI: 0.85 to -0.22) and -0.28 (95%CI: 0.54 to -0.02), respectively. Also, the analysis confirmed the low likelihood of publication bias and the robustness of these findings. CONCLUSION Our study highlighted the significant improvement in ASD behavioral symptoms through probiotic supplementation. The need for personalized treatment approaches and further research to confirm efficacy and safety of probiotics in ASD management is emphasized.
Collapse
Affiliation(s)
- Saman Soleimanpour
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Antimicrobial Resistance Research Center, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abavisani
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Khoshrou
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Rao K, Garey KW. From Chaos to Clarity? The Quest for Effective Probiotics in Antibiotic-Associated Diarrhea. Open Forum Infect Dis 2024; 11:ofae616. [PMID: 39529940 PMCID: PMC11551611 DOI: 10.1093/ofid/ofae616] [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: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Antibiotic-associated diarrhea (AAD) frequently complicates treatment of infections. A recent randomized, double-blind, placebo-controlled trial tested a proprietary probiotic mixture and found that it reduced the incidence of AAD by 16%. This is encouraging for patients, but future progress on probiotics for AAD and other conditions depends on transparency around strain selection, probiotic design guided by preclinical mechanistic studies, and rigorously conducted human studies.
Collapse
Affiliation(s)
- Krishna Rao
- University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kevin W Garey
- College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
31
|
Li IC, Lee YL, Li TJ, Tsai YS, Chen YL, Chen CC. Whole-Genome Sequencing of Three Lactiplantibacillus plantarum Strains Reveals Potential Metabolites for Boosting Host Immunity Safely. J Microbiol Biotechnol 2024; 34:2079-2090. [PMID: 39263794 PMCID: PMC11540610 DOI: 10.4014/jmb.2402.02013] [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: 02/08/2024] [Revised: 07/03/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024]
Abstract
In response to the growing demand for immune-related products, this study evaluated the safety and immune-modulating potential of three newly discovered Lactiplantibacillus plantarum strains (GKM3, GKK1, and GKD7) through toxicity tests and whole-genome sequencing. Safety evaluations, including the analysis of antimicrobial resistance genes, virulence factors, plasmids, and prophages, classified these strains as safe for human consumption. Acute oral toxicity tests further supported their safety. To evaluate their immune-modulating potential, dendritic cells were exposed to these strains, and the secretion of key cytokines (IFN-β and IL-12) was measured. Among the strains, GKK1 exhibited the highest enhancement of IFN-β and IL-12 production, suggesting its potential as an immune-stimulating probiotic. Bioinformatics analysis revealed potential metabolic pathways and secondary metabolites, including predicted bacteriocins, associated with immune modulation. The presence of a nitrate reductase region in the GKK1 strain indicated its ability to produce nitric oxide, a critical molecule involved in immune regulation and host defense. The presence of glucorhamnan-related gene clusters in GKK1 also suggested immune-enhancing effects. Nitrate reductase expression was confirmed using qPCR, with the highest levels detected in GKK1. Moreover, this study is the first to show an anti-inflammatory effect of plantaricin A, linked to its presence in strain GKM3 and its potential therapeutic applications due to sequence similarity to known anti-inflammatory peptides. Overall, these three L. plantarum strains demonstrated a safe profile and GKK1 showed potential as an immunity-enhancing probiotic. However, additional investigation is required to confirm the involvement of specific metabolic pathways, secondary metabolites, and bacteriocins in immune responses.
Collapse
Affiliation(s)
- I-Chen Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan
| | - Tsung-Ju Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan City 325, Taiwan
- Department of Food Science, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei City 104, Taiwan
- Institute of Food Science and Technology, National Taiwan University, Taipei City 106, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City 320, Taiwan
| |
Collapse
|
32
|
Huang X, Bao J, Yang M, Li Y, Liu Y, Zhai Y. The role of Lactobacillus plantarum in oral health: a review of current studies. J Oral Microbiol 2024; 16:2411815. [PMID: 39444695 PMCID: PMC11497578 DOI: 10.1080/20002297.2024.2411815] [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: 01/25/2024] [Revised: 07/09/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024] Open
Abstract
Background Oral non-communicable diseases, particularly dental caries and periodontal disease, impose a significant global health burden. The underlying microbial dysbiosis is a prominent factor, driving interest in strategies that promote a balanced oral microbiome. Lactobacillus plantarum, a gram-positive lactic acid bacterium known for its adaptability, has gained attention for its potential to enhance oral health. Recent studies have explored the use of probiotic L. plantarum in managing dental caries, periodontal disease, and apical periodontitis. However, a comprehensive review on its effects in this context is still lacking. Aims This narrative review evaluates current literature on L. plantarum's role in promoting oral health and highlights areas for future research. Content In general, the utilization of L. plantarum in managing non-communicable biofilm-dependent oral diseases is promising, but additional investigations are warranted. Key areas for future study include: exploring its mechanisms of action, identifying optimal strains or strain combinations of L. plantarum, determining effective delivery methods and dosages, developing commercial antibacterial agents from L. plantarum, and addressing safety considerations related to its use in oral care.
Collapse
Affiliation(s)
- Xinyan Huang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Jianhang Bao
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Mingzhen Yang
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| | - Yingying Li
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Youwen Liu
- Orthopedic Department, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yuankun Zhai
- School of Stomatology, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, China
| |
Collapse
|
33
|
Šimiaková M, Bielik V. The pros and cons of probiotic use in pediatric oncology patients following treatment for acute lymphoblastic leukemia. Front Pediatr 2024; 12:1427185. [PMID: 39502562 PMCID: PMC11534854 DOI: 10.3389/fped.2024.1427185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/27/2024] [Indexed: 11/08/2024] Open
Abstract
Acute lymphoblastic leukemia (ALL) treatment, involving chemotherapy, radiotherapy, and pharmacotherapy (antibiotics, antineoplastics) perturbs the gut microbiota in pediatric patients, with enduring effects post-treatment. ALL treatments diminish microbial richness and diversity, favoring pathogenic bacteria. Probiotics may offer promise in mitigating these disruptions and associated side effects. This mini-review explores the impact of ALL treatment on the gut microbiota and the potential benefits of probiotics in pediatric oncology. Probiotics have shown promise in restoring gut microbial balance, reducing treatment-associated side effects, and potentially improving quality of life. However, potential adverse effects, particularly in immunocompromised patients, warrant caution. Notably, there's emerging interest in probiotics' role in bone health and mineral bioaccessibility. Further research is needed to elucidate probiotics' mechanisms and their broader impact on pediatric health. Integration of probiotics into ALL treatment and post-treatment regimens offers significant potential for improving patient outcomes and reducing treatment-related complications and long-lasting disruptions, although careful monitoring is essential.
Collapse
Affiliation(s)
| | - Viktor Bielik
- Department of Biological and Medical Science, Faculty of Physical Education and Sports, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
34
|
Bai Z, Wu Y, Gao D, Dong Y, Pan Y, Gu S. Gut Microbiome and Metabolome Alterations in Overweight or Obese Adult Population after Weight-Loss Bifidobacterium breve BBr60 Intervention: A Randomized Controlled Trial. Int J Mol Sci 2024; 25:10871. [PMID: 39456659 PMCID: PMC11507383 DOI: 10.3390/ijms252010871] [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: 09/08/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Probiotics, known for regulating gut microbiota, may aid those with overweight or obesity, but their mechanisms require more research. This study involved 75 overweight or obese young adults, randomly assigned to either a Bifidobacterium breve BBr60 (BBr60) group or a placebo group. Both groups received diet guidance and took either BBr60 (1 × 1010 CFU/day) or a placebo for 12 weeks. Researchers analyzed body composition, serum glucose, lipids, liver and kidney function, comprehensive metabolome, and intestinal homeostasis before and after the intervention. After 12 weeks, BBr60 significantly reduced weight and BMI compared to pretreatment levels and outperformed the placebo. The BBr60 group also showed improved blood biochemistry, with notably lower fasting blood glucose (FBG) levels than the placebo group (p < 0.05). Additionally, BBr60 influenced vital serum and fecal metabolites related to three amino acid metabolic pathways and regulated the bacteria Dialister, Klebsiella, and Bacteroides, which correlated strongly with serum metabolites. These findings indicate that BBr60 can safely and effectively regulate BMI, body weight, serum glucose, lipids, and liver function markers, which may involve BBr60's impact on key gut bacteria, which influence metabolites related to the valine, leucine, and isoleucine biosynthesis; glycine, serine, and threonine metabolism; and alanine, aspartate, and glutamate metabolism.
Collapse
Affiliation(s)
- Zhouya Bai
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Z.B.); (Y.W.); (D.G.); (Y.P.)
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Z.B.); (Y.W.); (D.G.); (Y.P.)
- Henan Engineering Research Center of Food Microbiology, Luoyang 471000, China
| | - Dejiao Gao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Z.B.); (Y.W.); (D.G.); (Y.P.)
| | - Yao Dong
- Germline Stem Cells and Microenvironment Laboratory, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
| | - Yujia Pan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Z.B.); (Y.W.); (D.G.); (Y.P.)
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang 471000, China; (Z.B.); (Y.W.); (D.G.); (Y.P.)
- Henan Engineering Research Center of Food Microbiology, Luoyang 471000, China
| |
Collapse
|
35
|
Jessen LR, Werner M, Singleton D, Prior C, Foroutan F, Ferran AA, Arenas C, R Bjørnvad C, Lavy E, Allerton F, Allensbach K, Guardabassi L, Unterer S, Bodnárová T, Windahl U, Brennan ML, Weese JS, Scahill K. European Network for Optimization of Veterinary Antimicrobial Therapy (ENOVAT) guidelines for antimicrobial use in canine acute diarrhoea. Vet J 2024; 307:106208. [PMID: 39074542 DOI: 10.1016/j.tvjl.2024.106208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
Acute diarrhoea is a common presentation in dogs, and a common reason for antimicrobial prescription and nutraceutical use. This evidence-based guideline provides recommendations for antimicrobial and probiotic treatment of canine acute diarrhoea (CAD). A multidisciplinary panel developed the recommendations by adhering to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) framework. The opinions of stakeholders (general veterinary practitioners and dog owners) were collected and incorporated to ensure the applicability of this guideline. Four strong recommendations informed by high certainty evidence, and three conditional recommendations informed by very low or low certainty evidence, were drafted by the panel, along with an ungraded section on diagnostic work-up of dogs with acute diarrhoea. The ENOVAT guidelines initiative encourages national or regional guideline makers to use the evidence presented in this document, and the supporting systematic review, to draft national or local guidance documents.
Collapse
Affiliation(s)
- L R Jessen
- Department of Veterinary Clinical Science, University of Copenhagen, Frederiksberg C 1870, Denmark.
| | - M Werner
- Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich 8057, Switzerland
| | - D Singleton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, CH64 7TE, United Kingdom
| | - C Prior
- Willows Veterinary Centre & Referral Centre, Solihull B90 4NH, United Kingdom
| | - F Foroutan
- Faculty of Health Sciences, McMasters University, ON L8S 4L8, Canada
| | - A A Ferran
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse 31076, France
| | - C Arenas
- Internal Medicine Service, AniCura Valencia Sur Hospital Veterinario, Valencia 46460, Spain; VetCT Teleconsulting, Cambridge CB30FA, United Kingdom
| | - C R Bjørnvad
- Department of Veterinary Clinical Science, University of Copenhagen, Frederiksberg C 1870, Denmark
| | - E Lavy
- Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - F Allerton
- Willows Veterinary Centre & Referral Centre, Solihull B90 4NH, United Kingdom
| | - K Allensbach
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, United States
| | - L Guardabassi
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg C 1870, Denmark
| | - S Unterer
- Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich 8057, Switzerland
| | - T Bodnárová
- Veterinary Clinic Podebrady, Poděbrady 290 01, Czech Republic; Centre for Evidence-based Veterinary Medicine, The University of Nottingham, LE12 5RD, United Kingdom
| | - U Windahl
- Department of Animal Health and Antimicrobial Strategies, National Veterinary Institute (SVA), Uppsala SE-75189, Sweden
| | - M L Brennan
- Centre for Evidence-based Veterinary Medicine, The University of Nottingham, LE12 5RD, United Kingdom
| | - J S Weese
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G2W1, Canada
| | - K Scahill
- Evidensia Södra Djursjukhuset Kungens Kurva, Månskärarvägen 13, Kungens Kurva 14175, Sweden; University of Edinburgh, College of Medicine and Veterinary Medicine, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
36
|
Miranda MH, Nader-Macías MEF. Pharmabiotic/phytobiotic formulas approach and their intravaginal effect on different parameters. Vet Res Commun 2024; 48:3019-3033. [PMID: 38980588 DOI: 10.1007/s11259-024-10450-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024]
Abstract
Postpartum reproductive infections in cows generate significant economic losses. The use of lactic acid bacteria in animal health is an alternative tool to avoid antibiotic therapy in the prevention/treatment of bovine reproductive infections. In previous studies, 6 lactic bacteria from bovine mammary glands and vagina with beneficial, safe and technological characteristics were selected, and included in probiotic/phytobiotic formulas (combined with Malva and Lapacho extracts). In this work, probiotic and phytobiotic formulations were designed and their long-term viability determined. They were administered intravaginally to 30 females pregnant bovine pre and postpartum. The modification of the native microbiota and permanence/colonization of cultivable bacteria was evaluated, and also the safety of the designed products through the application of nutritional, clinical, hematological and biochemical parameters. The microorganisms maintained their viability up to 9 months at refrigeration temperature. The number of cultivable bacteria showed different pattern: total aerobic mesophylls increased slightly in all experimental groups, while Enterobacteriaceae increased after delivery, except in beneficial acid lactic bacteria + vegetable extract cows. Control and vegetable extract females showed the highest numbers of Enterobacteriaceae at the end of the trial (30 days postpartum). The number of lactic acid bacteria increased significantly in all the groups between 15 days pre and postpartum. The different parameters evaluated demonstrate the safety and harmlessness of the designed formulas, without producing local and systemic adverse effects in the cows.
Collapse
Affiliation(s)
- María Hortencia Miranda
- Centro de Referencia para Lactobacilos, CONICET. Chacabuco 145, San Miguel de Tucumán, CP4000, Tucumán, Argentina
| | | |
Collapse
|
37
|
Kim DH, Kim SH, Kim SA, Kwak MJ, Han NS, Lee CH. Pathway and Production Differences in Branched-Chain Hydroxy Acids as Bioactive Metabolites in Limosilactobacillus fermentum, Ligilactobacillus salivarius, and Latilactobacillus sakei. Int J Mol Sci 2024; 25:10112. [PMID: 39337595 PMCID: PMC11432647 DOI: 10.3390/ijms251810112] [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: 08/28/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Branched-chain hydroxy acids (BCHAs) as bioactive metabolites of Lactobacillaceae include 2-hydroxy isovaleric acid (HIVA), 2-hydroxy isocaproic acid (HICA), and 2-hydroxy-3-methyl isovaleric acid (HMVA). Combining targeted and untargeted metabolomics, this study elucidates differences in extracellular BCHA production in Limosilactobacillus fermentum, Ligilactobacillus salivarius, and Latilactobacillus sakei alongside comparing comprehensive metabolic changes. Through targeted metabolomics, BCHA production among 38 strains exhibited strain specificity, except for L. sakei, which showed significantly lower BCHA production. Explaining the lower production in L. sakei, which lacks the branched-chain amino acid (BCAA)-utilizing pathway, comparison of BCHA production by precursor reaction revealed that the pathway utilizing BCAAs is more dominant than the pathway utilizing pyruvate. Expanding upon the targeted approach, untargeted metabolomics revealed the effects of the reaction compound on other metabolic pathways besides BCHAs. Metabolism alterations induced by BCAA reactions varied among species. Significant differences were observed in glycine, serine, and threonine metabolism, pyruvate metabolism, butanoate metabolism, and galactose metabolism (p < 0.05). These results emphasize the importance of the synergy between fermentation strains and substrates in influencing nutritional components of fermented foods. By uncovering novel aspects of BCAA metabolism pathways, this study could inform the selection of fermentation strains and support the targeted production of BCHAs.
Collapse
Affiliation(s)
- Dong-Hyuk Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Su-Hyun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Seul-Ah Kim
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Min Jeong Kwak
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Nam Soo Han
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
38
|
Wang T, Zhang X, Fan L, Zhao Y, Zhang Z, Cao Z, Xu Y, Lee S, Lim C, Zhang S. Complete genome sequence and anti-obesity potential of Lactiplantibacillus plantarum HOM2217 in 3T3-L1 cells and high-fat diet-fed rats. Front Microbiol 2024; 15:1436378. [PMID: 39323881 PMCID: PMC11422070 DOI: 10.3389/fmicb.2024.1436378] [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: 05/22/2024] [Accepted: 08/27/2024] [Indexed: 09/27/2024] Open
Abstract
The global prevalence of obesity is rising year by year, which has become a public health problem worldwide. Many animal and clinical studies have shown that Lactiplantibacillus plantarum is considered an ideal probiotic and potential supplement for the treatment of obesity. In this study, we aimed to complete the genome sequence of L. plantarum HOM2217, which was isolated from human milk, and study its physiological characteristics and anti-obesity effects in 3T3-L1 cells and rats fed a high-fat diet (HFD) to determine its potential as a starter for functional food products. Whole-genome analysis demonstrated that HOM2217 contained a single circular chromosome of 3,267,529 bp with a GC content of 44.5% and one plasmid (62,350 bp) with a GC content of 38.5%. Compared to the reference strains, HOM2217 demonstrated superior tolerance to gastrointestinal conditions, higher adhesion to intestinal epithelial cell lines, potent antimicrobial activity against Enterobacter cloacae ATCC 13047, and effective cholesterol removal ability in vitro. Treatment with heat-killed HOM2217 significantly reduced lipid accumulation and intracellular triglyceride production in 3T3-L1 adipocytes. Daily treatment of HFD-fed rats with HOM2217 for 7 weeks decreased body weight, body weight gain, and body fat without changes in food intake. HOM2217 also significantly increased the serum high-density lipoprotein cholesterol (HDL-C) level, decreased the serum tumor necrosis factor (TNF-α) and increased short-chain fatty acid (SCFA) (formic acid, acetic acid, and butyric acid) levels in the cecum. Thus, HOM2217 could potentially prevent obesity in rats by inhibiting inflammatory responses and regulating lipid metabolism and SCFAs expression. Therefore, HOM2217 has potential as an alternative treatment for obesity.
Collapse
Affiliation(s)
- Tingting Wang
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Xiao Zhang
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Linlin Fan
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Ying Zhao
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Zhengwen Zhang
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Zhonghua Cao
- Research Center, Beijing Hanmi Pharmaceutical Co., Ltd., Beijing, China
| | - Ying Xu
- Food & Biotech R&D Center, Coree Beijing Co., Ltd., Beijing, China
| | - Suwon Lee
- Food & Biotech R&D Center, Coree Beijing Co., Ltd., Beijing, China
| | - Chongyoon Lim
- Food & Biotech R&D Center, Coree Beijing Co., Ltd., Beijing, China
| | - Shiqi Zhang
- Food & Biotech R&D Center, Coree Beijing Co., Ltd., Beijing, China
| |
Collapse
|
39
|
Chen X, Li Q, Xie J, Nie S. Immunomodulatory Effects of Probiotic-Derived Extracellular Vesicles: Opportunities and Challenges. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19259-19273. [PMID: 39177683 DOI: 10.1021/acs.jafc.4c04223] [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: 08/24/2024]
Abstract
Probiotics are known to modulate host immune responses in the course of many diseases. Recently, bacterial extracellular vesicles (EVs), which contain bioactive proteins, lipids, nucleic acids, and metabolites released by bacteria, have been identified as potentially important mediators of bacteria-bacterium and bacteria-host interactions. With the deepening of research, it has been found that probiotic-derived EVs play a significant role in regulating host immune function and, thus, exerting health-promoting effects. Nevertheless, current research is in its early stages, and there remains a long way to go to bridge the gap between basic research and clinical practice. In this review, we describe the fundamental aspects of probiotic-derived EVs, including their biogenesis, cargo sorting mechanism, and transport capabilities. We further discussed the potential mechanisms of probiotic-derived EVs in regulating the host's gut microbiota and immune responses. Finally, we speculate about the potential of probiotic-derived EVs as new postbiotics for applications in functional food, disease treatment substitutes, and immune regulatory adjuvants.
Collapse
Affiliation(s)
- Xinyang Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Qiqiong Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Junhua Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
40
|
Dhakephalkar T, Pisu V, Margale P, Chandras S, Shetty D, Wagh S, Dagar SS, Kapse N, Dhakephalkar PK. Strain-Dependent Adhesion Variations of Shouchella clausii Isolated from Healthy Human Volunteers: A Study on Cell Surface Properties and Potential Probiotic Benefits. Microorganisms 2024; 12:1771. [PMID: 39338446 PMCID: PMC11434523 DOI: 10.3390/microorganisms12091771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
The probiotic potential of Shouchella clausii is widely recognized, but little is known about its adhesive properties. Hence, this study aims to investigate the adhesion potential and cell surface properties of four human-origin S. clausii strains (B619/R, B603/Nb, B106, and B637/Nm). We evaluated epithelial adhesion, Extracellular Matrix (ECM) binding, aggregation ability, and cell surface hydrophobicity and used genome analysis for validation. Our results demonstrate that adhesion capability is a strain-specific attribute, with significant variations observed among the four strains. B619/R, B603/Nb, and B106 displayed stronger adhesion properties than B637/Nm. Supplementary adhesion assays showed that B637/Nm displayed high hydrophobicity, significant auto-aggregation, and significant mucin-binding abilities. Conversely, B619/R, B603/Nb, and B106 had mildly hydrophobic surfaces and low aggregation abilities. Genome annotation revealed the presence of various adhesion proteins in four strains. Notably, the reduced adhesion potential of B637/Nm was supported by the absence of the cell wall surface anchor family protein (LPxTG motif), which is crucial for interactions with intestinal epithelial cells or mucus components. Further, docking studies provided insights into the interaction of adhesion proteins with gut mucins. These findings contribute to a better understanding of how S. clausii strains interact with the gut environment, facilitating the development of probiotic formulations tailored for improved gut health and well-being.
Collapse
Affiliation(s)
- Tanisha Dhakephalkar
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Vaidehi Pisu
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Prajakta Margale
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Siddhi Chandras
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Deepa Shetty
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Shilpa Wagh
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Sumit Singh Dagar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Neelam Kapse
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Prashant K Dhakephalkar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| |
Collapse
|
41
|
Theodorou IM, Kapoukranidou D, Theodorou M, Tsetis JK, Menni AE, Tzikos G, Bareka S, Shrewsbury A, Stavrou G, Kotzampassi K. Cosmeceuticals: A Review of Clinical Studies Claiming to Contain Specific, Well-Characterized Strains of Probiotics or Postbiotics. Nutrients 2024; 16:2526. [PMID: 39125405 PMCID: PMC11314542 DOI: 10.3390/nu16152526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The skin serves as a critical barrier against external threats-dehydration, ultraviolet exposure, and infections-playing a significant role in internal homeostasis and moisture retention. Additionally, and equally importantly, it interacts dynamically with the complex microbiome resident in it, which is essential for maintaining skin health. Recent interest has focused on the use of probiotics and postbiotics, besides their ability to modulate the skin microbiome, to enhance barrier function, and exhibit anti-inflammatory properties, to be involved in skincare, by having the potential to improve skin hydration, elasticity, and overall appearance, as well as in reducing signs of aging, such as wrinkles and fine lines. The products-being a combination of a cosmetic regime plus probiotic[s] or postbiotic[s]-are named cosmeceuticals. However, to comply with the regulations for the characterization of a microorganism as a specific probiotic strain, the pro- or postbiotics incorporated into the cosmetic regime should be both genetically and phenotypically defined. Thus, in this review, we present 14 published clinical trials using such cosmetic products with specific, well-characterized strains of probiotics or postbiotics applied to volunteers with healthy skin. Looking at the results of these studies collectively, we can say that these genetically and phenotypically defined strains of either live or inanimate bacteria and/or their components seem to keep the treated skin at least fully hydrated, with intact epithelial tone, increased radiance, and with decreased wrinkle depth, while normalizing the commensal skin microbiota. Future advancements in personalized skin care may lead to genomic sequencing and metabolomics to tailor probiotic and postbiotic treatments to individual skin microbiomes, promising a new frontier in cosmeceuticals.
Collapse
Affiliation(s)
| | - Dorothea Kapoukranidou
- Department of Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | | | | | | | - Georgios Tzikos
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stella Bareka
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Anne Shrewsbury
- Department of Surgery, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - George Stavrou
- Department of Surgery, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK
| | | |
Collapse
|
42
|
Zaidi SMH, Haider R, Kazmi SAB, Husnain A, Khan S, Merchant S, Tayyab H, Wazeen FR, Chaudhary AJ. Beyond Antibiotics: Novel Approaches in the Treatment of Recurrent Clostridioides difficile Infection. ACG Case Rep J 2024; 11:e01333. [PMID: 39081300 PMCID: PMC11286250 DOI: 10.14309/crj.0000000000001333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/06/2024] [Indexed: 08/02/2024] Open
Affiliation(s)
| | - Ramsha Haider
- Karachi Medical and Dental College, Karachi, Pakistan
| | | | - Ali Husnain
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Saniah Khan
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | | | - Hamnah Tayyab
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Fazl Rahim Wazeen
- Department of Medicine, Greater Baltimore Medical Center, Towson, MD
| | | |
Collapse
|
43
|
Kim B, Lee Y, Lee C, Jung ES, Kang H, Holzapfel WH. Comprehensive Amelioration of Metabolic Dysfunction through Administration of Lactiplantibacillus plantarum APsulloc 331261 (GTB1™) in High-Fat-Diet-Fed Mice. Foods 2024; 13:2227. [PMID: 39063311 PMCID: PMC11276112 DOI: 10.3390/foods13142227] [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: 06/10/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
The beneficial effects of probiotics for the improvement of metabolic disorders have been studied intensively; however, these effects are evident in a probiotic strain-specific and disease-specific manner. Thus, it is still essential to evaluate the efficacy of each strain against a target disease. Here, we present an anti-obese and anti-diabetic probiotic strain, Lactiplantibacillus plantarum APsulloc331261 (GTB1™), which was isolated from green tea and tested for safety previously. In high-fat-diet-induced obese mice, GTB1™ exerted multiple beneficial effects, including significant reductions in adiposity, glucose intolerance, and dyslipidemia, which were further supported by improvements in levels of circulating hormones and adipokines. Lipid metabolism in adipose tissues was restored through the activation of PPAR/PGC1α signaling by GTB1™ treatment, which was facilitated by intestinal microbiota composition changes and short-chain fatty acid production. Our findings provide evidence to suggest that GTB1™ is a potential candidate for probiotic supplementation for comprehensive improvement in metabolic disorders.
Collapse
Affiliation(s)
- Bobae Kim
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| | - Yuri Lee
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| | - Chungho Lee
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
| | - Eun Sung Jung
- Multi-Omics Center, HEM Pharma Inc., Suwon 16229, Republic of Korea;
| | - Hyeji Kang
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Global Green Research Institute, Handong Global University, Pohang 37554, Republic of Korea
| | - Wilhelm H. Holzapfel
- Basic Research Center, HEM Pharma Inc., Pohang 37554, Republic of Korea; (B.K.); (Y.L.); (C.L.)
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| |
Collapse
|
44
|
Singh A, Mazumder A, Das S, Tyagi PK, Chaitanya MVNL. Probiotics in Action: Enhancing Immunity and Combatting Diseases for Optimal Health. JOURNAL OF NATURAL REMEDIES 2024:1153-1167. [DOI: 10.18311/jnr/2024/35894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/22/2024] [Indexed: 01/03/2025]
Abstract
This review offers an in-depth examination of the mechanisms underlying the microbiome's defense against viral infections, with a specific focus on probiotic interventions. Mycotoxins, secondary compounds produced by microfungi, pose significant health risks. Yet, certain strains of Lactic Acid Bacteria (LAB) have exhibited remarkable efficacy in eliminating aflatoxin B1 (AFB1), the most toxic member of the aflatoxin family. Experimental setups demonstrated AFB1 binding to specific LAB strains, persisting even after gastric digestion. Laboratory studies revealed a potential protective mechanism wherein pre-incubation of probiotics with mycotoxins reduced their adhesion to mucus. Animal trials further underscored the benefits of oral probiotic administration, showcasing increased fecal excretion of mycotoxins and mitigation of associated health risks. Cyanobacteria-generated microcystins in drinking water pose a significant threat to human health. Probiotic bacteria, particularly strains like Bifidobacterium longum and Lactobacillus rhamnosus, have demonstrated exceptional efficacy in removing the cyanobacterial peptide toxin microcystin-LR. Optimized conditions resulted in rapid toxin elimination, highlighting the potential of probiotics in water purification. Engineered probiotics represent a cutting-edge approach to tailor microorganisms for specific therapeutic applications, exhibiting promise in treating metabolic disorders, Alzheimer's disease, and type 1 diabetes. Additionally, they serve as innovative diagnostic tools, capable of detecting pathogens and inflammation markers within the body. In the realm of antimicrobial peptide production, probiotics offer a promising platform, with genetically modified strains engineered to produce human β-defensin 2 (HBD2) for treating Crohn's disease, showcasing their potential in targeted theurapetic delivery. Biocontainment strategies have been implemented to prevent unintended environmental impacts.
Collapse
|
45
|
Bermúdez-Humarán LG, Chassaing B, Langella P. Exploring the interaction and impact of probiotic and commensal bacteria on vitamins, minerals and short chain fatty acids metabolism. Microb Cell Fact 2024; 23:172. [PMID: 38867272 PMCID: PMC11167913 DOI: 10.1186/s12934-024-02449-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
There is increasing evidence that probiotic and commensal bacteria play a role in substrate metabolism, energy harvesting and intestinal homeostasis, and may exert immunomodulatory activities on human health. In addition, recent research suggests that these microorganisms interact with vitamins and minerals, promoting intestinal and metabolic well-being while producing vital microbial metabolites such as short-chain fatty acids (SCFAs). In this regard, there is a flourishing field exploring the intricate dynamics between vitamins, minerals, SCFAs, and commensal/probiotic interactions. In this review, we summarize some of the major hypotheses beyond the mechanisms by which commensals/probiotics impact gut health and their additional effects on the absorption and metabolism of vitamins, minerals, and SCFAs. Our analysis includes comprehensive review of existing evidence from preclinical and clinical studies, with particular focus on the potential interaction between commensals/probiotics and micronutrients. Finally, we highlight knowledge gaps and outline directions for future research in this evolving field.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Philippe Langella
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| |
Collapse
|
46
|
Pal BB, Bandagi RV, Pebbili KK, Rathod R, Kotak B, Dhanaki G, Shah S. Effectiveness of Saccharomyces boulardii CNCM I-745 in Adult Indian Patients with Diarrhoea: A Real-world, Multicentre, Retrospective, Comparative Study. Drugs Real World Outcomes 2024; 11:309-316. [PMID: 38581564 PMCID: PMC11176121 DOI: 10.1007/s40801-024-00424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Multiple clinical studies have described the benefits of probiotic Saccharomyces boulardii (S. boulardii) CNCM I-745 against diarrhoea, but the real-world evidence supporting its use is lacking. OBJECTIVE To evaluate effectiveness of the S. boulardii CNCM I-745 group in a real-world setting. METHODS This was an electronic medical record (EMR)-based, retrospective, multicentre, comparative study in Indian adult patients presenting with diarrhoea managed between January 2020 and January 2022. Data of patients at the baseline visit, with a follow-up visit within 15 days, and who were administered S. boulardii CNCM I-745 (for the test group) or any other treatment modality excluding probiotics (for the control group) were considered. Effectiveness was evaluated on the basis of number of patients who did not complain of diarrhoea at follow-up. RESULTS Of 30,385 adult patients with diarrhoea, 270 patients prescribed S. boulardii CNCM I-745 were included, while the control group comprised 1457 patients. The baseline median age of the test group was 47 years (range 19-86 years), while it was 44 years (range 19-100 years) for the control group. The majority of patients in both study groups were females (56.7% in the test and 51.5% in the control group). Median duration between visits was 5 days (range 1-15 days) in both study groups. In all, 77.8% patients (95% CI 72.34-82.59) in the test group did not complain of diarrhoea at follow-up, while the proportion was 15.8% (95% CI 13.95-17.76) in the control group (p < 0.05). Odds ratio (OR) for absence of diarrhoea in the S. boulardii CNCM I-745 group versus the control group was 18.7 (95% CI 13.6-25.7, p < 0.05). For subgroups on concomitant antibiotics, a significant advantage was noted again for the test versus the control group (76.8% versus 18.4%; p < 0.05; OR: 14.7 with 95% CI 8.8-24.4; p < 0.05). CONCLUSION The effect of S. boulardii CNCM I-745 probiotic in controlling diarrhoea was better than anti-diarrhoeal and/or oral rehydration therapy in real-world clinical practice. The effect was similar even with concomitant antibiotic usage.
Collapse
Affiliation(s)
| | | | - Kranthi Kiran Pebbili
- Department of Medical Affairs, Dr Reddy's Laboratories Ltd, Hyderabad, Telangana, India
| | - Rahul Rathod
- Department of Medical Affairs, Dr Reddy's Laboratories Ltd, Hyderabad, Telangana, India
| | - Bhavesh Kotak
- Department of Medical Affairs, Dr Reddy's Laboratories Ltd, Hyderabad, Telangana, India
| | - Gauri Dhanaki
- Department of Medical Affairs, Dr Reddy's Laboratories Ltd, Hyderabad, Telangana, India
| | - Snehal Shah
- Department of Clinical Insights, Healthplix Technologies, Bangalore, India
| |
Collapse
|
47
|
Chen K, Zhou Z, Nie Y, Cao Y, Yang P, Zhang Y, Xu P, Yu Q, Shen Y, Ma W, Jin S, Liu C. Adjunctive efficacy of Bifidobacterium animalis subsp. lactis XLTG11 for functional constipation in children. Braz J Microbiol 2024; 55:1317-1330. [PMID: 38381349 PMCID: PMC11153453 DOI: 10.1007/s42770-024-01276-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Functional constipation (FC) can seriously affect the physical and mental health of children. The goal of this study is to assess the efficacy and safety of Bifidobacterium animalis subsp. lactis XLTG11 in treating FC in children through a randomized, double-blinded, placebo-controlled approach. Eligible children were randomized into either the intervention group (IG, n = 65, receiving conventional treatment with probiotics) or the control group (CG, n = 66, receiving conventional treatment without probiotics). The primary outcome measure was fecal frequency. Fecal gut microbiota analysis and PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) were used to predict gene family abundances based on 16S information. Over the course of treatment, the weekly frequency of feces within each group increased significantly (F = 41.97, p < 0.001). The frequency of feces (times/week (t/w)) in the IG was significantly higher than that in the CG (3.69 ± 2.62 t/w vs.3.18 ± 1.43 t/w, 4.03 ± 2.54 t/w vs. 2.89 ± 1.39 t/w and 3.74 ± 2.36 t/w vs. 2.94 ± 1.18 t/w and 3.45 ± 1.98 vs. 3.17 ± 1.41 t/w for the 1st, 2nd, 3rd, and 4th week after intervention, respectively) (F = 7.60, p = 0.0067). After the intervention, dominate species shifted to Bifidobacterium longum, Bifidobacterium breve, and Escherichia coli in the IG. Additionally, genes related to short-chain fatty acid (SCF) metabolism were upregulated, while methane metabolism was downregulated. Administration of XLTG11 at a dose of 1 × 1010 CFU/day to children increased fecal frequency, induced beneficial changes in gut microbiota, and regulated SCFs and methane metabolism-related genes.
Collapse
Affiliation(s)
- Ke Chen
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Zengyuan Zhou
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Nie
- Department of Child Health Care, Chongzhou Maternal and Child Health Care Hospital, Chengdu, China
| | - Yanmei Cao
- Department of Child Health Care, Dayi Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Yang
- Department of Child Health Care, Xindu Maternal and Child Health Care Hospital, Chengdu, China
| | - Ying Zhang
- Department of Child Health Care, Jinniu Maternal and Child Health Care Hospital, Chengdu, China
| | - Ping Xu
- Department of Child Health Care, Qingbaijiang Maternal and Child Health Care Hospital, Chengdu, China
| | - Qinghua Yu
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co, Limited, Shanghai, China
| | - Yang Shen
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co, Limited, Shanghai, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shanshan Jin
- Department of Nutrition, School of Medicine, Chengdu Women's & Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Changqi Liu
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, USA
| |
Collapse
|
48
|
Aljohani A, Rashwan N, Vasani S, Alkhawashki A, Wu TT, Lu X, Castillo DA, Xiao J. The Health Benefits of Probiotic Lactiplantibacillus plantarum: A Systematic Review and Meta-Analysis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10287-3. [PMID: 38816672 DOI: 10.1007/s12602-024-10287-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 06/01/2024]
Abstract
To ensure effective administration of probiotics in clinical practice, it is crucial to comprehend the specific strains and their association with human health. Therefore, we conducted a systematic review and meta-analysis to evaluate the scientific evidence on the impact of Lactiplantibacillus plantarum probiotic consumption on human health. Out of 11,831 records, 135 studies were assessed qualitatively, and 18 studies were included in the meta-analysis. This systematic review demonstrated that probiotic supplementation with L. plantarum, either alone or in combination, can significantly improve outcomes for patients with specific medical conditions. Meta-analysis revealed notable benefits in periodontal health, evidenced by reduced pocket depth and bleeding on probing (p < 0.001); in gastroenterological health, marked by significant reductions in abdominal pain (p < 0.001); and in infectious disease, through a reduction in C-reactive protein levels (p < 0.001). Cardiovascular benefits included lowered total cholesterol and low-density lipoprotein cholesterol in the L. plantarum intervention group (p < 0.05). Our study's clinical significance highlights the importance of considering probiotic strain and their application to specific diseases when planning future studies and clinical interventions, emphasizing the need for further research in this area.
Collapse
Affiliation(s)
- Amal Aljohani
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Noha Rashwan
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Shruti Vasani
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Ahmed Alkhawashki
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
- Pediatrics, King Fahd Medical City, Riyadh, Saudi Arabia
| | - Tong Tong Wu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, USA
| | - Xingyi Lu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, USA
| | - Daniel A Castillo
- Miner Library, University of Rochester Medical Center, Rochester, NY, USA
| | - Jin Xiao
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
49
|
Meng J, Liu S, Wu X. Engineered probiotics as live biotherapeutics for diagnosis and treatment of human diseases. Crit Rev Microbiol 2024; 50:300-314. [PMID: 36946080 DOI: 10.1080/1040841x.2023.2190392] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/09/2023] [Indexed: 03/23/2023]
Abstract
The use of probiotics to regulate the intestinal microbiota to prevent and treat a large number of disorders and diseases has been an international research hotspot. Although conventional probiotics have a certain regulatory role in nutrient metabolism, inhibiting pathogens, inducing immune regulation, and maintaining intestinal epithelial barrier function, they are unable to treat certain diseases. In recent years, aided by the continuous development of synthetic biology, engineering probiotics with desired characteristics and functionalities to benefit human health has made significant progress. In this article, we summarise the mechanism of action of conventional probiotics and their limitations and highlight the latest developments in the design and construction of probiotics as living diagnostics and therapeutics for the detection and treatment of a series of diseases, including pathogen infections, cancer, intestinal inflammation, metabolic disorders, vaccine delivery, cognitive health, and fatty liver. Besides we discuss the concerns regarding engineered probiotics and corresponding countermeasures and outline the desired features in the future development of engineered live biotherapeutics.
Collapse
Affiliation(s)
- Jiao Meng
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
| | - Shufan Liu
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology; College of Bioengineering, Tianjin University of Science and Technology, Tianjin, China
| | - Xin Wu
- Laboratory of Nutrient Resources and Synthetic Biology, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Science, Tianjin, China
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
50
|
Belotserkovsky I, Stabryla LM, Hunter M, Allegretti J, Callahan BJ, Carlson PE, Daschner PJ, Goudarzi M, Guyard C, Jackson SA, Rao K, Servetas SL, Sokol H, Wargo JA, Novick S. Standards for fecal microbiota transplant: Tools and therapeutic advances. Biologicals 2024; 86:101758. [PMID: 38518435 DOI: 10.1016/j.biologicals.2024.101758] [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: 05/26/2023] [Accepted: 03/04/2024] [Indexed: 03/24/2024] Open
Abstract
Fecal microbiota transplantation (FMT) has been demonstrated to be efficacious in preventing recurrent Clostridioides difficile (C. difficile) infections, and is being investigated for treatment of several other diseases including inflammatory bowel disease, cancer, obesity, liver disease, and diabetes. To speed up the translation of FMT into clinical practice as a safe and standardized therapeutic intervention, additional evidence-based technical and regulatory guidance is needed. To this end in May of 2022, the International Alliance for Biological Standardization (IABS) and the BIOASTER Microbiology Technology Institute hosted a second webinar to discuss key issues still impeding the advancement and standardization of FMT. The goal of this two-day webinar was to provide a forum for scientific experts to share and discuss data and key challenges with one another. Discussion included a focus on the evaluation of safety, efficacy, clinical trial design, reproducibility and accuracy in obtained microbiome measurements and data reporting, and the potential for standardization across these areas. It also focused on increasing the application potential and visibility of FMT beyond treating C. difficile infections.
Collapse
Affiliation(s)
| | - Lisa M Stabryla
- Complex Microbial Systems Group, Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Monique Hunter
- Complex Microbial Systems Group, Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Jessica Allegretti
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Benjamin J Callahan
- Bioinformatics Research Center, North Carolina State University, Raleigh, 27606, USA; Department of Population Health and Pathobiology, North Carolina State University, Raleigh, 27607, USA
| | - Paul E Carlson
- Laboratory of Mucosal Pathogens and Cellular Immunology, Division of Bacterial, Parasitic, and Allergenic Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Phillip J Daschner
- Division of Cancer Biology, National Cancer Institute, Bethesda, MD, USA
| | | | - Cyril Guyard
- BIOSTER Technological Research Institute, Lyon, France
| | - Scott A Jackson
- Complex Microbial Systems Group, Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Krishna Rao
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephanie L Servetas
- Complex Microbial Systems Group, Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, MD, USA
| | - Harry Sokol
- Assistance Publique des Hôpitaux de Paris, Saint-Antoine Hospital, Gastroenterology Department, Paris, France
| | - Jennifer A Wargo
- Departments of Surgical Oncology and Genomic Medicine, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shawn Novick
- BioPhia Consulting, Inc., 7307 W. Green Lake Dr. N., Seattle, WA, 98103, USA.
| |
Collapse
|