1
|
Gelli HP, Vazquez-Uribe R, Buckley ST, Andersen JT, Alexander Sommer MO. Advanced microbiome therapeutics for oral delivery of peptides and proteins: Advances, challenges, and opportunities. Adv Drug Deliv Rev 2025:115603. [PMID: 40349728 DOI: 10.1016/j.addr.2025.115603] [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: 04/03/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Peptide and protein medicines have changed the therapeutic landscape for many diseases, yet oral delivery remains a significant challenge due to enzymatic degradation, instability, and poor permeability in the gastrointestinal tract. Advanced Microbiome Therapeutics (AMTs) could overcome some of these barriers by producing and releasing therapeutic peptides directly in the gastrointestinal tract. AMTs can localize peptide production at the site of absorption, providing either sustained or controlled release while potentially reducing side effects associated with systemic administration. Here, this review assesses the status of AMTs for oral peptide delivery and discusses the potential integration of enzyme inhibitors, permeation enhancers, and mucoadhesive to improve oral bioavailability further. Combining these approaches could pave the way for more widespread oral delivery strategies for peptide and protein medicines.
Collapse
Affiliation(s)
- Hitesh P Gelli
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | | | - Jan Terje Andersen
- Department of Pharmacology, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | | |
Collapse
|
2
|
Bijle MN, Pudipeddi A, Yiu C. Effect of direct arginine supplementation on growth of Lacticaseibacillus rhamnosus GG. J Dent 2025; 158:105800. [PMID: 40324576 DOI: 10.1016/j.jdent.2025.105800] [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: 03/18/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVES The study aimed to examine the effect of direct arginine (Arg) supplementation on Lacticaseibacillus rhamnosus GG (LRG) growth. METHODS The probiotic LRG ATCC 53103 was routinely cultured in MRS broth and adjusted to 107 CFU/ml. l-Arg at 0.25 % and 0.5 % w/v. was directly supplemented to MRS broth containing LRG. Then, LRG with/without supplemented Arg was characterised to determine growth curves for 72 h under a CO2-driven, environment-controlled spectrophotometer. Whereas, end-point McFarland spectrophotometric, metabolic assays (XTT and WST-8), colony forming units (CFU) assessment, live/dead bacterial confocal imaging, and viable DNA quantification were undertaken after 24 h CO2 incubation to determine LRG growth with/without supplemented Arg. RESULTS Growth curve analysis revealed similar absorbance profiles with LRG and LRG+0.25 % Arg with no overlaps to LRG+0.5 % Arg; with a similar trend observed as per end-point McFarland spectrophotometric assessment. The % viability estimated by metabolic assays with LRG and LRG+0.25 % Arg was significantly lower than LRG+0.5 % Arg (p < 0.05). As per CFU assessment, CFU/ml for LRG+0.25 % Arg and LRG+0.5 % Arg was significantly higher than LRG control (p < 0.05). LRG live-dead proportions determined using confocal imaging denote no differences between the groups (p > 0.05), with live cell proportions significantly higher than the dead cells (p < 0.05). However, LRG cellular density appeared to increase with increasing concentrations of Arg. Additionally, the viable cells quantified with PMA-qPCR significantly increased with increasing concentrations of Arg (p < 0.05). CONCLUSION Direct supplementation of 0.5 % Arg to LRG enhances LRG growth, demonstrating a promising synbiotic potential. CLINICAL SIGNIFICANCE (48 WORDS): Primary caries prevention is initiated at biofilms using biotic strategies that modulate the microbiome matrix to maintain ecological homeostasis. The results of the present study establish a deliverable synbiotic of Arg and probiotic Lacticaseibacillus rhamnosus GG, demonstrating a promising synergism between interventions in the milieu targeted for high caries-risk individuals.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- Dr. D. Y. Patil Dental College & Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune, Maharashtra, India.
| | | | - Cynthia Yiu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
3
|
Loupy KM, Dawud LM, Zambrano CA, Lee T, Heinze JD, Elsayed AI, Hassell JE, D'Angelo HM, Frank MG, Maier SF, Brenner LA, Lowry CA. Effects of Oral Administration of the Probiotic Lactobacillus rhamnosus GG on the Proteomic Profiles of Cerebrospinal Fluid and Immunoregulatory Signaling in the Hippocampus of Adult Male Rats. Neuroimmunomodulation 2025; 32:94-109. [PMID: 40031897 DOI: 10.1159/000544842] [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: 11/24/2024] [Accepted: 02/08/2025] [Indexed: 03/05/2025] Open
Abstract
INTRODUCTION The microbiome-gut-brain axis, by modulating bidirectional immune, metabolic, and neural signaling pathways in the host, has emerged as a target for the prevention and treatment of psychiatric and neurological disorders. Oral administration of the probiotic bacterium Lactobacillus rhamnosus GG (LGG; ATCC 53103) exhibits anti-inflammatory effects, although the precise mechanisms by which LGG benefits host physiology and behavior are not known. The goal of this study was to explore the general effects of LGG on the cerebrospinal fluid (CSF) proteome and a biological signature of anti-inflammatory signaling in the central nervous system (CNS) of undisturbed, adult male rats. METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were conducted using CSF samples collected after 21 days of oral treatment with live LGG (3.34 × 107 colony-forming units (CFU)/mL in the drinking water (resulting in an estimated delivery of ∼1.17 × 109 CFU/day/rat) or water vehicle. Gene enrichment analysis (using DAVID, v. 6.8) and protein-protein interactions (using STRING, v. 11) were used to explore physiological network changes in CSF. Real-time reverse transcription polymerase chain reaction (real-time RT-PCR) was performed to assess gene expression changes of anti-inflammatory cytokines in the hippocampus. Genes associated with anti-inflammatory signaling that were analyzed included Il10, Tgfb1, Il4, and IL-4-responsive genes, Cd200, Cd200r1, and Mrc1 (Cd206). RESULTS Oral LGG administration altered the abundance of CSF proteins, increasing the abundance of five proteins (cochlin, NPTXR, reelin, Sez6l, and VPS13C) and decreasing the abundance of two proteins (CPQ, IGFBP-7) in the CSF. Simultaneously, LGG increased the expression of Il10 mRNA, encoding the anti-inflammatory cytokine interleukin 10, in the hippocampus. CONCLUSION Oral LGG altered the abundance of CSF proteins associated with extracellular scaffolding, synaptic plasticity, and glutamatergic signaling. These data are consistent with the hypothesis that oral administration of LGG improves memory and cognition, and promotes a physiological resilience to neurodegenerative disease, by increasing glutamatergic signaling and promoting an anti-inflammatory environment in the brain.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lamya'a M Dawud
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, Colorado, USA
| | - Jared D Heinze
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - James E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
| | - Lisa A Brenner
- Rocky Mountain Regional VA Medical Center (RMRVAMC), Aurora, Colorado, USA
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, Colorado, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, Colorado, USA
- Rocky Mountain Regional VA Medical Center (RMRVAMC), Aurora, Colorado, USA
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, Colorado, USA
- Center for Microbial Exploration, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
4
|
Phùng TTT, Dupont S, Beney L, Chanut J, Karbowiak T. Unlocking Probiotic Potential: Physicochemical Approaches to Evaluate Probiotic Bacterial Adhesion Potential to the Intestinal Tract. Mol Nutr Food Res 2025:e202400705. [PMID: 39846848 DOI: 10.1002/mnfr.202400705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 01/24/2025]
Abstract
Bacterial adhesion in the gut is critical to evaluate their effectiveness as probiotics. Understanding the bacterial adhesion within the complex gut environment is challenging. This study explores the adhesion mechanisms and the adhesion potential of five selected bacterial strains (Escherichia coli, Lactiplantibacillus plantarum, Faecalibacterium duncaniae, Bifidobacterium longum, and Bifidobacterium longum subsp. infantis) at the initial stages when bacterial cells arriving in the gut, using different physicochemical approaches. Bacterial morphology, rheology, and surface properties were evaluated. Surprisingly, previous methods such as bacterial adhesion to hydrocarbon and the interfacial tension between bacterial suspensions and mineral oil did not fully capture the bacterial adhesion to intestinal mucus. Consequently, this study introduced a novel approach to assess bacterial adhesion to mucus, based on contact angle measurements, calculation of surface tension, and work of adhesion. Interestingly, both small and large intestinal mucus are rather hydrophilic, and thus highly hydrophilic bacteria such as E. coli and B. infantis tend to adhere better. Additionally, a multicriteria evaluation of bacterial adhesion to the gut, from the bulk liquid transport stage until the irreversible adhesion, was proposed. E. coli and B. infantis demonstrated the highest overall adhesion potential in the intestinal tract, followed by Lpb. plantarum, B. longum, and F. duncaniae, respectively. This work contributed original physicochemical approaches to comprehensively examine bacterial adhesion in the gut.
Collapse
Affiliation(s)
- Thị-Thanh-Trúc Phùng
- L'institut Agro, Université Bourgogne Europe, INRAe, UMR PAM, Dijon, F-21000, France
| | - Sébastien Dupont
- L'institut Agro, Université Bourgogne Europe, INRAe, UMR PAM, Dijon, F-21000, France
| | - Laurent Beney
- L'institut Agro, Université Bourgogne Europe, INRAe, UMR PAM, Dijon, F-21000, France
| | - Julie Chanut
- L'institut Agro, Université Bourgogne Europe, INRAe, UMR PAM, Dijon, F-21000, France
| | - Thomas Karbowiak
- L'institut Agro, Université Bourgogne Europe, INRAe, UMR PAM, Dijon, F-21000, France
| |
Collapse
|
5
|
Bingöl FG, Ağagündüz D, Budán F. Probiotic Bacterium-Derived p40, p75, and HM0539 Proteins as Novel Postbiotics and Gut-Associated Immune System (GAIS) Modulation: Postbiotic-Gut-Health Axis. Microorganisms 2024; 13:23. [PMID: 39858791 PMCID: PMC11767761 DOI: 10.3390/microorganisms13010023] [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: 12/08/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
It is known that probiotics have direct and indirect effects on many systems in the body, especially the gastrointestinal system. Interest in using probiotic strain-derived cell components and metabolites has also increased as a result of the significant benefits of probiotics. Although many terminologies and definitions are used for these components and metabolites, the International Scientific Association of Probiotics and Prebiotics (ISAPP) recommended the use of the term postbiotic in 2021, which is defined as "a preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics are bioactive metabolites such as organic acids, peptides/proteins, cell wall components, functional enzymes, short-chain fatty acids, vitamins, and phenols. These molecules mediate many positive effects such as immunomodulatory, antimicrobial, and antioxidant effects. These positive effects on maintaining health have enabled the identification of many new postbiotic proteins such as p40, p75, and HM0539. In this review, the postbiotic proteins p40, p75, and HM0539 derived from lactobacilli and their functional effects are systematically summarized. The p40 protein, in particular, has been shown to support gut barrier activity and reduce inflammation, potentially through mechanisms involving epidermal growth factor receptor-dependent signaling. Additionally, p40 and p75 proteins exhibit protective effects on intestinal epithelial tight junctions, suggesting their therapeutic potential in preventing intestinal damage and diseases such as colitis. HM0539 enhances intestinal barrier integrity, exhibits antiinflammatory properties, and protects against bacterial infection, suggesting its possible as a therapeutic for inflammatory bowel disease. This review may contribute to future studies on the therapeutic use of p40, p75, and HM0539 postbiotic proteins in inflammatory gastrointestinal system diseases.
Collapse
Affiliation(s)
- Feray Gençer Bingöl
- Department of Nutrition and Dietetics, Faculty of Health Science, Burdur Mehmet Akif Ersoy University, 15200 Burdur, Türkiye;
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Science, Gazi University, 06490 Ankara, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
6
|
Pino A, Hiippala K, Ronkainen A, Vaccalluzzo A, Caggia C, Satokari R, Randazzo CL. Adhesion Properties and Pathogen Inhibition of Vaginal-Derived Lactobacilli. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10390-5. [PMID: 39565565 DOI: 10.1007/s12602-024-10390-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/21/2024]
Abstract
In the present study, twenty-seven (27) lactobacilli strains, isolated from the vagina of healthy Italian women of reproductive age, were screened for probiotic properties. The strains were evaluated for antagonistic activity against pathogens, adhesion abilities, and potential to displace and/or inhibit the adhesion of previously adhered pathogens as a primary strain selection criterion. Overall, all the tested lactobacilli inhibited at least three pathogens, and the majority of them exhibited antimicrobial activity against Enterobacter cloacae DSM 30054, Pseudomonas aeruginosa DSM 3227, and Pseudomonas aeruginosa DSM 1117. The complete neutralization of antimicrobial activity after cell-free supernatant (CFS) neutralization suggested a pivotal role for lactic acid or other organic acids secreted by the lactobacilli. The strains showed variability in their adhesion levels, but all tested strains adhered to both human colonic epithelial cells (HT-29) and vaginal cells (VK2/E6E7) with adhesion percentages exceeding 1%. The ability to displace or inhibit pathogens was dependent on the pathogen and the lactobacilli strain; the pathogen displacement levels ranged from 9 to 82%, while pathogen exclusion levels varied from 1 to 99%. In conclusion, this study demonstrates the protective effect of vaginal lactobacilli against pathogens and confirms the suitability of the vaginal microbiota as a source of potential probiotic strains. The selected lactobacilli hold promise for the formulation of supplements to enhance genitourinary tract health.
Collapse
Affiliation(s)
- Alessandra Pino
- Department of Agriculture, Food and Environment (Di3A), University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy
- ProBioEtna SRL, Spin-Off of the University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy
| | - Kaisa Hiippala
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, 00014, Helsinki, Finland
| | - Aki Ronkainen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, 00014, Helsinki, Finland
| | - Amanda Vaccalluzzo
- Department of Agriculture, Food and Environment (Di3A), University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy
| | - Cinzia Caggia
- Department of Agriculture, Food and Environment (Di3A), University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy
- ProBioEtna SRL, Spin-Off of the University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy
| | - Reetta Satokari
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Haartmaninkatu 3, 00014, Helsinki, Finland
| | - Cinzia Lucia Randazzo
- Department of Agriculture, Food and Environment (Di3A), University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy.
- ProBioEtna SRL, Spin-Off of the University of Catania, Santa Sofia Street, 100, 95123, Catania, Italy.
| |
Collapse
|
7
|
Eveliina S, Halahlah A, Räikkönen H, Yousefvand A, Saris PEJ, Mikkonen KS, Ho TM. Wood hemicelluloses as protective materials for preserving the viability of probiotic Lacticaseibacillus rhamnosus GG during spray drying. Int J Biol Macromol 2024; 282:137216. [PMID: 39515729 DOI: 10.1016/j.ijbiomac.2024.137216] [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: 03/13/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Wood hemicelluloses from forest industry side-streams are promising economic and sustainable alternatives for encapsulating bioactive compounds. This study explores their suitability for probiotic encapsulation, specifically for maintaining cell viability and structure. The ability of galactoglucomannans (GGM) and glucuronoxylans (GX) to support the survival of Lacticaseibacillus rhamnosus GG (LGG) during spray drying at solid feed concentrations of 15 and 20 % and inlet air temperatures of 105 and 140 °C (outlet air temperature of 50 °C) was investigated and compared to the results obtained using maltodextrin (MD). Across all investigated conditions, LGG survival rates exceeded 85 % (>107 cfu/g) in GX and GGM microcapsule powders, which similar to that in MD microcapsules despite the differences in pH, particle size, and viscosity of their feed dispersions. The GX microcapsules demonstrated the highest process yield (50-58 %), followed by MD (39-51 %). All the microcapsule powders exhibited an amorphous structure consisting of spherical particles with an average diameter of 10 μm, sufficient for LGG accommodation. Atomic force microscopy analysis confirmed the encapsulation of LGG cells within microcapsules with intact rod-shaped chains post-spray drying. Overall, the spray-dried microencapsulation of probiotics using wood hemicelluloses maintains high probiotic viability and offers an eco-friendly, cost-effective alternative to traditional materials.
Collapse
Affiliation(s)
- Suutari Eveliina
- Department of Food and Nutrition, P.O. Box 66, FIN-00014, University of Helsinki, Finland
| | - Abedalghani Halahlah
- Department of Food and Nutrition, P.O. Box 66, FIN-00014, University of Helsinki, Finland
| | - Heikki Räikkönen
- Faculty of Pharmacy, P.O. Box 56, FIN-00014, University of Helsinki, Finland
| | - Amin Yousefvand
- Department of Microbiology, P.O. Box 56, FIN-00014, University of Helsinki, Finland; Helsinki Institute of Sustainability Science (HELSUS), P.O. Box 65, FIN-00014, University of Helsinki, Finland
| | | | - Kirsi S Mikkonen
- Department of Food and Nutrition, P.O. Box 66, FIN-00014, University of Helsinki, Finland; Helsinki Institute of Sustainability Science (HELSUS), P.O. Box 65, FIN-00014, University of Helsinki, Finland
| | - Thao M Ho
- Department of Food and Nutrition, P.O. Box 66, FIN-00014, University of Helsinki, Finland; Helsinki Institute of Sustainability Science (HELSUS), P.O. Box 65, FIN-00014, University of Helsinki, Finland.
| |
Collapse
|
8
|
Xuan H, Umar S, Zhong C, Yu W, Ahmed I, Wheatley JL, Sampath V, Chavez-Bueno S. Lactobacillus rhamnosus modulates murine neonatal gut microbiota and inflammation caused by pathogenic Escherichia coli. BMC Microbiol 2024; 24:452. [PMID: 39506682 PMCID: PMC11539828 DOI: 10.1186/s12866-024-03598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Pathogenic Escherichia coli strains produce neonatal septicemia after colonizing the neonatal gut. While the probiotic Lactobacillus rhamnosus GG (LGG) effectively reduces neonatal sepsis, LGG's effects on the neonatal intestinal microbiota alterations and inflammation triggered by E. coli are incompletely understood. We hypothesized that LGG significantly modulates the specific neonatal gut microbial populations changes and the inflammatory response elicited by the enteral introduction of septicemia-producing E. coli. To test this hypothesis, newborn rats were pretreated orally with LGG or placebo prior to infection with the neonatal E. coli septicemia clinical isolate SCB34. Amplicon 16S rRNA gene sequencing was performed on intestinal samples. Intestinal injury and expression of inflammatory mediators and apoptosis were determined. RESULTS Alpha diversity of gut microbiota was greater in SCB34-infected pups in comparison to sham-infected pups, these changes were not modified by LGG pretreatment. Beta diversity analyses also showed differences between SCB34-infected vs. uninfected pups. LGG pretreatment before SCB34 infection did not result in significant beta diversity changes compared to placebo. Moreover, individual genera and species abundance analyses by linear discriminant analysis effect size (LEfSe) showed significant changes in Gram-negative, Gram-positive, and anaerobic populations resulting from LGG pretreatment and SCB34 infection. LGG significantly suppressed the expression of inflammatory cytokines but did not attenuate SCB34-induced apoptosis or histologic injury. CONCLUSIONS LGG modulates clinically significant microbiota features and inflammation triggered by pathogenic E. coli intestinal infection shortly after birth. This new knowledge can potentially be harnessed to design novel interventions against gut-derived neonatal sepsis.
Collapse
Affiliation(s)
- Hao Xuan
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Shahid Umar
- Department of Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | - Wei Yu
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Ishfaq Ahmed
- Department of Math, Science and Computer Technology, Kansas City Kansas Community College, Kansas City, KS, USA
| | - Joshua L Wheatley
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Pediatrics, Division of Neonatology, Children's Mercy Kansas City, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA
| | - Susana Chavez-Bueno
- Department of Pediatrics, Division of Infectious Diseases, Children's Mercy Kansas City, Kansas City, MO, USA.
- University of Missouri Kansas City School of Medicine, 2401 Gillham Road, Kansas City, MO, 64108, USA.
| |
Collapse
|
9
|
Daisley BA, Allen‐Vercoe E. Microbes as medicine. Ann N Y Acad Sci 2024; 1541:63-82. [PMID: 39392836 PMCID: PMC11580781 DOI: 10.1111/nyas.15237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Over the last two decades, advancements in sequencing technologies have significantly deepened our understanding of the human microbiome's complexity, leading to increased concerns about the detrimental effects of antibiotics on these intricate microbial ecosystems. Concurrently, the rise in antimicrobial resistance has intensified the focus on how beneficial microbes can be harnessed to treat diseases and improve health and offer potentially promising alternatives to traditional antibiotic treatments. Here, we provide a comprehensive overview of both established and emerging microbe-centric therapies, from probiotics to advanced microbial ecosystem therapeutics, examine the sophisticated ways in which microbes are used medicinally, and consider their impacts on microbiome homeostasis and health outcomes through a microbial ecology lens. In addition, we explore the concept of rewilding the human microbiome by reintroducing "missing microbes" from nonindustrialized societies and personalizing microbiome modulation to fit individual microbial profiles-highlighting several promising directions for future research. Ultimately, the advancements in sequencing technologies combined with innovative microbial therapies and personalized approaches herald a new era in medicine poised to address antibiotic resistance and improve health outcomes through targeted microbiome management.
Collapse
Affiliation(s)
- Brendan A. Daisley
- Department of Molecular and Cellular BiologyUniversity of GuelphGuelphOntarioCanada
| | - Emma Allen‐Vercoe
- Department of Molecular and Cellular BiologyUniversity of GuelphGuelphOntarioCanada
| |
Collapse
|
10
|
Bijle MN, Abdalla MM, Yiu C. The effect of arginine on the growth of probiotics. J Dent 2024; 149:105272. [PMID: 39074576 DOI: 10.1016/j.jdent.2024.105272] [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: 05/17/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVE(S) The study objective was to examine the effect of arginine (Arg) supplementation on the growth of probiotics. METHODS Lacticaseibacillus rhamnosus GG, Lactiplantibacillus plantarum, and Lactobacillus acidophilus were identified as potential probiotics. L. rhamnosus GG and L. plantarum were selected for further experimentation. The probiotics were co-treated with 0.9 % NaCl (negative control), 0.5 % Arg, and 1.0 % Arg in a 1:1 ratio for 24 h at 5 % CO2, 37 °C. The probiotics were tested for growth profiles, spectroscopic turbidity assay, metabolic assays (XTT and WST-8), live/dead cell assessment using confocal laser scanning microscopy (CLSM), and colony forming units (CFU). RESULTS The growth profiles of L. rhamnosus GG and L. plantarum were found to be similar, whereas L. acidophilus showed minimal or no transition from the initial lag phase. In the turbidity assay, the end-point absorbance for L. rhamnosus GG with 1.0 % Arg was significantly lower than 0.9 % NaCl and 0.5 % Arg (p < 0.05). For metabolic assays and CFU, increasing concentrations of Arg increased the viable cells for L. rhamnosus GG (p < 0.05), but decreased viability for L. plantarum (p < 0.05). Metabolic assays with dual-species bacterial suspensions indicated that Arg co-treatment inhibited viable proportions compared to control (p < 0.05). The dead cell proportion was significantly lower than live cell proportion for all tested interventions and probiotics (p < 0.05). CONCLUSION Increasing concentrations of Arg promote the growth of L. rhamnosus GG, while conversely inhibiting the growth of L. plantarum. Therefore, the effect of prebiotic Arg on probiotics is concentration-dependent, leading to a selective promotion or inhibition of growth. CLINICAL SIGNIFICANCE The present study results show that Arg supplementation can selectively enhance the growth of L. rhamnosus GG while inhibit the growth of L. plantarum. This underscores the need to consider strain-specific responses in probiotic formulations when developing Arg-based synbiotics for modulating biofilms and creating ecologically homeostatic biofilm microenvironments.
Collapse
Affiliation(s)
| | - Mohamed Mahmoud Abdalla
- Faculty of Dentistry, The University of Hong Kong, Hong Kong; Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt
| | - Cynthia Yiu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
11
|
Cui Y, Liu Y, Yang J, Duan H, Wang P, Guo L, Guo Y, Li S, Zhao Y, Wang J, Qi G, Guan J. Microencapsulated Lactobacillus plantarum promotes intestinal development through gut colonization of layer chicks. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:1-16. [PMID: 38989011 PMCID: PMC11231655 DOI: 10.1016/j.aninu.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 01/29/2024] [Accepted: 03/12/2024] [Indexed: 07/12/2024]
Abstract
The effects of Lactobacillus plantarum in microencapsulation (LPM) on intestinal development in layer chicks were investigated in this study, as well as the colonization of L. plantarum in the gut. A total of 480 healthy Hy-Line Brown layer chicks at 0 d old were randomly divided into 4 groups (8 replicates each treatment), and the diets of these birds were supplemented with nothing (control), L. plantarum (0.02 g/kg feed; 109 CFU/kg feed), LPM (1.0 g/kg feed; 109 CFU/kg feed) and wall material of LPM (WM; 0.98 g/kg feed), respectively. Compared to control, LPM improved growth performance and intestinal development of layer chicks, evidenced by significantly increased body weight, average daily gain, average daily feed intake, villus height, villus height/crypt depth, as well as weight and length of the duodenum, jejunum and ileum (P < 0.05). These results could be attributed to the increased colonization of L. plantarum in the gut, which was verified by significant increases in lactic acid content, viable counts in chyme and mucosa (P < 0.05), as well as a visible rise in number of strains labeled with fluorescein isothiocyanate. Meanwhile, the relative abundances of Lactobacillus and Bifidobacterium significantly increased in response to microencapsulated L. plantarum supplementation (P < 0.05), accompanied by the significant up-regulation of colonization related genes (P < 0.05), encoding solute carrier family, monocarboxylate transporter, activin A receptor, succinate receptor and secretogranin II. To sum up, microencapsulated L. plantarum supplementation promoted intestinal development, which could be attributed to the enhancement of L. plantarum colonization in the intestine through the mutual assistance of Bifidobacterium and interactions with colonization related transmembrane proteins.
Collapse
Affiliation(s)
- Yaoming Cui
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Yanxia Liu
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Jing Yang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Haitao Duan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan 450046, China
| | - Peng Wang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Linna Guo
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Yanjiao Guo
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Suying Li
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Yating Zhao
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Jinrong Wang
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| | - Guanghai Qi
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Junjun Guan
- School of Biological Engineering, Henan University of Technology, Zhengzhou, Henan 450001, China
| |
Collapse
|
12
|
Xiao L, Zhao X, Lin L, Mahsa GC, Ma K, Zhang C, Rui X, Li W. Contribution of Surface Adhesins of Lacticaseibacillus paracasei S-NB to Its Intestinal Adhesion and Colonization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18986-19002. [PMID: 39140151 DOI: 10.1021/acs.jafc.4c04256] [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: 08/15/2024]
Abstract
The intestinal retention and persistence of lactic acid bacteria (LAB) are strain-specific and affected by the bacterial surface components. However, the contribution of surface adhesins of LAB to intestinal adhesion and colonization remains unclear. In the present study, seven gene knockout mutants (genes related to surface adhesin synthesis) of Lacticaseibacillus paracasei S-NB were derived based on the Cre-lox-based recombination system. Results showed that the capsule layer appeared thinner in the cell wall of S-NBΔ7576, S-NBΔdlt, and S-NBΔsrtA mutants when compared with the wild-type (WT) S-NB. The effects of S-NB_7576 (wzd and wze genes, responsible for capsular polysaccharide synthesis) and S-NB_srtA (sortase A) mutation on the hydrophobicity, surface charge, and adhesion ability seem to vary strongly among seven mutant strains. In vivo colonization experiments showed a decrease in the colonization numbers of S-NBΔ7576 and S-NBΔsrtA in both the ileal and colon lumen from 2 to 8 days when compared with those of the WT S-NB. In conclusion, the synthesis of capsular polysaccharides and the transport of surface proteins are closely related to the adhesion ability and intestinal colonization of L. paracasei S-NB.
Collapse
Affiliation(s)
- Luyao Xiao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Xiaogan Zhao
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Long Lin
- Key Laboratory of Biological Interactions and Crop Health, College of Plant Protection, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Ghahvechi Chaeipeima Mahsa
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Kai Ma
- Jiangsu New-Bio Biotechnology Co., Ltd., Jiangyin, Jiangsu 214400, PR China
- Jiangsu Biodep Biotechnology Co., Ltd., Jiangyin, Jiangsu 214400, PR China
| | - Changliang Zhang
- Jiangsu New-Bio Biotechnology Co., Ltd., Jiangyin, Jiangsu 214400, PR China
- Jiangsu Biodep Biotechnology Co., Ltd., Jiangyin, Jiangsu 214400, PR China
| | - Xin Rui
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| | - Wei Li
- Sanya Institute of Nanjing Agricultural University, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, PR China
| |
Collapse
|
13
|
Zhou Y, Zhang D, Cheng H, Wu J, Liu J, Feng W, Peng C. Repairing gut barrier by traditional Chinese medicine: roles of gut microbiota. Front Cell Infect Microbiol 2024; 14:1389925. [PMID: 39027133 PMCID: PMC11254640 DOI: 10.3389/fcimb.2024.1389925] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.
Collapse
Affiliation(s)
- Yaochuan Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Prajapati A, Palva A, von Ossowski I, Krishnan V. The crystal structure of the N-terminal domain of the backbone pilin LrpA reveals a new closure-and-twist motion for assembling dynamic pili in Ligilactobacillus ruminis. Acta Crystallogr D Struct Biol 2024; 80:474-492. [PMID: 38935340 DOI: 10.1107/s2059798324005114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Sortase-dependent pili are long surface appendages that mediate attachment, colonization and biofilm formation in certain genera and species of Gram-positive bacteria. Ligilactobacillus ruminis is an autochthonous gut commensal that relies on sortase-dependent LrpCBA pili for host adherence and persistence. X-ray crystal structure snapshots of the backbone pilin LrpA were captured in two atypical bent conformations leading to a zigzag morphology in the LrpCBA pilus structure. Small-angle X-ray scattering and structural analysis revealed that LrpA also adopts the typical linear conformation, resulting in an elongated pilus morphology. Various conformational analyses and biophysical experiments helped to demonstrate that a hinge region located at the end of the flexible N-terminal domain of LrpA facilitates a new closure-and-twist motion for assembling dynamic pili during the assembly process and host attachment. Further, the incongruent combination of flexible domain-driven conformational dynamics and rigid isopeptide bond-driven stability observed in the LrpCBA pilus might also extend to the sortase-dependent pili of other bacteria colonizing a host.
Collapse
Affiliation(s)
- Amar Prajapati
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, NCR, Biotech Science Cluster, Faridabad 121 001, India
| | - Airi Palva
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | | | - Vengadesan Krishnan
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, NCR, Biotech Science Cluster, Faridabad 121 001, India
| |
Collapse
|
15
|
Wonglapsuwan M, Pahumunto N, Teanpaisan R, Surachat K. Unlocking the genetic potential of Lacticaseibacillus rhamnosus strains: Medical applications of a promising probiotic for human and animal health. Heliyon 2024; 10:e29499. [PMID: 38655288 PMCID: PMC11035056 DOI: 10.1016/j.heliyon.2024.e29499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Lacticaseibacillus rhamnosus is a group of probiotic strains that have gained popularity for their potential health benefits such as promoting digestive health, boosting the immune system, improving lactose digestion, preventing and treating antibiotic-associated diarrhea, reducing the severity and duration of certain infections, and preventing the formation of dental plaque. In particular, L. rhamnosus strains SD4 and SD11 have promising human and animal health applications due to their ability to inhibit the growth of harmful pathogens. This study presents an in silico genomic analysis of L. rhamnosus strains SD4 and SD11. We analyzed draft genomes and conducted comparative genome analyses against several other probiotic strains, aiming to gain insights into the genomes of the two strains and to compare them to related strains isolated from other sources. We also aimed to clarify the functional mechanisms and adaptation of these strains to specific environments. Comprehensive insights into the genomes of L. rhamnosus SD4 and SD11 could enhance our understanding of their capacity to colonize, adapt, and exhibit probiotic properties after administration. This study holds significance in advancing our understanding of the potential health benefits associated with these strains and in elucidating the underlying mechanisms responsible for their effectiveness in humans and animals.
Collapse
Affiliation(s)
- Monwadee Wonglapsuwan
- Division of Biological Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Nuntiya Pahumunto
- Research Center of Excellence for Oral Health, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
- Department of Oral Diagnostic Sciences, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Rawee Teanpaisan
- Medical Science Research and Innovation Institute, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Komwit Surachat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| |
Collapse
|
16
|
Leser T, Baker A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG ® Probiotic Function. Microorganisms 2024; 12:794. [PMID: 38674738 PMCID: PMC11051730 DOI: 10.3390/microorganisms12040794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
To advance probiotic research, a comprehensive understanding of bacterial interactions with human physiology at the molecular and cellular levels is fundamental. Lacticaseibacillus rhamnosus LGG® is a bacterial strain that has long been recognized for its beneficial effects on human health. Probiotic effector molecules derived from LGG®, including secreted proteins, surface-anchored proteins, polysaccharides, and lipoteichoic acids, which interact with host physiological processes have been identified. In vitro and animal studies have revealed that specific LGG® effector molecules stimulate epithelial cell survival, preserve intestinal barrier integrity, reduce oxidative stress, mitigate excessive mucosal inflammation, enhance IgA secretion, and provide long-term protection through epigenetic imprinting. Pili on the cell surface of LGG® promote adhesion to the intestinal mucosa and ensure close contact to host cells. Extracellular vesicles produced by LGG® recapitulate many of these effects through their cargo of effector molecules. Collectively, the effector molecules of LGG® exert a significant influence on both the gut mucosa and immune system, which promotes intestinal homeostasis and immune tolerance.
Collapse
Affiliation(s)
- Thomas Leser
- Future Labs, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark;
| | | |
Collapse
|
17
|
Zhang Z, Zhang HL, Yang DH, Hao Q, Yang HW, Meng DL, Meindert de Vos W, Guan LL, Liu SB, Teame T, Gao CC, Ran C, Yang YL, Yao YY, Ding QW, Zhou ZG. Lactobacillus rhamnosus GG triggers intestinal epithelium injury in zebrafish revealing host dependent beneficial effects. IMETA 2024; 3:e181. [PMID: 38882496 PMCID: PMC11170971 DOI: 10.1002/imt2.181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 06/18/2024]
Abstract
Lactobacillus rhamnosus GG (LGG), the well-characterized human-derived probiotic strain, possesses excellent properties in the maintenance of intestinal homeostasis, immunoregulation and defense against gastrointestinal pathogens in mammals. Here, we demonstrate that the SpaC pilin of LGG causes intestinal epithelium injury by inducing cell pyroptosis and gut microbial dysbiosis in zebrafish. Dietary SpaC activates Caspase-3-GSDMEa pathways in the intestinal epithelium, promotes intestinal pyroptosis and increases lipopolysaccharide (LPS)-producing gut microbes in zebrafish. The increased LPS subsequently activates Gaspy2-GSDMEb pyroptosis pathway. Further analysis reveals the Caspase-3-GSDMEa pyroptosis is initiated by the species-specific recognition of SpaC by TLR4ba, which accounts for the species-specificity of the SpaC-inducing intestinal pyroptosis in zebrafish. The observed pyroptosis-driven gut injury and microbial dysbiosis by LGG in zebrafish suggest that host-specific beneficial/harmful mechanisms are critical safety issues when applying probiotics derived from other host species and need more attention.
Collapse
Affiliation(s)
- Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
- Faculty of Land and Food Systems The University of British Columbia Vancouver Canada
| | - Hong-Ling Zhang
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Da-Hai Yang
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai China
| | - Qiang Hao
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Hong-Wei Yang
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - De-Long Meng
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Willem Meindert de Vos
- Laboratory of Microbiology Wageningen University and Research Wageningen Netherlands
- Human Microbiome Research Program, Faculty of Medicine University of Helsinki Helsinki Finland
| | - Le-Luo Guan
- Faculty of Land and Food Systems The University of British Columbia Vancouver Canada
| | - Shu-Bin Liu
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Tsegay Teame
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
- Tigray Agricultural Research Institute Mekelle Ethiopia
| | - Chen-Chen Gao
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Ya-Lin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Yuan-Yuan Yao
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Qian-Wen Ding
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| | - Zhi-Gang Zhou
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research Chinese Academy of Agricultural Sciences Beijing China
| |
Collapse
|
18
|
Adnane M, Whiston R, Tasara T, Bleul U, Chapwanya A. Harnessing Vaginal Probiotics for Enhanced Management of Uterine Disease and Reproductive Performance in Dairy Cows: A Conceptual Review. Animals (Basel) 2024; 14:1073. [PMID: 38612312 PMCID: PMC11011061 DOI: 10.3390/ani14071073] [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: 03/12/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
Uterine disease in cattle impairs reproductive performance and profitability and increases antibiotic use and antimicrobial resistance. Thus, probiotics offer a promising alternative therapy. This review presents conceptual findings on the efficacy of probiotics in managing uterine diseases and fertility in cows. Probiotics containing Lactobacillus spp. and Bifidobacterium spp. individually or as composite formulations are known to improve fertility. Strategic intravaginal administration of these formulations would likely enhance uterine immunity, particularly during the postpartum period. While current findings on the benefits to uterine health are encouraging, there is still significant knowledge missing, including a lack of empirical information from large-scale field trials. This review underscores the need for evidence-based guidelines for probiotics, such as genomic selection of formulations, targeted delivery, or potential synergy with other interventions. Future research should address these gaps to maximize the potential of probiotics in managing uterine diseases and enhancing the reproductive health of dairy cattle.
Collapse
Affiliation(s)
- Mounir Adnane
- Department of Biomedicine, Institute of Veterinary Sciences, University Ibn Khaldoun of Tiaret, Tiaret 14000, Algeria
- USDA, Faculty Exchange Program Fellow, University of Georgia, Athens, GA 30602, USA
| | - Ronan Whiston
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre 00265, Saint Kitts and Nevis; (R.W.); (A.C.)
| | - Taurai Tasara
- Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland;
| | - Ulrich Bleul
- Department of Farm Animals, Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland;
| | - Aspinas Chapwanya
- Department of Clinical Sciences, Ross University School of Veterinary Medicine, Basseterre 00265, Saint Kitts and Nevis; (R.W.); (A.C.)
| |
Collapse
|
19
|
Wang W, Zhang L, Zhang Y, Liu X, Song A, Ren J, Qu X. A Self-Adaptive Pyroptosis Inducer: Optimizing the Catalytic Microenvironment of Nanozymes by Membrane-Adhered Microbe Enables Potent Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310063. [PMID: 38153294 DOI: 10.1002/adma.202310063] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Indexed: 12/29/2023]
Abstract
Pyroptosis has garnered increasing attention in cancer immunotherapy. Moreover, increasing plasma membrane damage by reactive oxygen species (ROS) is considered an effective strategy for promoting pyroptosis. However, the current tactics for enhancing membrane rupture in pyroptosis are limited by the inherent drawbacks of ROS and the immunosuppressive tumor microenvironment. Herein, a self-adaptive pyroptosis inducer (LPZ) is designed by integrating Lactobacillus rhamnosus GG (LGG) and an enzyme-like metal-organic framework to achieve potent pyroptosis immunotherapy. LPZ can adhere to cancer cell membranes through the interaction between the pili of LGG and the mucin of cancer cells. In particular, the adaptive formula can gradually enhance the ability of nanozymes to produce ROS by creating an acidic microenvironment through anaerobic respiration. These results verify that LPZ could generate high levels of ROS both on the membrane and within cancer cells, leading to pyroptotic cell death and strong antitumor immunity. Meanwhile, LGG are eventually killed by ROS in this process to halt their respiration and prevent potential biosafety concerns. Overall, this work provides new inspiration for the design of self-adaptive nanocatalytic drugs for cancer immunotherapy.
Collapse
Affiliation(s)
- Wenjie Wang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Yanjie Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xuemeng Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Anjun Song
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
20
|
Matsuzaki C, Takagi H, Saiga S, Kinoshita Y, Yamaguchi M, Higashimura Y, Yamamoto K, Yamaguchi M. Prebiotic effect of galacto- N-biose on the intestinal lactic acid bacteria as enhancer of acetate production and hypothetical colonization. Appl Environ Microbiol 2024; 90:e0144523. [PMID: 38411084 PMCID: PMC10952502 DOI: 10.1128/aem.01445-23] [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: 08/22/2023] [Accepted: 01/28/2024] [Indexed: 02/28/2024] Open
Abstract
Galacto-N-biose (GNB) is an important core structure of glycan of mucin glycoproteins in the gastrointestinal (GI) mucosa. Because certain beneficial bacteria inhabiting the GI tract, such as bifidobacteria and lactic acid bacteria, harbor highly specialized GNB metabolic capabilities, GNB is considered a promising prebiotic for nourishing and manipulating beneficial bacteria in the GI tract. However, the precise interactions between GNB and beneficial bacteria and their accompanying health-promoting effects remain elusive. First, we evaluated the proliferative tendency of beneficial bacteria and their production of beneficial metabolites using gut bacterial strains. By comparing the use of GNB, glucose, and inulin as carbon sources, we found that GNB enhanced acetate production in Lacticaseibacillus casei, Lacticaseibacillus rhamnosus, Lactobacillus gasseri, and Lactobacillus johnsonii. The ability of GNB to promote acetate production was also confirmed by RNA-seq analysis, which indicated the upregulation of gene clusters that catalyze the deacetylation of N-acetylgalactosamine-6P and biosynthesize acetyl-CoA from pyruvate, both of which result in acetate production. To explore the in vivo effect of GNB in promoting acetate production, antibiotic-treated BALB/cA mice were administered with GNB with L. rhamnosus, resulting in a fecal acetate content that was 2.7-fold higher than that in mice administered with only L. rhamnosus. Moreover, 2 days after the last administration, a 3.7-fold higher amount of L. rhamnosus was detected in feces administered with GNB with L. rhamnosus than in feces administered with only L. rhamnosus. These findings strongly suggest the prebiotic potential of GNB in enhancing L. rhamnosus colonization and converting L. rhamnosus into higher acetate producers in the GI tract. IMPORTANCE Specific members of lactic acid bacteria, which are commonly used as probiotics, possess therapeutic properties that are vital for human health enhancement by producing immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. The long residence time of probiotic lactic acid bacteria in the GI tract prolongs their beneficial health effects. Moreover, the colonization property is also desirable for the application of probiotics in mucosal vaccination to provoke a local immune response. In this study, we found that GNB could enhance the beneficial properties of intestinal lactic acid bacteria that inhabit the human GI tract, stimulating acetate production and promoting intestinal colonization. Our findings provide a rationale for the addition of GNB to lactic acid bacteria-based functional foods. This has also led to the development of therapeutics supported by more rational prebiotic and probiotic selection, leading to an improved healthy lifestyle for humans.
Collapse
Affiliation(s)
- Chiaki Matsuzaki
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Hiroki Takagi
- Department of Production Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Sorachi Saiga
- Department of Production Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Yuun Kinoshita
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Misako Yamaguchi
- Department of Organic Bio Chemistry, Faculty of Education, Wakayama University, Wakayama, Japan
| | - Yasuki Higashimura
- Department of Food Science, Ishikawa Prefectural University, Nonoichi, Ishikawa, Japan
| | - Kenji Yamamoto
- Center for Innovative and Joint Research, Wakayama University, Wakayama, Japan
| | - Masanori Yamaguchi
- Department of Organic Bio Chemistry, Faculty of Education, Wakayama University, Wakayama, Japan
| |
Collapse
|
21
|
Quintieri L, Fanelli F, Monaci L, Fusco V. Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides. Foods 2024; 13:601. [PMID: 38397577 PMCID: PMC10888271 DOI: 10.3390/foods13040601] [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: 12/21/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Milk is a source of many valuable nutrients, including minerals, vitamins and proteins, with an important role in adult health. Milk and dairy products naturally containing or with added probiotics have healthy functional food properties. Indeed, probiotic microorganisms, which beneficially affect the host by improving the intestinal microbial balance, are recognized to affect the immune response and other important biological functions. In addition to macronutrients and micronutrients, biologically active peptides (BPAs) have been identified within the amino acid sequences of native milk proteins; hydrolytic reactions, such as those catalyzed by digestive enzymes, result in their release. BPAs directly influence numerous biological pathways evoking behavioral, gastrointestinal, hormonal, immunological, neurological, and nutritional responses. The addition of BPAs to food products or application in drug development could improve consumer health and provide therapeutic strategies for the treatment or prevention of diseases. Herein, we review the scientific literature on probiotics, BPAs in milk and dairy products, with special attention to milk from minor species (buffalo, sheep, camel, yak, donkey, etc.); safety assessment will be also taken into consideration. Finally, recent advances in foodomics to unveil the probiotic role in human health and discover novel active peptide sequences will also be provided.
Collapse
Affiliation(s)
| | - Francesca Fanelli
- National Research Council of Italy, Institute of Sciences of Food Production (CNR-ISPA), 70126 Bari, Italy; (L.Q.); (L.M.); (V.F.)
| | | | | |
Collapse
|
22
|
Marole TA, Sibanda T, Buys EM. Assessing probiotic viability in mixed species yogurt using a novel propidium monoazide (PMAxx)-quantitative PCR method. Front Microbiol 2024; 15:1325268. [PMID: 38389538 PMCID: PMC10882272 DOI: 10.3389/fmicb.2024.1325268] [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: 10/20/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Viability is a prerequisite for any therapeutic benefits associated with the ingestion of probiotic bacteria. Current culture-based techniques are inadequate for the enumeration of probiotics in mixed-species food products. This study utilized a quantitative PCR (qPCR) method coupled with propidium monoazide (PMAxx), and novel species-specific tuf gene primers to selectively enumerate Lacticaseibacillus rhamnosus, Bifidobacterium spp., and yogurt starter cultures in mixed-species probiotic yogurt. The method was optimized for PMAxx concentration and specificity and evaluated for efficiency and applicability. PMAxx-qPCR showed high specificity to the target organisms in mixed-species yogurt, quantifying only viable cells. The linear dynamic ranges were established over five to seven orders of magnitude. The assay was reliable with an efficiency of 91-99%, R2 values > 0.99, and a good correlation to the plate count method (r = 0.882). The results of this study demonstrate the high selectivity, improved lead time, and reliability of PMAxx-qPCR over the culture-dependent method, making it a valuable tool for inline viability verification during processing and improving probiotic quality assurance for processors and consumers.
Collapse
Affiliation(s)
- Tlaleo A Marole
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria, South Africa
| | - Thulani Sibanda
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria, South Africa
| | - Elna M Buys
- Department of Consumer and Food Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
23
|
Buljubašić E, Bambace MF, Christensen MHL, Ng K, Huertas‐Díaz L, Sundekilde U, Marietou A, Schwab C. Novel Lactobacillaceae strains and consortia to produce propionate-containing fermentates as biopreservatives. Microb Biotechnol 2024; 17:e14392. [PMID: 38380951 PMCID: PMC10880516 DOI: 10.1111/1751-7915.14392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/30/2023] [Accepted: 12/04/2023] [Indexed: 02/22/2024] Open
Abstract
Biopreservation refers to the use of natural or controlled microbial single strains or consortia, and/or their metabolites such as short-chain carboxylic acids (SCCA), to improve the shelf-life of foods. This study aimed at establishing a novel Lactobacillaceae-driven bioprocess that led to the production of the SCCA propionate through the cross-feeding on 1,2-propanediol (1,2-PD) derived from the deoxyhexoses rhamnose or fucose. When grown as single cultures in Hungate tubes, strains of Lacticaseibacillus rhamnosus preferred fucose over rhamnose and produced 1,2-PD in addition to lactate, acetate, and formate, while Limosilactobacillus reuteri metabolized 1,2-PD into propionate, propanol and propanal. Loigolactobacillus coryniformis used fucose to produce 1,2-PD and only formed propionate when supplied with 1,2-PD. Fermentates collected from batch fermentations in bioreactor using two-strain consortia (L. rhamnosus and L. reuteri) or fed-batch fermentations of single strain cultures of L. coryniformis with rhamnose contained mixtures of SCCA consisting of mainly lactate and acetate and also propionate. Synthetic mixtures that contained SCCA at concentrations present in the fermentates were more antimicrobial against Salmonella enterica if propionate was present. Together, this study (i) demonstrates the potential of single strains and two-strain consortia to produce propionate in the presence of deoxyhexoses extending the fermentation metabolite profile of Lactobacillaceae, and (ii) emphasizes the potential of applying propionate-containing fermentates as biopreservatives.
Collapse
Affiliation(s)
- Ena Buljubašić
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | | | | | - Ker‐Sin Ng
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Lucía Huertas‐Díaz
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | | | - Angeliki Marietou
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| |
Collapse
|
24
|
Chen JF, Hsia KC, Kuo YW, Chen SH, Huang YY, Li CM, Hsu YC, Tsai SY, Ho HH. Safety Assessment and Probiotic Potential Comparison of Bifidobacterium longum subsp. infantis BLI-02, Lactobacillus plantarum LPL28, Lactobacillus acidophilus TYCA06, and Lactobacillus paracasei ET-66. Nutrients 2023; 16:126. [PMID: 38201957 PMCID: PMC10780348 DOI: 10.3390/nu16010126] [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: 10/27/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Bifidobacterium longum subsp. infantis BLI-02, Lactobacillus paracasei ET-66, Lactobacillus plantarum LPL28, and Lactobacillus acidophilus TYCA06, isolated from healthy breast milk, miso, and the healthy human gut, were assessed for safety in this study. BLI-02, LPL28, TYCA06, and ET-66 exhibited no antibiotic resistance and mutagenic activity in the Ames test at the highest dosage (5000 μg/plate). No genotoxicity was observed in micronucleus and chromosomal aberration assays in rodent spermatogonia at the maximum dosage of 10 g/kg body weight (BW). No acute and sub-chronic toxicity occurred in mice and rats at the maximum tested dosage of 10 g/kg BW and 1.5 g/kg BW, respectively. The lyophilized powder of these strains survived a low pH and high bile salt environment, adhering strongly to Caco-2 cells. Unique antimicrobial activities were noted in these strains, with BLI-02 demonstrating the best growth inhibition against Vibrio parahaemolyticus, LPL28 exhibiting the best growth inhibition against Helicobacter pylori, and ET-66 showing the best growth inhibition against Aggregatibacter actinomycetemcomitans. Based on the present study, the lyophilized powder of these four strains appears to be a safe probiotic supplement at tested dosages. It should be applicable for clinical or healthcare applications.
Collapse
Affiliation(s)
- Jui-Fen Chen
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Ko-Chiang Hsia
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Yi-Wei Kuo
- Functional Investigation Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan;
| | - Shu-Hui Chen
- Process Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan;
| | - Yen-Yu Huang
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Ching-Min Li
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Yu-Chieh Hsu
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Shin-Yu Tsai
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
| | - Hsieh-Hsun Ho
- Research Product Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan; (J.-F.C.); (K.-C.H.); (Y.-Y.H.); (C.-M.L.); (Y.-C.H.); (S.-Y.T.)
- Functional Investigation Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan;
- Process Department, R&D Center, Glac Biotech Co., Ltd., Tainan City 744, Taiwan;
| |
Collapse
|
25
|
Walsh C, Owens RA, Bottacini F, Lane JA, van Sinderen D, Hickey RM. HMO-primed bifidobacteria exhibit enhanced ability to adhere to intestinal epithelial cells. Front Microbiol 2023; 14:1232173. [PMID: 38163079 PMCID: PMC10757668 DOI: 10.3389/fmicb.2023.1232173] [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: 05/31/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024] Open
Abstract
The ability of gut commensals to adhere to the intestinal epithelium can play a key role in influencing the composition of the gut microbiota. Bifidobacteria are associated with a multitude of health benefits and are one of the most widely used probiotics for humans. Enhanced bifidobacterial adhesion may increase host-microbe, microbe-nutrient, and/or microbe-microbe interactions, thereby enabling consolidated health benefits to the host. The objective of this study was to determine the ability of human milk oligosaccharides (HMOs) to enhance bifidobacterial intestinal adhesion in vitro. This study assessed the colonisation-promoting effects of HMOs on four commercial infant-associated Bifidobacterium strains (two B. longum subsp. infantis strains, B. breve and B. bifidum). HT29-MTX cells were used as an in vitro intestinal model for bacterial adhesion. Short-term exposure of four commercial infant-associated Bifidobacterium strains to HMOs derived from breastmilk substantially increased the adherence (up to 47%) of these probiotic strains. Interestingly, when strains were incubated with HMOs as a four-strain combination, the number of viable bacteria adhering to intestinal cells increased by >90%. Proteomic analysis of this multi-strain bifidobacterial mixture revealed that the increased adherence resulting from exposure to HMOs was associated with notable increases in the abundance of sortase-dependent pili and glycosyl hydrolases matched to Bifidobacterium bifidum. This study suggests that HMOs may prime infant gut-associated Bifidobacterium for colonisation to intestinal epithelial cells by influencing the expression of various colonization factors.
Collapse
Affiliation(s)
- Clodagh Walsh
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- Health and Happiness Group, H&H Research, Cork, Ireland
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| | | | - Francesca Bottacini
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
- Biological Sciences and ADAPT Research Centre, Munster Technological University, Cork, Ireland
| | | | - Douwe van Sinderen
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| | - Rita M. Hickey
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
26
|
Chung Y, Kang SB, Son D, Lee JY, Chung MJ, Lim S. Characterization of the probiotic properties of Lacticaseibacillus rhamnosus LR6 isolated from the vaginas of healthy Korean women against vaginal pathogens. Front Microbiol 2023; 14:1308293. [PMID: 38098667 PMCID: PMC10720895 DOI: 10.3389/fmicb.2023.1308293] [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: 10/11/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
The human microbiome exhibits intricate populations across the body, with the vaginal tract serving as an ecosystem characterized by the prevalence of the genus Lactobacillus. Disruptions in the vaginal microbiota, which are frequently linked to variables such as sexual activity, hormonal fluctuations, and excessive use of antibiotics, can result in vaginal dysbiosis and the development of diseases such as bacterial vaginosis (BV) and candidiasis. Lactobacillus species, owing to their capacity to create an acidic environment through the production of lactic acid, have a key function within this complex microbial community: they inhibit the growth of harmful microorganisms. This study aimed to investigate the genomic characteristics of L. rhamnosus LR6, a newly discovered strain isolated from the vaginal microbiota of 20 healthy women to assess its potential as a vaginal probiotic. We performed a comparative investigation of the genetic traits of L. rhamnosus using 45 publicly available genomes from various sources. We evaluated the genetic characteristics related to carbohydrate utilization, adhesion to host cells, and the presence of bacteriocin clusters. A comprehensive study was conducted by integrating in silico evaluations with experimental techniques to authenticate the physiological characteristics of strain LR6. We further used a rat model to assess the impact of L. rhamnosus LR6 administration on the changes in the gastrointestinal tract and the vaginal microbiome. The assessments revealed a significantly high inhibitory activity against pathogens, enhanced adherence to host cells, and high lactic acid production. Rat experiments revealed changes in both the fecal and vaginal microbiota; in treated rats, Firmicutes increased in both; Lactobacillaceae increased in the fecal samples; and Enterobacteriaceae decreased but Enterococcaceae, Streptococcaceae, and Morganellaceae increased in the vaginal samples. The study results provide evidence of the genetic characteristics and probiotic properties of LR6, and suggest that oral administration of L. rhamnosus LR6 can alter both gut and vaginal microbiome. Collectively, these findings establish L. rhamnosus LR6 as a highly promising candidate for improving vaginal health.
Collapse
Affiliation(s)
- Yusook Chung
- R&D Center, Cell Biotech, Co., Ltd., Gimpo-si, Republic of Korea
| | - Seung Beom Kang
- R&D Center, Cell Biotech, Co., Ltd., Gimpo-si, Republic of Korea
| | - Dooheon Son
- R&D Center, Cell Biotech, Co., Ltd., Gimpo-si, Republic of Korea
| | - Ji Young Lee
- Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Myung Jun Chung
- R&D Center, Cell Biotech, Co., Ltd., Gimpo-si, Republic of Korea
| | - Sanghyun Lim
- R&D Center, Cell Biotech, Co., Ltd., Gimpo-si, Republic of Korea
| |
Collapse
|
27
|
Gizachew S, Van Beeck W, Spacova I, Dekeukeleire M, Alemu A, Mihret W, Lebeer S, Engidawork E. Characterization of potential probiotic starter cultures of lactic acid bacteria isolated from Ethiopian fermented cereal beverages, Naaqe, and Cheka. J Appl Microbiol 2023; 134:lxad237. [PMID: 37858306 DOI: 10.1093/jambio/lxad237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/21/2023] [Accepted: 10/18/2023] [Indexed: 10/21/2023]
Abstract
AIMS To test the in vitro probiotic potential and starter culture capacity of lactic acid bacteria (LAB) isolated from Naaqe and Cheka, cereal-based Ethiopian traditional fermented beverages. METHODS AND RESULTS A total of 44 strains were isolated from spontaneously fermented Ethiopian cereal-based beverages, Naaqe and Cheka with 24 putatively identified as LAB and 14 identified up to the species level. The species Limosilactobacillus fermentum (6/12; 50%) and Weissella confusa (5/12, 41.67%) were the predominant species identified from Naaqe, while the two Cheka isolates were L. fermentum and Pediococcus pentosaceus. Six LAB strains inhibited eight of the nine gastrointestinal indicator key pathogens in Ethiopia, including Escherichia coli, Salmonella enterica subsp. enterica var. Typhimurium, Staphylococcus aureus, Shigella flexneri, and Listeria monocytogenes. Three of the LAB isolates exhibited strain-specific immunostimulation in human monocytes. Based on these probiotic properties and growth, six strains were selected for in situ evaluation in a mock fermentation of Naaqe and Cheka. During primary fermentations, L. fermentum 73B, P. pentosaceus 74D, L. fermentum 44B, W. confusa 44D, L. fermentum 82C, and Weissella cibaria 83E and their combinations demonstrated higher pH-lowering properties and colony-forming unit counts compared to the control spontaneous fermentation. The same pattern was also observed in the secondary mock fermentation by the Naaqe LAB isolates. CONCLUSIONS In this study, we selected six LAB strains with antipathogenic, immunostimulatory, and starter culture potentials that can be used as autochthonous probiotic starters for Naaqe and Cheka fermentations once their health benefit is ascertained in a clinical trial as a next step.
Collapse
Affiliation(s)
- Seyoum Gizachew
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
- Department of Bioscience Engineering, Faculty of Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Wannes Van Beeck
- Department of Bioscience Engineering, Faculty of Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Irina Spacova
- Department of Bioscience Engineering, Faculty of Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Max Dekeukeleire
- Department of Bioscience Engineering, Faculty of Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Ashenafi Alemu
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Wude Mihret
- Bacterial and Viral Diseases Research Directorate, Armauer Hansen Research Institute, P.O. Box 1005, Addis Ababa, Ethiopia
| | - Sarah Lebeer
- Department of Bioscience Engineering, Faculty of Sciences, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | - Ephrem Engidawork
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia
| |
Collapse
|
28
|
You L, Lv R, Jin H, Ma T, Zhao Z, Kwok LY, Sun Z. A large-scale comparative genomics study reveals niche-driven and within-sample intra-species functional diversification in Lacticaseibacillus rhamnosus. Food Res Int 2023; 173:113446. [PMID: 37803772 DOI: 10.1016/j.foodres.2023.113446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 10/08/2023]
Abstract
Lacticaseibacillus rhamnosus (L. rhamnosus) is widely recognized as a probiotic species, and it exists in a variety of environments including host gut and dairy products. This work aimed at conducting a large-scale comparative genomics analysis of 384 L. rhamnosus genomes (257 whole-sequence or metagenomic-assembled genomes from gut-associated isolates [122 and 135 retrieved from the UHGG and NCBI databases, respectively] and 127 genomes from dairy isolates [34 from the NCBI database; 93 isolated from a cheese sample and sequenced here]). Our results showed that L. rhamnosus had a large and open pan-genome (15,253 pan-genes identified from all 384 genomes; 15,028 pan-genes if the 93 cheese-originated isolates were excluded). The core-gene phylogenetic tree constructed from the 384 L. rhamnosus genomes comprised five phylogenetic branches, with a random distribution of dairy and gut-associated isolates/genomes across the tree. No significant difference was identified in the overall profile of metabolism-related genes between dairy and gut-associated genomes; however, notably, the gut-associated strains/isolates contained more genes coding for specific metabolic pathways and carbohydrate-active enzymes, e.g., lacto-N-biosidase (EC 3.2.1.140; GT20) and lacto-N-biose phosphorylase/galacto-N-biose phosphorylase (EC 2.4.1.211; GH112). Further, we found that there was obvious intra-species diversification of the 93 cheese-originated L. rhamnosus isolates, forming three clades (Clades A, B, and C) in the reconstructed core-gene phylogenetic tree. There were numerous single nucleotide variations (over 10,000) across the three clades. Moreover, significant differences were observed in the content of metabolism-related genes across clades (p < 0.05, Adonis test), characterized by the enrichment in glycoside hydrolases in Clade C and the possession of unique metabolic pathways in each clade. These results implicated genomics/functional diversification of L. rhamnosus in a single food matrix and niche-driven adaptive evolution of isolates from dairy and host gut-associated origins. Our study shed insights into the selection of candidate strains for food industry applications.
Collapse
Affiliation(s)
- Lijun You
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Ruirui Lv
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Hao Jin
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Teng Ma
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhixin Zhao
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Lai-Yu Kwok
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Zhihong Sun
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China; Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
29
|
Donahue R, Sahoo JK, Rudolph S, Chen Y, Kaplan DL. Mucosa-Mimetic Materials for the Study of Intestinal Homeostasis and Disease. Adv Healthc Mater 2023; 12:e2300301. [PMID: 37329337 DOI: 10.1002/adhm.202300301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/11/2023] [Indexed: 06/19/2023]
Abstract
Mucus is a viscoelastic hydrogel that lines and protects the epithelial surfaces of the body that houses commensal microbiota and functions in host defense against pathogen invasion. As a first-line physical and biochemical barrier, intestinal mucus is involved in immune surveillance and spatial organization of the microbiome, while dysfunction of the gut mucus barrier is implicated in several diseases. Mucus can be collected from a variety of mammalian sources for study, however, established methods are challenging in terms of scale and efficiency, as well as with regard to rheological similarity to native human mucus. Therefore, there is a need for mucus-mimetic hydrogels that more accurately reflect the physical and chemical profile of the in vivo human epithelial environment to enable the investigation of the role of mucus in human disease and interactions with the intestinal microbiome. This review will evaluate the material properties of synthetic mucus mimics to date designed to address the above need, with a focus toward an improved understanding of the biochemical and immunological functions of these biopolymers related to utility for research and therapeutic applications.
Collapse
Affiliation(s)
- Rebecca Donahue
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Sara Rudolph
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|
30
|
Nejadmansouri M, Eskandari MH, Yousefi GH, Riazi M, Hosseini SMH. Promising application of probiotic microorganisms as Pickering emulsions stabilizers. Sci Rep 2023; 13:15915. [PMID: 37741896 PMCID: PMC10517997 DOI: 10.1038/s41598-023-43087-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023] Open
Abstract
The purpose of this work was to study the ability of nineteen food-grade microorganisms as Pickering emulsion (PE) stabilizers. Medium-chain triacylglycerol (MCT) oil-in-water (50:50) PEs were fabricated by 10 wt% or 15 wt% of thermally-inactivated yeast, cocci, Bacillus spp. and lactobacilli cells. The characteristics of microorganisms related to "Pickering stabilization" including morphology, surface charge, interfacial tension, and "contact angle" were firstly studied. After that, the cells-stabilized PEs were characterized from both kinetic and thermodynamic viewpoints, microstructure and rheological properties. The interfacial tension and "contact angle" values of various microorganisms ranged from 16.33 to 38.31 mN/m, and from 15° to 106°, respectively. The mean droplet size of PEs ranged from 11.51 to 57.69 µm. Generally, the physical stability of cell-stabilized PEs followed this order: lactobacilli > Bacillus spp. > cocci > yeast. These variations were attributed to the morphology and cell wall composition. Increasing the microorganism concentration significantly increased the physical stability of PEs from a maximum of 12 days at 10 wt% to 35 days at 15 wt% as a result of better interface coverage. Shear-thinning and dominant elastic behaviors were observed in PEs. Physical stability was affected by the free energy of detachment. Therefore, food-grade microorganisms are suggested for stabilizing PEs.
Collapse
Affiliation(s)
- Maryam Nejadmansouri
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Mohammad Hadi Eskandari
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Gholam Hossein Yousefi
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Riazi
- Enhanced Oil Recovery (EOR) Research Centre, IOR/EOR Research Institute, Shiraz University, Shiraz, Iran
- Department of Petroleum Engineering, School of Chemical and Petroleum Eng, Shiraz University, Shiraz, Iran
| | | |
Collapse
|
31
|
Perry EK, Tan MW. Bacterial biofilms in the human body: prevalence and impacts on health and disease. Front Cell Infect Microbiol 2023; 13:1237164. [PMID: 37712058 PMCID: PMC10499362 DOI: 10.3389/fcimb.2023.1237164] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/11/2023] [Indexed: 09/16/2023] Open
Abstract
Bacterial biofilms can be found in most environments on our planet, and the human body is no exception. Consisting of microbial cells encased in a matrix of extracellular polymers, biofilms enable bacteria to sequester themselves in favorable niches, while also increasing their ability to resist numerous stresses and survive under hostile circumstances. In recent decades, biofilms have increasingly been recognized as a major contributor to the pathogenesis of chronic infections. However, biofilms also occur in or on certain tissues in healthy individuals, and their constituent species are not restricted to canonical pathogens. In this review, we discuss the evidence for where, when, and what types of biofilms occur in the human body, as well as the diverse ways in which they can impact host health under homeostatic and dysbiotic states.
Collapse
Affiliation(s)
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech, South San Francisco, CA, United States
| |
Collapse
|
32
|
Luis AS, Hansson GC. Intestinal mucus and their glycans: A habitat for thriving microbiota. Cell Host Microbe 2023; 31:1087-1100. [PMID: 37442097 PMCID: PMC10348403 DOI: 10.1016/j.chom.2023.05.026] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/07/2023] [Accepted: 05/23/2023] [Indexed: 07/15/2023]
Abstract
The colon mucus layer is organized with an inner colon mucus layer that is impenetrable to bacteria and an outer mucus layer that is expanded to allow microbiota colonization. A major component of mucus is MUC2, a glycoprotein that is extensively decorated, especially with O-glycans. In the intestine, goblet cells are specialized in controlling glycosylation and making mucus. Some microbiota members are known to encode multiple proteins that are predicted to bind and/or cleave mucin glycans. The interactions between commensal microbiota and host mucins drive intestinal colonization, while at the same time, the microbiota can utilize the glycans on mucins and affect the colonic mucus properties. This review will examine this interaction between commensal microbes and intestinal mucins and discuss how this interplay affects health and disease.
Collapse
Affiliation(s)
- Ana S Luis
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Gunnar C Hansson
- Department of Medical Biochemistry and Cell Biology, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
33
|
Lee CG, Cha KH, Kim GC, Im SH, Kwon HK. Exploring probiotic effector molecules and their mode of action in gut-immune interactions. FEMS Microbiol Rev 2023; 47:fuad046. [PMID: 37541953 DOI: 10.1093/femsre/fuad046] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/17/2023] [Accepted: 08/03/2023] [Indexed: 08/06/2023] Open
Abstract
Probiotics, live microorganisms that confer health benefits when consumed in adequate amounts, have gained significant attention for their potential therapeutic applications. The beneficial effects of probiotics are believed to stem from their ability to enhance intestinal barrier function, inhibit pathogens, increase beneficial gut microbes, and modulate immune responses. However, clinical studies investigating the effectiveness of probiotics have yielded conflicting results, potentially due to the wide variety of probiotic species and strains used, the challenges in controlling the desired number of live microorganisms, and the complex interactions between bioactive substances within probiotics. Bacterial cell wall components, known as effector molecules, play a crucial role in mediating the interaction between probiotics and host receptors, leading to the activation of signaling pathways that contribute to the health-promoting effects. Previous reviews have extensively covered different probiotic effector molecules, highlighting their impact on immune homeostasis. Understanding how each probiotic component modulates immune activity at the molecular level may enable the prediction of immunological outcomes in future clinical studies. In this review, we present a comprehensive overview of the structural and immunological features of probiotic effector molecules, focusing primarily on Lactobacillus and Bifidobacterium. We also discuss current gaps and limitations in the field and propose directions for future research to enhance our understanding of probiotic-mediated immunomodulation.
Collapse
Affiliation(s)
- Choong-Gu Lee
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology, 679, Saimdang-ro, Gangneung 25451, Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, 679, Saimdang-ro, Seoul 02792, Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, 20, Ilsan-ro, Wonju 26493, Korea
| | - Gi-Cheon Kim
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, 77, Cheongam-ro, Pohang 37673, Korea
- Institute for Convergence Research and Education, Yonsei University, 50-1 Yonsei-ro, Seoul 03722, Korea
- ImmunoBiome Inc, Bio Open Innovation Center, 77, Cheongam-ro, Pohang 37673 , Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Korea
| |
Collapse
|
34
|
Sha Y, Yan Q, Liu J, Yu J, Xu S, He Z, Ren J, Qu J, Zheng S, Wang G, Dong W. Homologous genes shared between probiotics and pathogens affect the adhesion of probiotics and exclusion of pathogens in the gut mucus of shrimp. Front Microbiol 2023; 14:1195137. [PMID: 37389343 PMCID: PMC10301755 DOI: 10.3389/fmicb.2023.1195137] [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: 03/28/2023] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
Clarifying mechanisms underlying the selective adhesion of probiotics and competitive exclusion of pathogens in the intestine is a central theme for shrimp health. Under experimental manipulation of probiotic strain (i.e., Lactiplantibacillus plantarum HC-2) adhesion to the shrimp mucus, this study tested the core hypothesis that homologous genes shared between probiotic and pathogen would affect the adhesion of probiotics and exclusion of pathogens by regulating the membrane proteins of probiotics. Results indicated that the reduction of FtsH protease activity, which significantly correlated with the increase of membrane proteins, could increase the adhesion ability of L. plantarum HC-2 to the mucus. These membrane proteins mainly involved in transport (glycine betaine/carnitine/choline ABC transporter choS, ABC transporter, ATP synthase subunit a atpB, amino acid permease) and regulation of cellular processes (histidine kinase). The genes encoding the membrane proteins were significantly (p < 0.05) up-regulated except those encoding ABC transporters and histidine kinases in L. plantarum HC-2 when co-cultured with Vibrio parahaemolyticus E1, indicating that these genes could help L. plantarum HC-2 to competitively exclude pathogens. Moreover, an arsenal of genes predicted to be involved in carbohydrate metabolism and bacteria-host interactions were identified in L. plantarum HC-2, indicating a clear strain adaption to host's gastrointestinal tract. This study advances our mechanistic understanding of the selective adhesion of probiotics and competitive exclusion of pathogens in the intestine, and has important implications for screening and applying new probiotics for maintaining gut stability and host health.
Collapse
Affiliation(s)
- Yujie Sha
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Qingyun Yan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Jian Liu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Jiafeng Yu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Shicai Xu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Zhili He
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Jing Ren
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Jie Qu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Shiying Zheng
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
- Shandong Engineering Laboratory of Swine Health Big Data and Intelligent Monitoring, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Guomin Wang
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
| | - Weiying Dong
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, China
| |
Collapse
|
35
|
Pagnini C, Di Paolo MC, Urgesi R, Pallotta L, Fanello G, Graziani MG, Delle Fave G. Safety and Potential Role of Lactobacillus rhamnosus GG Administration as Monotherapy in Ulcerative Colitis Patients with Mild-Moderate Clinical Activity. Microorganisms 2023; 11:1381. [PMID: 37374884 DOI: 10.3390/microorganisms11061381] [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: 04/12/2023] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Probiotics are microorganisms that confer benefits to the host, and, for this reason, they have been proposed in several pathologic states. Specifically, probiotic bacteria have been investigated as a therapeutic option in ulcerative colitis (UC) patients, but clinical results are dishomogeneous. In particular, many probiotic species with different therapeutic schemes have been proposed, but no study has investigated probiotics in monotherapy in adequate trials for the induction of remission. Lactobacillus rhamnosus GG (LGG) is the more intensively studied probiotic and it has ideal characteristics for utilization in UC patients. The aim of the present study is to investigate the clinical efficacy and safety of LGG administration in an open trial, delivered in monotherapy at two different doses, in UC patients with mild-moderate disease. The UC patients with mild-moderate disease activity (Partial Mayo score ≥ 2) despite treatment with oral mesalamine were included. The patients stopped oral mesalamine and were followed up for one month, then were randomized to receive LGG supplement at dose of 1.2 or 2.4 × 1010 CFU/day for one month. At the end of the study, the clinical activity was evaluated and compared to that at the study entrance (efficacy). Adverse events were recorded (safety). The primary end-point was clinical improvement (reduction in the Partial Mayo score) and no serious adverse events, while the secondary end-points were the evaluation of different efficacies and safeties between the two doses of LGG. The patients with disease flares dropped out of the study and went back to standard therapy. The efficacy data were analyzed in an intention-to-treat (ITT) and per-protocol (PP) analysis. Out of the 76 patients included in the study, 75 started the probiotic therapy (n = 38 and 37 per group). In the ITT analysis, 32/76 (42%) responded to treatment, 21/76 (28%) remained stable, and 23/76 (30%) had a worsening of their clinical condition; 55 (72%) completed the treatment and were analyzed in a PP analysis: 32/55 (58%) had a clinical response, 21 (38%) remained stable, and 2 (4%) had a light worsening of their clinical condition (p < 0.0001). Overall, 37% of the patients had a disease remission. No severe adverse event was recorded, and only one patient stopped therapy due to obstinate constipation. No difference in the clinical efficacy and safety has been recorded between groups treated with different doses of LGG. The present prospective clinical trial demonstrates, for the first time, that LGG in monotherapy is safe and effective for the induction of remission in UC patients with mild-moderate disease activity (ClinicalTrials.gov identifier: NCT04102852).
Collapse
Affiliation(s)
- Cristiano Pagnini
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Carla Di Paolo
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Riccardo Urgesi
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Lorella Pallotta
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Fanello
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Giovanna Graziani
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Delle Fave
- Department of Gastroenterology, "Sapienza" University of Rome, 00185 Rome, Italy
- Onlus "S. Andrea", 00199 Rome, Italy
| |
Collapse
|
36
|
Bijle MN, Abdalla MM, Hung IFN, Yiu CKY. The effect of synbiotic-fluoride therapy on multi-species biofilm. J Dent 2023; 133:104523. [PMID: 37080530 DOI: 10.1016/j.jdent.2023.104523] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023] Open
Abstract
OBJECTIVES The study objective was to examine the effect of synbiotic-fluoride (SF) therapy within a multi-species cariogenic biofilm model system comprising of S. mutans, S. sanguinis, and S. gordonii. METHODS The SF therapy was prepared using 2% L-arginine (Arg), 0.2% NaF and probiotic L. rhamnosus GG (LRG). The 8 treatment groups were: Group 1: No treatment, Group 2: 2% Arg, Group 3: 0.2% NaF, Group 4: LRG, Group 5: 2% Arg+0.2% NaF, Group 6: 2% Arg+LRG, Group 7: 0.2% NaF+LRG, and Group 8: SF therapy (2% Arg+0.2% NaF +LRG). Multi-species biofilm model over 96 h comprising Streptococcus mutans, Streptococcus sanguinis, and Streptococcus gordonii was utilized. The biofilms received cariogenic challenge and SF therapy 2 × /day. The extracellular matrix components were analyzed for carbohydrates, proteins, and extra-cellular DNA (eDNA). The live/dead cells were imaged and quantified using confocal microscopy. The viable/dead bacterial concentrations were estimated using propidium monoazide-quantitative polymerase chain reaction (PMA-qPCR). The gene expressions for gtfB, sagP, arcA, argG, and argH were measured using real-time reverse transcriptase qPCR. RESULTS Carbohydrates and protein content with SF therapy were higher than non-LRG containing groups, while eDNA content was lower than other groups (p<0.05). Live bacterial proportions determined using confocal imaging with SF therapy were the lowest (p<0.05). The 2% Arg+LRG and SF therapy showed higher viable L. rhamnosus GG than 0.2% NaF+LRG (p<0.05). The dead S. mutans with SF therapy were higher than the other groups (p<0.05) with no difference from 2% Arg+0.2% NaF and 2% Arg+LRG (p>0.05). The SF therapy significantly downregulates gtfB and upregulates sagP, arcA, argG, argH gene expression (p<0.05). CONCLUSION Synbiotic-fluoride therapy effectuates multi-fold changes in the multi-species biofilm matrix and cellular components leading to superior ecological homeostasis than its individual contents, prebiotics (arginine), probiotic (L. rhamnosus GG), and fluorides (NaF). CLINICAL SIGNIFICANCE The ecological-based synbiotic-fluoride caries-preventive therapy aids in maintaining biofilm homeostasis to preempt/restore dysbiosis thereby sustaining dynamic-diverse health-associated microbial stability significant as a preventive regimen for high caries-risk patients.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- Assistant Professor in Paediatric Dentistry, Department of Clinical Sciences, College of Dentistry, Ajman University, United Arab Emirates; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Mohamed Mahmoud Abdalla
- Postdoctoral Fellow, Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong; Associate Professor, Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt.
| | - Ivan Fan Ngai Hung
- Ru Chien and Helen Lieh Professor in Health Sciences Pedagogy, Clinical Professor, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Cynthia Kar Yung Yiu
- Clinical Professor in Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
37
|
Bharindwal S, Goswami N, Jha P, Pandey S, Jobby R. Prospective Use of Probiotics to Maintain Astronaut Health during Spaceflight. Life (Basel) 2023; 13:life13030727. [PMID: 36983881 PMCID: PMC10058446 DOI: 10.3390/life13030727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
Maintaining an astronaut's health during space travel is crucial. Multiple studies have observed various changes in the gut microbiome and physiological health. Astronauts on board the International Space Station (ISS) had changes in the microbial communities in their gut, nose, and skin. Additionally, immune system cell alterations have been observed in astronauts with changes in neutrophils, monocytes, and T-cells. Probiotics help tackle these health issues caused during spaceflight by inhibiting pathogen adherence, enhancing epithelial barrier function by reducing permeability, and producing an anti-inflammatory effect. When exposed to microgravity, probiotics demonstrated a shorter lag phase, faster growth, improved acid tolerance, and bile resistance. A freeze-dried Lactobacillus casei strain Shirota capsule was tested for its stability on ISS for a month and has been shown to enhance innate immunity and balance intestinal microbiota. The usage of freeze-dried spores of B. subtilis proves to be advantageous to long-term spaceflight because it qualifies for all the aspects tested for commercial probiotics under simulated conditions. These results demonstrate a need to further study the effect of probiotics in simulated microgravity and spaceflight conditions and to apply them to overcome the effects caused by gut microbiome dysbiosis and issues that might occur during spaceflight.
Collapse
Affiliation(s)
- Sahaj Bharindwal
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Department of Biology, University of Naples Federico II, 80131 Naples, Italy
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| | - Nidhi Goswami
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| | - Pamela Jha
- Sunandan Divatia School of Science, NMIMS University Mumbai, Mumbai 400056, Maharashtra, India
| | - Siddharth Pandey
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
| | - Renitta Jobby
- Amity Centre of Excellence in Astrobiology, Amity University Mumbai, Mumbai 410206, Maharashtra, India
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai 410206, Maharashtra, India
| |
Collapse
|
38
|
Antibacterial and Immunostimulatory Activity of Potential Probiotic Lactic Acid Bacteria Isolated from Ethiopian Fermented Dairy Products. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9030258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Lactic acid bacteria (LAB) form a group of bacteria to which most probiotics belong and are commonly found in fermented dairy products. Fermented foods and beverages are foods made through desired microbial growth and enzymatic conversions of food components. In this study, 43 LAB were isolated from Ethiopian traditional cottage cheese, cheese, and yogurt and evaluated for their functional and safety properties as candidate probiotics. Twenty-seven isolates, representative of each fermented food type, were selected and identified to the species level. Limosilactobacillus fermentum was found to be the predominant species in all samples studied (70.4%), while 11.1% of isolates were identified as Lactiplantibacillus plantarum. All 27 isolates tested showed resistance to 0.5% bile salt, while 26 strains were resistant to pH 3. The LAB isolates were also evaluated for antagonistic properties against key pathogens, with strain-specific features observed for their antimicrobial activity. Five strains from cottage cheese (Lactiplantibacillus plantarum 54B, 54C, and 55A, Lactiplantibacillus pentosus 55B, and Pediococcus pentosaceus 95E) showed inhibitory activity against indicator pathogens that are key causes of gastrointestinal infections in Ethiopia, i.e., Escherichia coli, Salmonella enterica subsp. enterica var. Typhimurium, Staphylococcus aureus, Shigella flexneri, and Listeria monocytogenes. Strain-specific immunomodulatory activity monitored as nuclear factor kappa B (NF-κB) and interferon regulatory factor (IRF) activation was documented for Lactiplantibacillus plantarum 54B, 55A and P. pentosaceus 95E. Antibiotic susceptibility testing confirmed that all LAB isolates were safe concerning their antibiotic resistance profiles. Five isolates (especially Lactiplantibacillus plantarum 54B, 54C, and 55A, Lactiplantibacillus pentosus 55B, and P. pentosaceus 95E) showed promising results in all assays and are novel probiotic candidates of interest for clinical trial follow-up.
Collapse
|
39
|
Mendoza RM, Kim SH, Vasquez R, Hwang IC, Park YS, Paik HD, Moon GS, Kang DK. Bioinformatics and its role in the study of the evolution and probiotic potential of lactic acid bacteria. Food Sci Biotechnol 2023; 32:389-412. [PMID: 36911331 PMCID: PMC9992694 DOI: 10.1007/s10068-022-01142-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/30/2022] [Accepted: 07/13/2022] [Indexed: 11/04/2022] Open
Abstract
Due to their numerous well-established applications in the food industry, there have been many studies regarding the adaptation and evolution of lactic acid bacteria (LAB) in a wide variety of hosts and environments. Progress in sequencing technology and continual decreases in its costs have led to the availability of LAB genome sequence data. Bioinformatics has been central to the extraction of valuable information from these raw genome sequence data. This paper presents the roles of bioinformatics tools and databases in understanding the adaptation and evolution of LAB, as well as the bioinformatics methods used in the initial screening of LAB for probiotic potential. Moreover, the advantages, challenges, and limitations of employing bioinformatics for these purposes are discussed.
Collapse
Affiliation(s)
- Remilyn M. Mendoza
- Department of Animal Resources Science, Dankook University, 119 Dandae-ro, Cheonan, 31116 Republic of Korea
| | - Sang Hoon Kim
- Department of Animal Resources Science, Dankook University, 119 Dandae-ro, Cheonan, 31116 Republic of Korea
| | - Robie Vasquez
- Department of Animal Resources Science, Dankook University, 119 Dandae-ro, Cheonan, 31116 Republic of Korea
| | - In-Chan Hwang
- Department of Animal Resources Science, Dankook University, 119 Dandae-ro, Cheonan, 31116 Republic of Korea
| | - Young-Seo Park
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120 Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resource, Konkuk University, Seoul, 05029 Republic of Korea
| | - Gi-Seong Moon
- Division of Food Science and Biotechnology, Korea National University of Transportation, Jeungpyeong, 27909 Republic of Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, 119 Dandae-ro, Cheonan, 31116 Republic of Korea
| |
Collapse
|
40
|
In Silico Evidence of the Multifunctional Features of Lactiplantibacillus pentosus LPG1, a Natural Fermenting Agent Isolated from Table Olive Biofilms. Foods 2023; 12:foods12050938. [PMID: 36900455 PMCID: PMC10000683 DOI: 10.3390/foods12050938] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
In recent years, there has been a growing interest in obtaining probiotic bacteria from plant origins. This is the case of Lactiplantibacillus pentosus LPG1, a lactic acid bacterial strain isolated from table olive biofilms with proven multifunctional features. In this work, we have sequenced and closed the complete genome of L. pentosus LPG1 using both Illumina and PacBio technologies. Our intention is to carry out a comprehensive bioinformatics analysis and whole-genome annotation for a further complete evaluation of the safety and functionality of this microorganism. The chromosomic genome had a size of 3,619,252 bp, with a GC (Guanine-Citosine) content of 46.34%. L. pentosus LPG1 also had two plasmids, designated as pl1LPG1 and pl2LPG1, with lengths of 72,578 and 8713 bp (base pair), respectively. Genome annotation revealed that the sequenced genome consisted of 3345 coding genes and 89 non-coding sequences (73 tRNA and 16 rRNA genes). Taxonomy was confirmed by Average Nucleotide Identity analysis, which grouped L. pentosus LPG1 with other sequenced L. pentosus genomes. Moreover, the pan-genome analysis showed that L. pentosus LPG1 was closely related to the L. pentosus strains IG8, IG9, IG11, and IG12, all of which were isolated from table olive biofilms. Resistome analysis reported the absence of antibiotic resistance genes, whilst PathogenFinder tool classified the strain as a non-human pathogen. Finally, in silico analysis of L. pentosus LPG1 showed that many of its previously reported technological and probiotic phenotypes corresponded with the presence of functional genes. In light of these results, we can conclude that L. pentosus LPG1 is a safe microorganism and a potential human probiotic with a plant origin and application as a starter culture for vegetable fermentations.
Collapse
|
41
|
Van Holm W, Carvalho R, Delanghe L, Eilers T, Zayed N, Mermans F, Bernaerts K, Boon N, Claes I, Lebeer S, Teughels W. Antimicrobial potential of known and novel probiotics on in vitro periodontitis biofilms. NPJ Biofilms Microbiomes 2023; 9:3. [PMID: 36681674 PMCID: PMC9867767 DOI: 10.1038/s41522-023-00370-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
Several oral diseases are characterized by a shift within the oral microbiome towards a pathogenic, dysbiotic composition. Broad-spectrum antimicrobials are often part of patient care. However, because of the rising antibiotic resistance, alternatives are increasingly desirable. Alternatively, supplying beneficial species through probiotics is increasingly showing favorable results. Unfortunately, these probiotics are rarely evaluated comparatively. In this study, the in vitro effects of three known and three novel Lactobacillus strains, together with four novel Streptococcus salivarius strains were comparatively evaluated for antagonistic effects on proximal agar growth, antimicrobial properties of probiotic supernatant and the probiotic's effects on in vitro periodontal biofilms. Strain-specific effects were observed as differences in efficacy between genera and differences within genera. While some of the Lactobacillus candidates were able to reduce the periodontal pathobiont A. actinomycetemcomitans, the S. salivarius strains were not. However, the S. salivarius strains were more effective against periodontal pathobionts P. intermedia, P. gingivalis, and F. nucleatum. Vexingly, most of the Lactobacillus strains also negatively affected the prevalence of commensal species within the biofilms, while this was lower for S. salivarius strains. Both within lactobacilli and streptococci, some strains showed significantly more inhibition of the pathobionts, indicating the importance of proper strain selection. Additionally, some species showed reductions in non-target species, which can result in unexpected and unexplored effects on the whole microbiome.
Collapse
Affiliation(s)
- Wannes Van Holm
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium ,grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Rita Carvalho
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| | - Lize Delanghe
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Tom Eilers
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Naiera Zayed
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium ,grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium ,grid.411775.10000 0004 0621 4712Faculty of Pharmacy, Menoufia University, Shibin el Kom, Egypt
| | - Fabian Mermans
- grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Kristel Bernaerts
- grid.5596.f0000 0001 0668 7884Bio- and Chemical Systems Technology, Reactor Engineering and Safety, Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - Nico Boon
- grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | | | - Sarah Lebeer
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Wim Teughels
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
42
|
Assessing the Safety and Probiotic Characteristics of Lacticaseibacillus rhamnosus X253 via Complete Genome and Phenotype Analysis. Microorganisms 2023; 11:microorganisms11010140. [PMID: 36677432 PMCID: PMC9867440 DOI: 10.3390/microorganisms11010140] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Lacticaseibacillus rhamnosus is a generalist that can adapt to different ecological niches, serving as a valuable source of probiotics. The genome of L. rhamnosus X253 contains one chromosome and no plasmids, with a size of 2.99 Mb. Both single-copy orthologous gene-based phylogenetic analysis and average nucleotide identity indicated that dairy-derived L. rhamnosus X253 was most closely related to the human-intestine-derived strain L. rhamnosus LOCK908, rather than other dairy strains. The adaptation of L. rhamnosus X253 and the human-intestine-derived strain L. rhamnosus GG to different ecological niches was explained by structural variation analysis and COG annotation. Hemolytic assays, API ZYM assays, and antimicrobial susceptibility tests were performed to validate risk-related sequences such as virulence factors, toxin-encoding genes, and antibiotic-resistance genes in the genomes of L. rhamnosus X253 and GG. The results showed that L. rhamnosus GG was able to use L-fucose, had a higher tolerance to bile salt, and adhered better to CaCo-2 cells. In contrast, L. rhamnosus X253 was capable of utilizing D-lactose, withstood larger quantities of hydrogen peroxide, and possessed excellent antioxidant properties. This study confirmed the safety and probiotic properties of L. rhamnosus X253 via complete genome and phenotype analysis, suggesting its potential as a probiotic.
Collapse
|
43
|
Aksoyalp ZŞ, Temel A, Erdogan BR. Iron in infectious diseases friend or foe?: The role of gut microbiota. J Trace Elem Med Biol 2023; 75:127093. [PMID: 36240616 DOI: 10.1016/j.jtemb.2022.127093] [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: 04/09/2022] [Revised: 09/13/2022] [Accepted: 10/05/2022] [Indexed: 12/07/2022]
Abstract
Iron is a trace element involved in metabolic functions for all organisms, from microorganisms to mammalians. Iron deficiency is a prevalent health problem that affects billions of people worldwide, and iron overload could have some hazardous effect. The complex microbial community in the human body, also called microbiota, influences the host immune defence against infections. An imbalance in gut microbiota, dysbiosis, changes the host's susceptibility to infections by regulating the immune system. In recent years, the number of studies on the relationship between infectious diseases and microbiota has increased. Gut microbiota is affected by different parameters, including mode of delivery, hygiene habits, diet, drugs, and plasma iron levels during the lifetime. Gut microbiota may influence iron levels in the body, and iron overload and deficiency can also affect gut microbiota composition. Novel researches on microbiota shed light on the fact that the bidirectional interactions between gut microbiota and iron play a role in the pathogenesis of many diseases, especially infections. A better understanding of these interactions may help us to comprehend the pathogenesis of many infectious and metabolic diseases affecting people worldwide and following the development of more effective preventive and/or therapeutic strategies. In this review, we aimed to present the iron-mediated host-gut microbiota interactions, susceptibility to bacterial infections, and iron-targeted therapy approaches for infections.
Collapse
Affiliation(s)
- Zinnet Şevval Aksoyalp
- Izmir Katip Celebi University, Faculty of Pharmacy, Department of Pharmacology, Izmir, Turkey.
| | - Aybala Temel
- Izmir Katip Celebi University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Izmir, Turkey.
| | - Betul Rabia Erdogan
- Izmir Katip Celebi University, Faculty of Pharmacy, Department of Pharmacology, Izmir, Turkey.
| |
Collapse
|
44
|
Kennedy KM, de Goffau MC, Perez-Muñoz ME, Arrieta MC, Bäckhed F, Bork P, Braun T, Bushman FD, Dore J, de Vos WM, Earl AM, Eisen JA, Elovitz MA, Ganal-Vonarburg SC, Gänzle MG, Garrett WS, Hall LJ, Hornef MW, Huttenhower C, Konnikova L, Lebeer S, Macpherson AJ, Massey RC, McHardy AC, Koren O, Lawley TD, Ley RE, O'Mahony L, O'Toole PW, Pamer EG, Parkhill J, Raes J, Rattei T, Salonen A, Segal E, Segata N, Shanahan F, Sloboda DM, Smith GCS, Sokol H, Spector TD, Surette MG, Tannock GW, Walker AW, Yassour M, Walter J. Questioning the fetal microbiome illustrates pitfalls of low-biomass microbial studies. Nature 2023; 613:639-649. [PMID: 36697862 PMCID: PMC11333990 DOI: 10.1038/s41586-022-05546-8] [Citation(s) in RCA: 189] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/09/2022] [Indexed: 01/26/2023]
Abstract
Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.
Collapse
Affiliation(s)
- Katherine M Kennedy
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Marcus C de Goffau
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Wellcome Sanger Institute, Cambridge, UK
| | - Maria Elisa Perez-Muñoz
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Marie-Claire Arrieta
- International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peer Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Thorsten Braun
- Department of Obstetrics and Experimental Obstetrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frederic D Bushman
- Department of Microbiology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Willem M de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jonathan A Eisen
- Department of Evolution and Ecology, University of California, Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- UC Davis Genome Center, University of California, Davis, Davis, CA, USA
| | - Michal A Elovitz
- Maternal and Child Health Research Center, Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Stephanie C Ganal-Vonarburg
- Universitätsklinik für Viszerale Chirurgie und Medizin, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michael G Gänzle
- Department of Agriculture, Food and Nutrition Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
- Department of Medicine and Division of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
- Chair of Intestinal Microbiome, ZIEL-Institute for Food and Health, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Curtis Huttenhower
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liza Konnikova
- Departments of Pediatrics and Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Andrew J Macpherson
- Department for Biomedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Ruth C Massey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Alice Carolyn McHardy
- Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
- German Center for Infection Research (DZIF), Hannover Braunschweig site, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Trevor D Lawley
- Department of Vascular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Liam O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL, USA
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Jeroen Raes
- VIB Center for Microbiology, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thomas Rattei
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Eran Segal
- Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- European Institute of Oncology (IEO), IRCCS, Milan, Italy
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| | - Gordon C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Harry Sokol
- Gastroenterology Department, AP-HP, Saint Antoine Hospital, Centre de Recherche Saint-Antoine, CRSA, INSERM and Sorbonne Université, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, Jouy en Josas, France
| | - Tim D Spector
- Department of Twin Research, King's College London, London, UK
| | - Michael G Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gerald W Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Moran Yassour
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Microbiology and Molecular Genetics, IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jens Walter
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
- Department of Medicine, University College Cork, Cork, Ireland.
| |
Collapse
|
45
|
Food-Grade Bacteria Combat Pathogens by Blocking AHL-Mediated Quorum Sensing and Biofilm Formation. Foods 2022; 12:foods12010090. [PMID: 36613306 PMCID: PMC9818890 DOI: 10.3390/foods12010090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Disrupting bacterial quorum sensing (QS) signaling is a promising strategy to combat pathogenic biofilms without the development of antibiotic resistance. Here, we report that food-associated bacteria can interfere with the biofilm formation of a Gram-negative pathogenic bacterium by targeting its AHL (acyl-homoserine lactone) QS system. This was demonstrated by screening metabolic end-products of different lactobacilli and propionibacteria using Gram-negative and biofilm-forming Chromobacterium violaceum as the QS reporter and our anti-QS microscale screening platform with necessary modifications. The method was optimized in terms of the inoculation technique and the concentrations of D-glucose and L-tryptophan, two key factors controlling the synthesis of violacein, a purple pigment indicating the activation of the QS system in C. violaceum. These improvements resulted in ca. 16-times higher violacein yields and enabled revealing anti-QS effects of Lactobacillus acidophilus, Lentilactobacillus kefiri, Lacticaseibacillus rhamnosus and Propionibacterium freudenreichii, including new cheese-associated strains. Our findings also suggest that acetate and propionate excreted by these species are the main factors that interrupt the QS-mediated signaling and subsequent biofilm growth without affecting the cell viability of the C. violaceum reporter. Thus, the present study reports a revised anti-QS screening method to accurately define new bacteria with an ability to combat pathogens in a safe and sustainable way.
Collapse
|
46
|
Biagini F, Daddi C, Calvigioni M, De Maria C, Zhang YS, Ghelardi E, Vozzi G. Designs and methodologies to recreate in vitro human gut microbiota models. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00210-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AbstractThe human gut microbiota is widely considered to be a metabolic organ hidden within our bodies, playing a crucial role in the host’s physiology. Several factors affect its composition, so a wide variety of microbes residing in the gut are present in the world population. Individual excessive imbalances in microbial composition are often associated with human disorders and pathologies, and new investigative strategies to gain insight into these pathologies and define pharmaceutical therapies for their treatment are needed. In vitro models of the human gut microbiota are commonly used to study microbial fermentation patterns, community composition, and host-microbe interactions. Bioreactors and microfluidic devices have been designed to culture microorganisms from the human gut microbiota in a dynamic environment in the presence or absence of eukaryotic cells to interact with. In this review, we will describe the overall elements required to create a functioning, reproducible, and accurate in vitro culture of the human gut microbiota. In addition, we will analyze some of the devices currently used to study fermentation processes and relationships between the human gut microbiota and host eukaryotic cells.
Graphic abstract
Collapse
|
47
|
Li H, Wang J, Fu Y, Zhu K, Dong Z, Shan J, Di L, Jiang S, Yuan T. The Bioavailability of Glycyrrhizinic Acid Was Enhanced by Probiotic Lactobacillus rhamnosus R0011 Supplementation in Liver Fibrosis Rats. Nutrients 2022; 14:nu14245278. [PMID: 36558437 PMCID: PMC9782010 DOI: 10.3390/nu14245278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Glycyrrhizinic acid (GL) is clinically applied to treat liver injury, and the bioavailability of orally administered GL is closely related to the gut microbiota. Therefore, the dysbiosis of gut flora in liver injury could significantly influence GL bioavailability. Still, less is known about the impact of probiotic supplementation on the bio-absorption process of oral medication, especially under a pathological state. Herein, probiotic L. rhamnosus R0011 (R0011) with a high viability in the harsh gastrointestinal environment was selected, and the effect of R0011 on the GL bioavailability in rats was investigated. Four groups of rats (n = 6 per group) were included: the normal group (N group), the normal group supplemented with R0011 (NLGG group), CCl4-induced chronic liver injury model (M group), and the model group supplemented with R0011 (MLGG group). Our results showed that liver injury was successfully induced in the M and MLGG groups via an intraperitoneal injection of 50% (v/v) CCl4 solution. Healthy rats supplemented with R0011 could increase the bioavailability of GL by 1.4-fold compared with the normal group by plasma pharmacokinetic analysis. Moreover, the GL bioavailability of MLGG group was significantly increased by 4.5-fold compared with the model group. R0011 directly improved gut microbial glucuronidase and downregulated the host intestinal drug transporter gene expression of multidrug resistance protein 2 (MRP2). More critically, R0011 restored the gut microbiota composition and regulated the metabolic function, significantly enhancing the microbial tryptophan metabolic pathway compared with the pathological state, which may indirectly promote the bioavailability of GL. Overall, these data may provide possible strategies by which to address the low bioavailability of traditional medicine through probiotic intervention.
Collapse
Affiliation(s)
- Huifang Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jing Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yifan Fu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ke Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhiling Dong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Engineering Research Centre for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shu Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tianjie Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Correspondence:
| |
Collapse
|
48
|
Shanmugasundarasamy T, Karaiyagowder Govindarajan D, Kandaswamy K. A review on pilus assembly mechanisms in Gram-positive and Gram-negative bacteria. Cell Surf 2022; 8:100077. [PMID: 35493982 PMCID: PMC9046445 DOI: 10.1016/j.tcsw.2022.100077] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022] Open
Abstract
The surface of Gram-positive and Gram-negative bacteria contains long hair-like proteinaceous protrusion known as pili or fimbriae. Historically, pilin proteins were considered to play a major role in the transfer of genetic material during bacterial conjugation. Recent findings however elucidate their importance in virulence, biofilm formation, phage transduction, and motility. Therefore, it is crucial to gain mechanistic insights on the subcellular assembly of pili and the localization patterns of their subunit proteins (major and minor pilins) that aid the macromolecular pilus assembly at the bacterial surface. In this article, we review the current knowledge of pilus assembly mechanisms in a wide range of Gram-positive and Gram-negative bacteria, including subcellular localization patterns of a few pilin subunit proteins and their role in virulence and pathogenesis.
Collapse
|
49
|
Lahiri D, Nag M, Dutta B, Sarkar T, Pati S, Basu D, Abdul Kari Z, Wei LS, Smaoui S, Wen Goh K, Ray RR. Bacteriocin: A natural approach for food safety and food security. Front Bioeng Biotechnol 2022; 10:1005918. [PMID: 36353741 PMCID: PMC9637989 DOI: 10.3389/fbioe.2022.1005918] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/05/2022] [Indexed: 08/27/2023] Open
Abstract
The call to cater for the hungry is a worldwide problem in the 21st century. Food security is the utmost prime factor for the increasing demand for food. Awareness of human health when using chemical preservatives in food has increased, resulting in the use of alternative strategies for preserving food and enhancing its shelf-life. New preservatives along with novel preservation methods have been instigated, due to the intensified demand for extended shelf-life, along with prevention of food spoilage of dairy products. Bacteriocins are the group of ribosomally synthesized antimicrobial peptides; they possess a wide range of biological activities, having predominant antibacterial activity. The bacteriocins produced by the lactic acid bacteria (LAB) are considered to be of utmost importance, due to their association with the fermentation of food. In recent times among various groups of bacteriocins, leaderless and circular bacteriocins are gaining importance, due to their extensive application in industries. These groups of bacteriocins have been least studied as they possess peculiar structural and biosynthetic mechanisms. They chemically possess N-to-C terminal covalent bonds having a predominant peptide background. The stability of the bacteriocins is exhibited by the circular structure. Up till now, very few studies have been performed on the molecular mechanisms. The structural genes associated with the bacteriocins can be combined with the activity of various proteins which are association with secretion and maturation. Thus the stability of the bacteriocins can be used effectively in the preservation of food for a longer period of time. Bacteriocins are thermostable, pH-tolerant, and proteolytically active in nature, which make their usage convenient to the food industry. Several research studies are underway in the domain of biopreservation which can be implemented in food safety and food security.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Bandita Dutta
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Govt of West Bengal, Malda, India
| | - Siddhartha Pati
- NatNov Bioscience Private Limited, Balasore, India
- Skills Innovation and Academic Network (SIAN) Institute, Association for Biodiversity Conservation and Research (ABC), Balasore, India
| | - Debarati Basu
- Department of Biotechnology, University of Engineering and Management, Kolkata, India
| | - Zulhisyam Abdul Kari
- Department of Agricultural Sciences, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli Campus, Kelantan, Malaysia
| | - Lee Seong Wei
- Department of Agricultural Sciences, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli Campus, Kelantan, Malaysia
| | - Slim Smaoui
- Laboratory of Microorganisms and Biomolecules, Center of Biotechnology of Sfax, Sfax, Tunisia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| |
Collapse
|
50
|
Shulman RJ, Chichlowski M, Orozco FG, Harris CL, Wampler JL, Bokulich NA, Berseth CL. Infant behavioral state and stool microbiome in infants receiving Lactocaseibacillus rhamnosus GG in formula: randomized controlled trial. BMC Pediatr 2022; 22:580. [PMID: 36207675 PMCID: PMC9541012 DOI: 10.1186/s12887-022-03647-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Background Our aim was to evaluate infant behavioral state, stool microbiome profile and calprotectin in infants with infantile colic receiving a partially hydrolyzed protein formula with or without added Lacticaseibacillus (formerly Lactobacillus) rhamnosus GG (LGG). Methods In this single-center, double-blind, controlled, parallel, prospective study, term infants (14–28 days of age) identified with colic (using modified Wessel’s criteria: cried and/or fussed ≥ 3 h/day for ≥ 3 days/week, in a one-week period) were randomized to receive one of two formulas over a three-week feeding period: marketed partially hydrolyzed cow’s milk-based infant formula (PHF, n = 35) or a similar formula with added LGG (PHF-LGG, n = 36). Parent-reported infant behavior was recorded at three time points (Study Days 2–4, 10–12, and 18–20). Duration (hours/day) of crying/fussing (averaged over each three-day period) was the primary outcome. Stool samples were collected at Baseline and Study End (Days 19–21) to determine stool LGG colonization (by qPCR) and microbial abundance (using 16S rRNA gene sequencing) and calprotectin (μg/g). Results Duration of crying/fussing (mean ± SE) decreased and awake/content behavior increased over time with no significant group differences over the course of the study. There were no group differences in the percentage of infants who experienced colic by study end. Colic decreased by Study End vs Baseline in both groups. Change in fecal calprotectin also was similar between groups. Comparing Study End vs Baseline, LGG abundance was greater in the PHF-LGG group (P < 0.001) whereas alpha diversity was greater in the PHF group (P = 0.022). Beta diversity was significantly different between PHF and PHF-LGG at Study End (P = 0.05). By study end, relative abundance of L. rhamnosus was higher in the PHF-LGG vs PHF group and vs Baseline. Conclusions In this pilot study of infants with colic, both study formulas were well tolerated. Crying/fussing decreased and awake/content behavior increased in both study groups over the course of the study. Study results demonstrate a successful introduction of the probiotic to the microbiome. The partially hydrolyzed protein formula with added LGG was associated with significant changes in the gut microbiome. Trial registration ClinicalTrials.gov, ClinicalTrials.gov Identifier: NCT02340143. Registered 16/01/2015. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03647-x.
Collapse
Affiliation(s)
- Robert J Shulman
- Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Center for Pediatric Abdominal Pain Research, Baylor College of Medicine, Houston, TX, 77030, USA. .,Texas Children's Hospital, 6621 Fannin St., Houston, TX, 77030, USA. .,USDA/ARS Children's Nutrition Research Center, 1100 Bates St., Room 8072, Houston, TX, 77030, USA.
| | - Maciej Chichlowski
- Medical and Scientific Affairs, Reckitt
- Mead Johnson Nutrition Institute, Evansville, IN, 47721, USA
| | - Fabiola Gutierrez Orozco
- Medical and Scientific Affairs, Reckitt
- Mead Johnson Nutrition Institute, Evansville, IN, 47721, USA
| | - Cheryl L Harris
- Medical and Scientific Affairs, Reckitt
- Mead Johnson Nutrition Institute, Evansville, IN, 47721, USA
| | - Jennifer L Wampler
- Medical and Scientific Affairs, Reckitt
- Mead Johnson Nutrition Institute, Evansville, IN, 47721, USA
| | - Nicholas A Bokulich
- Laboratory of Food Systems Biotechnology, Institute of Food, Nutrition, and Health, ETH Zurich, Zurich, Switzerland
| | - Carol Lynn Berseth
- Medical and Scientific Affairs, Reckitt
- Mead Johnson Nutrition Institute, Evansville, IN, 47721, USA
| |
Collapse
|