1
|
Coxon J, Linder E, Sweet C, Magness S, Green L. Replicating Host-Microbiome Interactions: Harnessing Organ-on-a-Chip and Organoid Technologies to Model Vaginal and Lung Physiology. Annu Rev Biomed Eng 2025; 27:403-423. [PMID: 39971348 DOI: 10.1146/annurev-bioeng-110122-122343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Organ-on-a-chip (OOC) and organoid technologies are at the forefront of developing sophisticated in vitro systems that replicate complex host-microbiome interactions, including those associated with vaginal health and lung infection. We explore how these technologies provide insights into host-microbiome and host-pathogen interactions and the associated immune responses. Integrating omics data and high-resolution imaging in analyzing these models enhances our understanding of host-microbiome interactions' temporal and spatial aspects, paving the way for new diagnostic and treatment strategies. This review underscores the potential of OOC and organoid technologies in elucidating the complexities of vaginal health and lung disease, which have received less attention than other organ systems in recent organoid and OCC studies. Yet, each system presents notable characteristics, rendering them ideal candidates for these designs. Additionally, this review describes the key factors associated with each organ system and how to choose the technology setup to replicate human physiology.
Collapse
Affiliation(s)
- Jade Coxon
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA;
| | - Emily Linder
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Caden Sweet
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott Magness
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, North Carolina, USA
| | - Leopold Green
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
2
|
Mao W, Rong Y, Zhang H, Yuan F, Wang Y, Wang M, Wang L, Wang PG, Chen M, Wang S, Kong Y. Characterization and application in recombinant N-GlcNAc-protein production of a novel endo-β-N-acetylglucosaminidase from Listeria booriae. Bioorg Chem 2025; 157:108290. [PMID: 39983405 DOI: 10.1016/j.bioorg.2025.108290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/07/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Endo-β-N-acetylglucosaminidases (ENGases) are essential enzymes for hydrolyzing N-glycans, with applications in protein N-glycosylation analysis and glycoprotein synthesis. In this study, a novel GH18 family ENGase, Endo-LB, was identified from Listeria booriae FSL A5-0281. Composed of 593 amino acids (65.78 kDa), Endo-LB features with two domains: an Endo S-like catalytic domain and a mucin-binding protein (MucBP) domain. Recombinant Endo-LB, expressed in Escherichia coli BL21 (DE3) pLysS, exhibited a specific activity of 198.25 U/mg and hydrolyzed high mannose-type N-glycans at a temperature from 4 °C to 60 °C with optimal activity at 37 °C and pH 6.0 (range 3.0 to 10.0), making it versatile for various environmental conditions. The MucBP domain does not affect soluble Endo-LB activity but influences interaction with mucin on cell surface, suggesting potential application in targeting specific glycoproteins in complex biological environments. To address the heterogeneity of N-glycans in Pichia pastoris (Komagataella phaffii) expression, Endo-LB was further expressed in the Golgi of P. pastoris, efficiently producing glycoproteins, such as Erythropoietin (EPO) (37 mg/L) and Darbepoetin α (53 mg/L) with nearly complete N-glycans truncation, which can be further extended to generate diverse N-glycan structures. These findings highlight the versatility and potential utility of Endo-LB in glycoprotein engineering and biotechnological applications.
Collapse
Affiliation(s)
- Weian Mao
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Yongheng Rong
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Hongmei Zhang
- Department of Endocrinology, Zibo Central Hospital, Zibo 255020, China
| | - Fang Yuan
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Yankang Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China
| | - Mei Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Linhan Wang
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China
| | - Peng George Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Min Chen
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Shengjun Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266113, China.
| | - Yun Kong
- National Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| |
Collapse
|
3
|
Nori SRC, Walsh CJ, McAuliffe FM, Moore RL, Van Sinderen D, Feehily C, Cotter PD. Strain-level variation among vaginal Lactobacillus crispatus and Lactobacillus iners as identified by comparative metagenomics. NPJ Biofilms Microbiomes 2025; 11:49. [PMID: 40122890 PMCID: PMC11930926 DOI: 10.1038/s41522-025-00682-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 03/09/2025] [Indexed: 03/25/2025] Open
Abstract
The vaginal microbiome, a relatively simple, low diversity ecosystem crucial for female health, is often dominated by Lactobacillus spp. Detailed strain-level data, facilitated by shotgun sequencing, can provide a greater understanding of the mechanisms of colonization and host-microbe interactions. We analysed 354 vaginal metagenomes from pregnant women in Ireland to investigate metagenomic community state types and strain-level variation, focusing on cell surface interfaces. Our analysis revealed multiple subspecies, with Lactobacillus crispatus and Lactobacillus iners being the most dominant. We found genes, including putative mucin-binding genes, distinct to L. crispatus subspecies. Using 337 metagenome-assembled genomes, we observed a higher number of strain-specific genes in L. crispatus related to cell wall biogenesis, carbohydrate and amino acid metabolism, many under positive selection. A cell surface glycan gene cluster was predominantly found in L. crispatus but absent in L. iners and Gardnerella vaginalis. These findings highlight strain-specific factors associated with colonisation and host-microbe interactions.
Collapse
Affiliation(s)
- Sai Ravi Chandra Nori
- Teagasc Food Research Centre, Fermoy, Co, Cork, Ireland
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
- SFI Centre for Research Training in Genomics Data Science, School of Mathematics, Statistics & Applied Mathematics, University of Galway, Galway, Ireland
| | - Calum J Walsh
- The Centre for Pathogen Genomics, Department of Microbiology & Immunology, Peter Doherty Institute for Infection & Immunity, University of Melbourne, Melbourne, Australia
| | - Fionnuala M McAuliffe
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Rebecca L Moore
- UCD Perinatal Research Centre, School of Medicine, University College Dublin, National Maternity Hospital, Dublin, Ireland
| | - Douwe Van Sinderen
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Conor Feehily
- School of Infection and Immunity, University of Glasgow, Glasgow, G12 8TA, United Kingdom.
| | - Paul D Cotter
- Teagasc Food Research Centre, Fermoy, Co, Cork, Ireland.
- APC Microbiome Ireland, National University of Ireland, Cork, Ireland.
- School of Microbiology, University College Cork, Cork, Ireland.
| |
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
|
Seo H, Kim S, Beck S, Song HY. Perspectives on Microbiome Therapeutics in Infectious Diseases: A Comprehensive Approach Beyond Immunology and Microbiology. Cells 2024; 13:2003. [PMID: 39682751 PMCID: PMC11640688 DOI: 10.3390/cells13232003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Although global life expectancy has increased over the past 20 years due to advancements in managing infectious diseases, one-fifth of people still die from infections. In response to this ongoing threat, significant efforts are underway to develop vaccines and antimicrobial agents. However, pathogens evolve resistance mechanisms, complicating their control. The COVID-19 pandemic has underscored the limitations of focusing solely on the pathogen-killing strategies of immunology and microbiology to address complex, multisystemic infectious diseases. This highlights the urgent need for practical advancements, such as microbiome therapeutics, that address these limitations while complementing traditional approaches. Our review emphasizes key outcomes in the field, including evidence of probiotics reducing disease severity and insights into host-microbiome crosstalk that have informed novel therapeutic strategies. These findings underscore the potential of microbiome-based interventions to promote physiological function alongside existing strategies aimed at enhancing host immune responses and pathogen destruction. This narrative review explores microbiome therapeutics as next-generation treatments for infectious diseases, focusing on the application of probiotics and their role in host-microbiome interactions. While offering a novel perspective grounded in a cooperative defense system, this review also addresses the practical challenges and limitations in translating these advancements into clinical settings.
Collapse
Affiliation(s)
- Hoonhee Seo
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Sukyung Kim
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
| | - Samuel Beck
- Center for Aging Research, Department of Dermatology, Chobanian & Avedisian School of Medicine, Boston University, J-607, 609 Albany, Boston, MA 02118, USA
| | - Ho-Yeon Song
- Human Microbiome Medical Research Center (HM·MRC), School of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Sinchang-myeon, Asan-si 31538, Chungnam-do, Republic of Korea
- Department of Microbiology and Immunology, School of Medicine, Soonchunhyang University, 31, Suncheonhyang 6-gil, Cheonan-si 31151, Chungnam-do, Republic of Korea
| |
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
|
Janssen AWF, van der Lugt B, Duivenvoorde LPM, Vos AP, Bastiaan-Net S, Tomassen MMM, Verbokkem JAC, Blok-Heimerikx E, Hooiveld GJEJ, van Baarlen P, Ferrier L, van der Zande M. Comparison of iPSC-derived human intestinal epithelial cells with Caco-2 cells and human in vivo data after exposure to Lactiplantibacillus plantarum WCFS1. Sci Rep 2024; 14:26464. [PMID: 39488516 PMCID: PMC11531526 DOI: 10.1038/s41598-024-74802-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/30/2024] [Indexed: 11/04/2024] Open
Abstract
To investigate intestinal health and its potential disruptors in vitro, representative models are required. Human induced pluripotent stem cell (hiPSC)-derived intestinal epithelial cells (IECs) more closely resemble the in vivo intestinal tissue than conventional in vitro models like human colonic adenocarcinoma Caco-2 cells. However, the potential of IECs to study immune-related responses upon external stimuli has not been investigated in detail yet. The aim of the current study was to evaluate immune-related effects of IECs by challenging them with a pro-inflammatory cytokine cocktail. Subsequently, the effects of Lactiplantibacillus plantarum WCFS1 were investigated in unchallenged and challenged IECs. All exposures were compared to Caco-2 cells and in vivo data where possible. Upon the inflammatory challenge, IECs and Caco-2 cells induced a pro-inflammatory response which was strongest in IECs. Heat-killed L. plantarum exerted the strongest effect on immune parameters in the IEC model, while L. plantarum in the stationary growth phase had most pronounced effects on immune-related gene expression in Caco-2 cells. Unfortunately, comparison to in vivo transcriptomics data showed limited similarities, which could be explained by essential differences in the study setups. Altogether, hiPSC-derived IECs show a high potential as a model to study immune-related responses in the intestinal epithelium in vitro.
Collapse
Grants
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- KB37-001-003 Dutch ministries of Agriculture, Nature and Food Quality and Economic affairs
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
- LWV19125 Top consortium for Knowledge and Innovation (TKI) Agri & Food
Collapse
Affiliation(s)
- Aafke W F Janssen
- Wageningen Food Safety Research, Part of Wageningen University & Research, Akkermaalsbos 2, 6708 WB, Wageningen, The Netherlands
| | - Benthe van der Lugt
- Wageningen Food Safety Research, Part of Wageningen University & Research, Akkermaalsbos 2, 6708 WB, Wageningen, The Netherlands
| | - Loes P M Duivenvoorde
- Wageningen Food Safety Research, Part of Wageningen University & Research, Akkermaalsbos 2, 6708 WB, Wageningen, The Netherlands
| | - Arjan Paul Vos
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Monic M M Tomassen
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Janine A C Verbokkem
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Emmie Blok-Heimerikx
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Peter van Baarlen
- Host-Microbe Interactomics, Animal Sciences, Wageningen University & Research, Wageningen, The Netherlands
| | - Laurent Ferrier
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Meike van der Zande
- Wageningen Food Safety Research, Part of Wageningen University & Research, Akkermaalsbos 2, 6708 WB, Wageningen, The Netherlands.
| |
Collapse
|
8
|
Kim WJ, Jung G, Kim T, Kim J, Hurh BS, Kim H, Soung DY. Heat-Killed Lactobacillus paracasei SMB092 Reduces Halitosis by Stimulating the Expression of β-Defensins in Oral Keratinocytes. Microorganisms 2024; 12:2147. [PMID: 39597536 PMCID: PMC11596102 DOI: 10.3390/microorganisms12112147] [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: 09/30/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
The purpose of this study is to evaluate Lactobacillus paracasei SMB092 as a prophylactic agent for oral pathogens. We examined the physical interaction of SMB092 with a host by identifying the presence of mucus-binding (MuB) protein domains and the capacity of the mucin binding. We determined the role of heat-killed SMB092 in host oral immunity by quantifying the mRNA levels of β-defensins (BDs), Toll-like receptors (TLRs), and their cofactors (CD14/CD36) in normal human oral keratinocytes (HOK-16B cells). To assess the clinically relevant oral health effects of heat-killed SMB092, the growth of Porphyromonas (P.) gingivalis and the production of a volatile sulfur compound (H2S) were also measured in the filtered condition media (FCM) obtained from its cultures with HOK-16B cells. SMB092 possessed 14 putative MuB protein domains and was attached to mucin. Significant amounts of hBD1/2 and TLR2/6 were expressed in heat-killed SMB092-treated HOK-16B cells. The specific neutralization of TLR2 attenuated the expression of hBD1/2 and CD14/CD36. The FCM inhibited the growth of P. gingivalis and the production of H2S. Our data indicate that heat-killed SMB092 may contribute to a healthy oral microbiome as an immune stimulant in the production of BDs via the activation of the TLR2/6 signaling pathway.
Collapse
Affiliation(s)
- Won-Ju Kim
- Sempio Foods Company, R&D Center, Cheongju 28156, Republic of Korea; (W.-J.K.); (J.K.); (B.-S.H.)
| | - Gyubin Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea;
| | - Taewook Kim
- CJ Bioscience Inc., Seoul 04527, Republic of Korea;
| | - Jinseon Kim
- Sempio Foods Company, R&D Center, Cheongju 28156, Republic of Korea; (W.-J.K.); (J.K.); (B.-S.H.)
| | - Byung-Serk Hurh
- Sempio Foods Company, R&D Center, Cheongju 28156, Republic of Korea; (W.-J.K.); (J.K.); (B.-S.H.)
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Co., Ltd., Yongin 17104, Republic of Korea;
| | - Do Yu Soung
- Sempio Foods Company, R&D Center, Cheongju 28156, Republic of Korea; (W.-J.K.); (J.K.); (B.-S.H.)
| |
Collapse
|
9
|
Boyajian J, Abosalha A, Islam P, Kassab A, Santos M, Shum-Tim C, Renesteen E, Makhlouf S, Chen J, Prakash S. Select Probiotics Exhibit Antioxidative and Anti-Inflammatory Properties for Gut Modulation: In Vitro Analysis. THE EUROBIOTECH JOURNAL 2024; 8:149-157. [DOI: 10.2478/ebtj-2024-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Abstract
The gut microbiota harbors a complex ecosystem of bacteria that govern host health homeostasis. Alterations to the intestinal environment, known as gut dysbiosis, is associated with several diseases. Targeting the gut with microbiome-engineered therapeutics, such as probiotics, is a promising approach to restore microbial homeostasis and host health. Probiotics can effectively improve the gut environment, although strain-specific mechanisms remain largely unknown. Thus, this study aims to identify beneficial action of select probiotics to modulate the gut environment through antioxidative and anti-inflammatory properties. To this end, we tested the interaction among three probiotic strains – Lactobacillus gasseri A237 (LgA237), Lactobacillus plan-tarum WCFS1 (LpWCFS1) and Lactobacillus fermentum NCIMB 5221 (Lf5221) – and a human intestinal epithelial cell line, HT-29, for adhesion properties, radical scavenging abilities and anti-inflammatory activities. All three probiotics adhere well to HT-29 cells, indicating proper gut colonization. LpWCFS1 demonstrated the greatest adhesion capacity (68.3%), followed by LgA237 (35.5%) and Lf5221 (25.9%). The probiotics also exhibit excellent antioxidant properties via DPPH radical scavenging activity, comparable to quercetin, a known and potent antioxidant. Moreover, LgA237, LpWCFS1 and Lf5221 decrease interleukin-8 expression in lipopolysaccharide (LPS)-damaged HT-29 cells (41.19, 34.53 and 14.80% reduction, respectively), compared to non-treated cells. Further investigation of LpWCFS1 and LgA237 revealed a significant (p<0.0001) reduction in monocyte chemotactic and activating factor (MCAF) protein expression by 63.81 and 60.33%, respectively, in colitis-induced IECs. Overall, our results indicate adhesion, antioxidative and anti-inflammatory therapeutic potential of the tested probiotics through antioxidative and anti-inflammatory activities. These findings may be used to further understand the role of the tested probiotics in treating inflammation that underlies gut-related diseases. Such knowledge is essential for the development and translation of novel, targeted probiotic therapies to beneficially modulate the gut environment and reduce inflammation, improving host health.
Collapse
Affiliation(s)
- J.L. Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - A. Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy , Tanta University , Tanta , Egypt
| | - P. Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - A. Kassab
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - M. Santos
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - C. Shum-Tim
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - E. Renesteen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - S. Makhlouf
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - J. Chen
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| | - S. Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences , McGill University , Montreal, Quebec , Canada
| |
Collapse
|
10
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
11
|
Lv J, Lang G, Wang Q, Zhao W, Shi D, Zhou Z, Shen Y, Xia H, Han S, Li L. Lactobacillus helveticus attenuates alcoholic liver injury via regulation of gut microecology in mice. Microb Biotechnol 2024; 17:e70016. [PMID: 39431804 PMCID: PMC11492535 DOI: 10.1111/1751-7915.70016] [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: 12/25/2023] [Accepted: 09/04/2024] [Indexed: 10/22/2024] Open
Abstract
Previous reports have demonstrated that alcohol consumption significantly reduces the abundance of Lactobacillus in the gut. In this study, we selected five species of the genus Lactobacillus, commonly found in fermented foods, and acknowledged them as safe, edible, and effective in preventing or treating certain diseases, to evaluate their effects on alcoholic liver disease (ALD). By comparing the liver damage indices in each group, we found that the type strain of Lactobacillus helveticus (LH, ATCC 15009) had the most marked alleviating effect on ALD-induced liver injury. Furthermore, experiments combining microbiomics and metabolomics were conducted to explore the mechanisms underlying the hepatoprotective effects of LH. Finally, we discovered that LH mitigated ethanol-induced liver steatosis and inflammation in ALD mice by altering the structure and function of the gut microbiome, increasing intestinal levels of short-chain fatty acids (SCFAs), and enhancing gut barrier integrity. These findings suggest a potential strategy for the clinical management of patients with ALD.
Collapse
Affiliation(s)
- Jiawen Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Guanjing Lang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Wenlong Zhao
- Beijing Tsinghua Changgung Hospital, School of Clinical MedicineTsinghua UniversityBeijingChina
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Ziyuan Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Yangfan Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- College of MedicineZhejiang UniversityHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
12
|
Borah M, Mandal M, Konwar BK. Characterization of probiotic strains of Bacillus sp. from fermented palm wine (Nypa fructicans sp.) and exploration of cellulolytic potential for use as an addition in animal feed. Int Microbiol 2024:10.1007/s10123-024-00589-5. [PMID: 39289261 DOI: 10.1007/s10123-024-00589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
The main objective of this study was to assess cellulolytic probiotic strains from traditional fermented beverages such as palm wine in order to supplement the animal feed and strengthen the gut health of the animal for better digestibility and absorption. In the present study, different types of microbes were isolated from traditionally prepared palm wine and analyzed for their probiotic nature. For any microbe to be probiotic in nature, it has to sustain the harsh conditions of the human gastrointestinal tract such as acid tolerance, bile tolerance at the lower range of pH, and other properties like auto aggregation test, cell surface hydrophobicity test with non-polar hydrocarbons for evaluating its capabilities to adhere to the intestinal cells and antimicrobial nature against pathogens. Bacillus mycoides strain PR04 and Bacillus subtilis strain PR21 were found to be resistant to acid and bile in simulated artificial gastrointestinal tract model, found to be than 55% hydrophobic with xylene and n-hexadecane and also showed antimicrobial activity greater towards pathogenic strains like Pseudomonas aeruginosa, Staphylococcus aureus, Candida albicans, and Salmonella typhimurium respectively. The cellulolytic activity of the isolates PR04 and PR21 was evaluated in (0.2-2) % CMC (carboxymethyl cellulose) plate. Bacillus mycoides PR04 and Bacillus subtilis PR21 could degrade carboxymethyl cellulose, filter paper, and sugarcane bagasse. The degradation of sugarcane bagasse was confirmed by Scanning electron microscopy and filter paper degradation after 4 days of incubation at 37 °C. Cellulase gene of the identified Bacillus sp. strains was amplified by primers CF5'-ACAGGATCCGATGAAAACGGTCAATTTCTATTTT-3' and CR5'-ACTCTCGAGATTGGGTTCTGTTCCCAAT-3'. This study proposes potential probiotic Bacillus mycoides PR04 (Accession no. OR625070) and Bacillus subtilis PR21 (Accession no. OR625072) in the application as an animal feed additive to assist in its digestibility and encourage the gut health.
Collapse
Affiliation(s)
- Munmi Borah
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India.
| | - Manabendra Mandal
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India
| | - Bolin K Konwar
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India
| |
Collapse
|
13
|
Phùng TTT, Dupont S, Beney L, Moundanga S, Denimal E, Hồ PH, Karbowiak T. Ex-vivo investigation of probiotic bacterial adhesion to the intestinal mucus. Heliyon 2024; 10:e36339. [PMID: 39253206 PMCID: PMC11382035 DOI: 10.1016/j.heliyon.2024.e36339] [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: 02/19/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Recent research has promoted considerable interest in the potential health benefits of the new generation of probiotics. Despite the abundance of probiotic supplements, their adhesion and thereby colonization in the intestinal tract of the host, a determining factor of probiotic efficacy, remains questionable. Indeed, the gastrointestinal tract, a multi-component and complex system, obscures the comprehensive understanding of the probiotic adhesion mechanism. This study aimed to investigate the adhesion capacity of probiotic bacteria using two ex-vivo approaches that were specifically developed to investigate the bacteria-mucus agglomeration and the viable adhesion to intestinal mucus. Five probiotic bacterial strains including Escherichia coli, Lactiplantibacillus plantarum, Faecalibacterium duncaniae, Bifidobacterium longum, and Bifidobacterium longum str. infantis were selected for the investigation. In that context, higher adhesion to mucus was demonstrated by E. coli, L. plantarum, and B. infantis, emphasizing strain-specific differences. While total agglomeration capacity ranged from 8 % to 82 %, actual viable adhesion to mucus remained rather low (0.6 %-2.9 %). SEM images revealed that morphological characteristics, chain and/or cluster forming ability, as well as the presence of surface exopolysaccharides, might have an impact on bacterial adhesion. This study contributes knowledge on probiotic adhesion as well as simple and effective ex-vivo approaches to investigate the bacterial adhesion to the intestinal mucus, which is prerequisite for further colonization in the gut of the host.
Collapse
Affiliation(s)
- Thị-Thanh-Trúc Phùng
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Sébastien Dupont
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Laurent Beney
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Sylvie Moundanga
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Emmanuel Denimal
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| | - Phú-Hà Hồ
- Hanoi University of Science and Technology, School of Chemistry and Life Science, 1 Dai Co Viet Road, Hanoi, Viet Nam
| | - Thomas Karbowiak
- Université Bourgogne Franche-Comté, Institut Agro, Université de Bourgogne, INRAe, UMR PAM 1517, 1 Esplanade Erasme, 21000, Dijon, France
| |
Collapse
|
14
|
Nguyen HM, V Le KT, Nguyen NL, Tran-Van H, Ho GT, Nguyen TT, Haltrich D, Nguyen TH. Surface-Displayed Mannanolytic and Chitinolytic Enzymes Using Peptidoglycan Binding LysM Domains. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12655-12664. [PMID: 38775266 DOI: 10.1021/acs.jafc.4c01938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Using Lactiplantibacillus plantarum as a food-grade carrier to create non-GMO whole-cell biocatalysts is gaining popularity. This work evaluates the immobilization yield of a chitosanase (CsnA, 30 kDa) from Bacillus subtilis and a mannanase (ManB, 40 kDa) from B. licheniformis on the surface of L. plantarum WCFS1 using either a single LysM domain derived from the extracellular transglycosylase Lp_3014 or a double LysM domain derived from the muropeptidase Lp_2162. ManB and CsnA were fused with the LysM domains of Lp_3014 or Lp_2162, produced in Escherichia coli and anchored to the cell surface of L. plantarum. The localization of the recombinant proteins on the bacterial cell surface was successfully confirmed by Western blot and flow cytometry analysis. The highest immobilization yields (44-48%) and activities of mannanase and chitosanase on the displaying cell surface (812 and 508 U/g of dry cell weight, respectively) were obtained when using the double LysM domain of Lp_2162 as an anchor. The presence of manno-oligosaccharides or chito-oligosaccharides in the reaction mixtures containing appropriate substrates and ManB or CsnA-displaying cells was determined by high-performance anion exchange chromatography. This study indicated that non-GMO Lactiplantibacillus chitosanase- and mannanase-displaying cells could be used to produce potentially prebiotic oligosaccharides.
Collapse
Affiliation(s)
- Hoang-Minh Nguyen
- Department of Biotechnology, Faculty of Chemical Engineering, The University of Da Nang─University of Science and Technology, 54 Nguyen Luong Bang, Da Nang 550000, Vietnam
| | - Khanh-Trang V Le
- Food Biotechnology Laboratory, Department of Food Science and Technology, BOKU-University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
- Faculty of Biology and Environmental Science, The University of Da Nang - University of Science and Education, Da Nang 550000, Vietnam
| | - Ngoc-Luong Nguyen
- Hue University, College of Sciences, 77 Nguyen Hue, Hue 70000, Vietnam
| | - Hieu Tran-Van
- Laboratory of Biosensors, Faculty of Biology and Biotechnology, University of Science, Vietnam National University, Ho Chi Minh City 70000, Vietnam
| | - Giap T Ho
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh Street, District 4, Ho Chi Minh City 700000, Vietnam
| | - Tien-Thanh Nguyen
- School of Chemistry and Life Sciences, Hanoi University of Science and Technology, No.1 Dai Co Viet, Hai Ba Trung, Hanoi 100000, Vietnam
| | - Dietmar Haltrich
- Food Biotechnology Laboratory, Department of Food Science and Technology, BOKU-University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| | - Thu-Ha Nguyen
- Food Biotechnology Laboratory, Department of Food Science and Technology, BOKU-University of Natural Resources and Life Sciences, Muthgasse 18, A-1190 Vienna, Austria
| |
Collapse
|
15
|
Zhang S, Hung J, Yen TN, Huang S. Mutualistic interactions of lactate-producing lactobacilli and lactate-utilizing Veillonella dispar: Lactate and glutamate cross-feeding for the enhanced growth and short-chain fatty acid production. Microb Biotechnol 2024; 17:e14484. [PMID: 38801349 PMCID: PMC11129673 DOI: 10.1111/1751-7915.14484] [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: 01/28/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The human gut hosts numerous ecological niches for microbe-microbe and host-microbe interactions. Gut lactate homeostasis in humans is crucial and relies on various bacteria. Veillonella spp., gut lactate-utilizing bacteria, and lactate-producing bacteria were frequently co-isolated. A recent clinical trial has revealed that lactate-producing bacteria in humans cross-feed lactate to Veillonella spp.; however, their interspecies interaction mechanisms remain unclear. Veillonella dispar, an obligate anaerobe commonly found in the human gut and oral cavity, ferments lactate into acetate and propionate. In our study, we investigated the interaction between V. dispar ATCC 17748T and three representative phylogenetically distant strains of lactic acid bacteria, Lactobacillus acidophilus ATCC 4356T, Lacticaseibacillus paracasei subsp. paracasei ATCC 27216T, and Lactiplantibacillus plantarum ATCC 10241. Bacterial growth, viability, metabolism and gene level adaptations during bacterial interaction were examined. V. dispar exhibited the highest degree of mutualism with L. acidophilus. During co-culture of V. dispar with L. acidophilus, both bacteria exhibited enhanced growth and increased viability. V. dispar demonstrated an upregulation of amino acid biosynthesis pathways and the aspartate catabolic pathway. L. acidophilus also showed a considerable number of upregulated genes related to growth and lactate fermentation. Our results support that V. dispar is able to enhance the fermentative capability of L. acidophilus by presumably consuming the produced lactate, and that L. acidophilus cross-feed not only lactate, but also glutamate, to V. dispar during co-culture. The cross-fed glutamate enters the central carbon metabolism in V. dispar. These findings highlight an intricate metabolic relationship characterized by cross-feeding of lactate and glutamate in parallel with considerable gene regulation within both L. acidophilus (lactate-producing) and V. dispar (lactate-utilizing). The mechanisms of mutualistic interactions between a traditional probiotic bacterium and a potential next-generation probiotic bacterium were elucidated in the production of short-chain fatty acids.
Collapse
Affiliation(s)
- Shi‐Min Zhang
- Program in Molecular MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jia‐He Hung
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Tran Ngoc Yen
- Institute of Microbiology and ImmunologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Shir‐Ly Huang
- Institute of Microbiology and ImmunologyNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
16
|
Wei X, Tsai MS, Liang L, Jiang L, Hung CJ, Jelliffe-Pawlowski L, Rand L, Snyder M, Jiang C. Vaginal microbiomes show ethnic evolutionary dynamics and positive selection of Lactobacillus adhesins driven by a long-term niche-specific process. Cell Rep 2024; 43:114078. [PMID: 38598334 DOI: 10.1016/j.celrep.2024.114078] [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: 09/15/2023] [Revised: 03/01/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
The vaginal microbiome's composition varies among ethnicities. However, the evolutionary landscape of the vaginal microbiome in the multi-ethnic context remains understudied. We perform a systematic evolutionary analysis of 351 vaginal microbiome samples from 35 multi-ethnic pregnant women, in addition to two validation cohorts, totaling 462 samples from 90 women. Microbiome alpha diversity and community state dynamics show strong ethnic signatures. Lactobacillaceae have a higher ratio of non-synonymous to synonymous polymorphism and lower nucleotide diversity than non-Lactobacillaceae in all ethnicities, with a large repertoire of positively selected genes, including the mucin-binding and cell wall anchor genes. These evolutionary dynamics are driven by the long-term evolutionary process unique to the human vaginal niche. Finally, we propose an evolutionary model reflecting the environmental niches of microbes. Our study reveals the extensive ethnic signatures in vaginal microbial ecology and evolution, highlighting the importance of studying the host-microbiome ecosystem from an evolutionary perspective.
Collapse
Affiliation(s)
- Xin Wei
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Ming-Shian Tsai
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Liang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liuyiqi Jiang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China
| | - Chia-Jui Hung
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura Jelliffe-Pawlowski
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Larry Rand
- Department of Obstetrics, Gynecology & Reproductive Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Chao Jiang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
17
|
Angal A, Shidture S, Syed J, Tiwari DP, Dubey AK, Bhaduri A, Pujari R. In vitro adhesion and anti-inflammatory properties of Limosilactobacillus fermentum FS-10 isolated from infant fecal sample. Int Microbiol 2024; 27:227-238. [PMID: 37269431 DOI: 10.1007/s10123-023-00383-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/05/2023]
Abstract
In this study, seven strains of Limosilactobacillus fermentum were isolated from an infant fecal sample and characterized using in vitro studies. Lactobacillus rhamnosus GG was used as a comparison because it is a well-documented commercial probiotic. The isolates were tested for attributes such as acid and phenol tolerance, bile salt hydrolase (BSH) activity, and antibiotic sensitivity. One isolate, L. fermentum FS-10, displayed enhanced cell surface hydrophobicity (> 85%) and mucin adhesion. Mucin-binding helps colonization in the gut. The immunomodulatory property of L. fermentum FS-10 was evaluated by determining the modulation of pro- and anti-inflammatory factors such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-10, and nitric oxide (NO) in human acute monocytic leukemia (THP-1) cells under inflammatory conditions induced by lipopolysaccharide (LPS). L. fermentum FS-10 potently downregulated the expression of TNF-α and nitric oxide and upregulated IL-10 levels, indicating an anti-inflammatory response. Safety assessment of the strain revealed the absence of genes for virulence factors, toxin production, and antibiotic resistance, potentiating application as a probiotic strain.
Collapse
Affiliation(s)
- Ashvini Angal
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Shubham Shidture
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Jaserah Syed
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Deepika Pandey Tiwari
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Ashok Kumar Dubey
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Anirban Bhaduri
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India
| | - Radha Pujari
- Tata Chemicals Limited, Survey No 315, Hissa No 1-14, Paud Rd, Darawali, Pune, Maharashtra, 412111, India.
| |
Collapse
|
18
|
Kolesnyk PO, Paliy IH, Sydorchuk LP, Hoda ZP, Ivanchenko NO, Lych OS, Huley NR, Matsyura OI, Slyuzar ZL, Gerasymov SV. The role of nutritional support with probiotics in outpatients with symptomatic acute respiratory tract infections: a multicenter, randomized, double-blind, placebo-controlled dietary study. BMC Nutr 2024; 10:4. [PMID: 38178223 PMCID: PMC10768308 DOI: 10.1186/s40795-023-00816-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/22/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND A number of laboratory data and clinical studies have shown that probiotic bacteria may be beneficial in respiratory viral diseases. We investigated the role of probiotics in coronavirus disease-19 (COVID -19), post-disease symptoms, and humoral immune responses to viral antigens. METHODS This was a randomized, double-blind, placebo-controlled, prospective, multicenter study. We included symptomatic patients aged 18-65 years without risk of severe disease, and positive antigen/PCR test for SARS-CoV-2. Patients received (Bifidobacterium (B.) lactis BI040, B. longum BL020, Lactobacillus (L) rhamnosus LR110, L. casei LC130, L. acidophilus LA120, 5 billion CFU total) or placebo 1 capsule a day for 28 days and recorded symptoms. Three months later patients completed Post-COVID-19 Questionnaire (PCQ-19). On days 0-5 and 28-35, blood was sampled for IgG to nucleocapsid protein (NCP) and receptor binding domain (RBD)/spike 1 (S1) protein. The primary outcome measure was a patient global symptom score on day 10 of observation. The difference between groups was assessed using the Mann-Whitney U test. RESULTS Seventy-three patients were assessed for clinical endpoints and 44 patients were evaluated for antibody production. At day 10, the median global symptom score (interquartile range) was lower in the probiotic group (0.0 (0.0-2.0) vs. 2.0 (1.0-5.0), P < 0.05). The probiotic group had a shorter duration of fatigue and anxiety after COVID -19 (P < 0.05) and a greater change in IgG concentration on RBD/S1 (225.9 vs. 105.6 binding antibody units/mL, P < 0.05). CONCLUSIONS Use of probiotics alleviates acute and post-disease symptoms, and improves humoral immune response to viral antigens. TRIAL REGISTRATION Registered at clinicaltrials.gov as NCT04907877, June 1, 2021.
Collapse
Affiliation(s)
- Pavlo O Kolesnyk
- Family Medicine and Outpatient Care Department, Uzhgorod National University, Uzhgorod, Ukraine
| | - Iryna H Paliy
- Department of Internal and Family Medicine, National Pirogov Memorial Medical University, Vinnytsia, Ukraine
| | - Larysa P Sydorchuk
- Family Medicine Department, Bukovinian State Medical University, Chernivtsi, Ukraine
| | - Zoriana P Hoda
- Lviv State Center for Disease Control and Prevention of Ministry of Health of Ukraine, Lviv, Ukraine
| | - Nataliya O Ivanchenko
- Lviv State Center for Disease Control and Prevention of Ministry of Health of Ukraine, Lviv, Ukraine
| | - Oksana S Lych
- Lviv State Center for Disease Control and Prevention of Ministry of Health of Ukraine, Lviv, Ukraine
| | - Natalia R Huley
- Lviv Municipal Non-Profit Enterprise Third City Clinical Hospital, Lviv, Ukraine
| | | | | | - Sergiy V Gerasymov
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine.
- MedianaStatistics, CRO, Mykhaila Horynia Str. 15-A, Lviv, 79012, Ukraine.
| |
Collapse
|
19
|
Forouhandeh H, Soofiyani SR, Hosseini K, Beirami SM, Ahangari H, Moammer Y, Ebrahimzadeh S, Nejad MK, Farjami A, Khodaiefar F, Tarhriz V. Modulation of the Immune System Mechanisms using Probiotic Bacteria in Allergic Diseases: Focus on Allergic Retinitis and Food Allergies. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2024; 18:11-26. [PMID: 37842889 DOI: 10.2174/0127722708246899230928080651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 10/17/2023]
Abstract
Allergic illnesses occur when an organism's immune system is excessively responsive to certain antigens, such as those that are presented in the environment. Some people suffer from a wide range of immune system-related illnesses including allergic rhinitis, asthma, food allergies, hay fever, and even anaphylaxis. Immunotherapy and medications are frequently used to treat allergic disorders. The use of probiotics in bacteriotherapy has lately gained interest. Probiotics are essential to human health by modulating the gut microbiota in some ways. Due to probiotics' immunomodulatory properties present in the gut microbiota of all animals, including humans, these bacterial strains can prevent a wide variety of allergic disorders. Probiotic treatment helps allergy patients by decreasing inflammatory cytokines and enhancing intestinal permeability, which is important in the battle against allergy. By altering the balance of Th1 and Th2 immune responses in the intestinal mucosa, probiotics can heal allergic disorders. Numerous studies have shown a correlation between probiotics and a reduced risk of allergy disorders. A wide range of allergic disorders, including atopic dermatitis, asthma, allergic retinitis and food allergies has been proven to benefit from probiotic bacteria. Therefore, the use of probiotics in the treatment of allergic diseases offers a promising perspective. Considering that probiotic intervention in the treatment of diseases is a relatively new field of study, more studies in this regard seem necessary.
Collapse
Affiliation(s)
- Haleh Forouhandeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saiedeh Razi Soofiyani
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sohrab Minaei Beirami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Ahangari
- Department of Food Science and Technology, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yusif Moammer
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Ebrahimzadeh
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoomeh Kashef Nejad
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Farjami
- Food and Drug Safety Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Khodaiefar
- Department of Traditional Medicine, Faculty of Traditional Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
20
|
Müller M, Drexel R, Burkhart M, Dähnhardt-Pfeiffer S, Wien L, Herrmann C, Knoll T, Metzger C, Briesen H, Wagner S, Meier F, Kohl Y. Ex vivo models for intestinal translocation studies of cellulose nanocrystals. IN VITRO MODELS 2023; 2:181-194. [PMID: 39872170 PMCID: PMC11756450 DOI: 10.1007/s44164-023-00056-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 01/29/2025]
Abstract
Purpose Cellulose nanocrystals (CNC) play a promising role in the development of new advanced materials. The growing demand of CNC-containing products in the food industry will lead to an increased human exposure through oral uptake. To date, there is a dearth of studies reporting on the risks which CNC pose to human health following ingestion. In vitro models, which lack physiological accuracy, are often used to justify animal experiments in the field of nanosafety assessment. Nevertheless, ex vivo models of the intestine pose promising alternatives to in vivo experiments. Methods Two ex vivo models, a microfluidic chip based on porcine intestinal mucus and the Ussing chamber apparatus with tissue from abattoirs, which aim to complement in vitro models, are characterized by investigating the transport and toxicity of CNC through them in comparison to an in vitro triple co-culture model. Silver nanoparticles were included in this study as well-known and characterized nanomaterials for comparative purposes. Results Study results show that CNC cross the intestinal mucus layer but do not pass the intestinal tissue barrier ex vivo and in vitro; furthermore, no toxic effects were observed under exposure conditions tested. Conclusion These ex vivo models present complementary methods to the existing standardized in vitro and in silico methods to support data generation under physiologically relevant conditions without the use of animals. This multi-model approach offers an enhanced understanding of the complex interaction between new materials and human tissue and aligns with the flexible approach of IATA (Integrated Approaches to Testing and Assessment) and NAMs (New Approach Methods) for chemical and drug safety assessment. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-023-00056-x.
Collapse
Affiliation(s)
- Michelle Müller
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Roland Drexel
- Postnova Analytics GmbH, Rankinestr. 1, 86899 Landsberg am Lech, Germany
| | - Marie Burkhart
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | | | - Lena Wien
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Christine Herrmann
- Process Systems Engineering, School of Life Sciences, Technical University Munich, Gregor-Mendel-Str. 4, 85354 Freising, Germany
| | - Thorsten Knoll
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Christoph Metzger
- Process Systems Engineering, School of Life Sciences, Technical University Munich, Gregor-Mendel-Str. 4, 85354 Freising, Germany
| | - Heiko Briesen
- Process Systems Engineering, School of Life Sciences, Technical University Munich, Gregor-Mendel-Str. 4, 85354 Freising, Germany
| | - Sylvia Wagner
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| | - Florian Meier
- Postnova Analytics GmbH, Rankinestr. 1, 86899 Landsberg am Lech, Germany
| | - Yvonne Kohl
- Department Bioprocessing & Bioanalytics, Fraunhofer Institute for Biomedical Engineering IBMT, Joseph-von-Fraunhofer-Weg 1, 66280 Sulzbach, Germany
| |
Collapse
|
21
|
Antony MA, Chowdhury A, Edem D, Raj R, Nain P, Joglekar M, Verma V, Kant R. Gut microbiome supplementation as therapy for metabolic syndrome. World J Diabetes 2023; 14:1502-1513. [PMID: 37970133 PMCID: PMC10642415 DOI: 10.4239/wjd.v14.i10.1502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/31/2023] [Accepted: 08/28/2023] [Indexed: 10/09/2023] Open
Abstract
The gut microbiome is defined as an ecological community of commensal symbiotic and pathogenic microorganisms that exist in our body. Gut microbiome dysbiosis is a condition of dysregulated and disrupted intestinal bacterial homeostasis, and recent evidence has shown that dysbiosis is related to chronic inflammation, insulin resistance, cardiovascular diseases (CVD), type 2 diabetes mellitus (T2DM), and obesity. It is well known that obesity, T2DM and CVD are caused or worsened by multiple factors like genetic predisposition, environmental factors, unhealthy high calorie diets, and sedentary lifestyle. However, recent evidence from human and mouse models suggest that the gut microbiome is also an active player in the modulation of metabolic syndrome, a set of risk factors including obesity, hyperglycemia, and dyslipidemia that increase the risk for CVD, T2DM, and other diseases. Current research aims to identify treatments to increase the number of beneficial microbiota in the gut microbiome in order to modulate metabolic syndrome by reducing chronic inflammation and insulin resistance. There is increasing interest in supplements, classified as prebiotics, probiotics, synbiotics, or postbiotics, and their effect on the gut microbiome and metabolic syndrome. In this review article, we have summarized current research on these supplements that are available to improve the abundance of beneficial gut microbiota and to reduce the harmful ones in patients with metabolic syndrome.
Collapse
Affiliation(s)
- Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Aniqa Chowdhury
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Dinesh Edem
- Department of Endocrinology, Diabetes and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AK 72205, United States
| | - Rishi Raj
- Department of Endocrinology, Diabetes and Metabolism, Pikeville Medical Center, Pikeville, KY 41501, United States
| | - Priyanshu Nain
- Department of Graduate Medical Education, Maulana Azad Medical College, Delhi 110002, India
| | - Mansi Joglekar
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| | - Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina/AnMed Campus, Anderson, SC 29621, United States
| |
Collapse
|
22
|
Jang YJ, Min B, Lim JH, Kim BY. In Vitro Evaluation of Probiotic Properties of Two Novel Probiotic Mixtures, Consti-Biome and Sensi-Biome. J Microbiol Biotechnol 2023; 33:1149-1161. [PMID: 37386724 PMCID: PMC10580887 DOI: 10.4014/jmb.2303.03011] [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: 03/09/2023] [Revised: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 07/01/2023]
Abstract
Changes in the gut microbiome cause recolonization by pathogens and inflammatory responses, leading to the development of intestinal disorders. Probiotics administration has been proposed for many years to reverse the intestinal dysbiosis and to enhance intestinal health. This study aimed to evaluate the inhibitory effects of two newly designed probiotic mixtures, Consti-Biome and Sensi-Biome, on two enteric pathogens Staphylococcus aureus and Escherichia coli that may cause intestinal disorders. Additionally, the study was designed to evaluate whether Consti-Biome and Sensi-Biome could modulate the immune response, produce short-chain fatty acids (SCFAs), and reduce gas production. Consti-Biome and Sensi-Biome showed superior adhesion ratios to HT-29 cells and competitively suppressed pathogen adhesion. Moreover, the probiotic mixtures decreased the levels of pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-6 and IL-1β. Cell-free supernatants (CFSs) were used to investigate the inhibitory effects of metabolites on growth and biofilms of pathogens. Consti-Biome and Sensi-Biome CFSs exhibited antimicrobial and anti-biofilm activity, where microscopic analysis confirmed an increase in the number of dead cells and the structural disruption of pathogens. Gas chromatographic analysis of the CFSs revealed their ability to produce SCFAs, including acetic, propionic, and butyric acid. SCFA secretion by probiotics may demonstrate their potential activities against pathogens and gut inflammation. In terms of intestinal symptoms regarding abdominal bloating and discomfort, Consti-Biome and Sensi-Biome also inhibited gas production. Thus, these two probiotic mixtures have great potential to be developed as dietary supplements to alleviate the intestinal disorders.
Collapse
Affiliation(s)
- You Jin Jang
- R&D Center, Chong Kun Dang Healthcare, Seoul 07249, Republic of Korea
| | - Bonggyu Min
- R&D Center, Chong Kun Dang Healthcare, Seoul 07249, Republic of Korea
| | - Jong Hyun Lim
- R&D Center, Chong Kun Dang Healthcare, Seoul 07249, Republic of Korea
| | - Byung-Yong Kim
- R&D Center, Chong Kun Dang Healthcare, Seoul 07249, Republic of Korea
| |
Collapse
|
23
|
Calvigioni M, Panattoni A, Biagini F, Donati L, Mazzantini D, Massimino M, Daddi C, Celandroni F, Vozzi G, Ghelardi E. Development of an In Vitro Model of the Gut Microbiota Enriched in Mucus-Adhering Bacteria. Microbiol Spectr 2023; 11:e0033623. [PMID: 37289064 PMCID: PMC10433972 DOI: 10.1128/spectrum.00336-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023] Open
Abstract
Culturing the gut microbiota in in vitro models that mimic the intestinal environment is increasingly becoming a promising alternative approach to study microbial dynamics and the effect of perturbations on the gut community. Since the mucus-associated microbial populations in the human intestine differ in composition and functions from their luminal counterpart, we attempted to reproduce in vitro the microbial consortia adhering to mucus using an already established three-dimensional model of the human gut microbiota. Electrospun gelatin structures supplemented or not with mucins were inoculated with fecal samples and compared for their ability to support microbial adhesion and growth over time, as well as to shape the composition of the colonizing communities. Both scaffolds allowed the establishment of long-term stable biofilms with comparable total bacterial loads and biodiversity. However, mucin-coated structures harbored microbial consortia especially enriched in Akkermansia, Lactobacillus, and Faecalibacterium, being therefore able to select for microorganisms commonly considered mucosa-associated in vivo. IMPORTANCE These findings highlight the importance of mucins in shaping intestinal microbial communities, even those in artificial gut microbiota systems. We propose our in vitro model based on mucin-coated electrospun gelatin structures as a valid device for studies evaluating the effects of exogenous factors (nutrients, probiotics, infectious agents, and drugs) on mucus-adhering microbial communities.
Collapse
Affiliation(s)
- Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Adelaide Panattoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Biagini
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Leonardo Donati
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mariacristina Massimino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Costanza Daddi
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Research Center “Nutraceuticals and Food for Health – Nutrafood”, University of Pisa, Pisa, Italy
| |
Collapse
|
24
|
Kochetkov N, Smorodinskaya S, Vatlin A, Nikiforov-Nikishin D, Nikiforov-Nikishin A, Danilenko V, Anastasia K, Reznikova D, Grishina Y, Antipov S, Marsova M. Ability of Lactobacillus brevis 47f to Alleviate the Toxic Effects of Imidacloprid Low Concentration on the Histological Parameters and Cytokine Profile of Zebrafish ( Danio rerio). Int J Mol Sci 2023; 24:12290. [PMID: 37569666 PMCID: PMC10418720 DOI: 10.3390/ijms241512290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
In the present article, the possible mitigation of the toxic effect of imidacloprid low-concentration chronic exposure on Danio rerio by the probiotic strain Lactobacillus brevis 47f (1 × 108 CFU/g) was examined. It was found that even sublethal concentration (2500 µg/L) could lead to the death of some fish during the 60-day chronic experiment. However, the use of Lactobacillus brevis 47f partially reduced the toxic effects, resulting in an increased survival rate and a significant reduction of morphohistological lesions in the intestines and kidneys of Danio rerio. The kidneys were found to be the most susceptible organ to toxic exposure, showing significant disturbances. Calculation of the histopathological index, measurement of morphometric parameters, and analysis of principal components revealed the most significant parameters affected by the combined action of imidacloprid and Lactobacillus brevis 47f. This effect of imidacloprid and the probiotic strain had a multidirectional influence on various pro/anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-8). Therefore, the results suggest the possibility of further studying the probiotic strain Lactobacillus brevis 47f as a strain that reduces the toxic effects of xenobiotics. Additionally, the study established the possibility of using imidacloprid as a model toxicant to assess the detoxification ability of probiotics on the kidney and gastrointestinal tract of fish.
Collapse
Affiliation(s)
- Nikita Kochetkov
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Svetlana Smorodinskaya
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Aleksey Vatlin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Dmitry Nikiforov-Nikishin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Alexei Nikiforov-Nikishin
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Valery Danilenko
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Klimuk Anastasia
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Diana Reznikova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky Lane 9, 141700 Dolgoprudny, Russia
| | - Yelena Grishina
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Sergei Antipov
- Department of Biophysics and Biotechnology, Voronezh State University, University Square, 1, 394063 Voronezh, Russia;
| | - Maria Marsova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| |
Collapse
|
25
|
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
|
26
|
Bnfaga AA, Lee KW, Than LTL, Amin-Nordin S. Antimicrobial and immunoregulatory effects of Lactobacillus delbrueckii 45E against genitourinary pathogens. J Biomed Sci 2023; 30:19. [PMID: 36959635 PMCID: PMC10037868 DOI: 10.1186/s12929-023-00913-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND Lactobacilli are essential microbiota that maintain a healthy, balanced vaginal environment. Vaginitis is a common infection in women during their reproductive years. Many factors are associated with vaginitis; one of them is the imbalance of microbiota in the vaginal environment. This study aimed to evaluate the antimicrobial properties of Lactobacillus delbrueckii 45E (Ld45E) against several species of bacteria, namely, Group B Streptococcus (GBS), Escherichia coli, Klebsiella spp., and Candida parapsilosis, as well as to determine the concentration of interleukin-17 (IL-17) in the presence of Ld45E. METHODS The probiotic characteristics of Ld45E were evaluated by examining its morphology, pH tolerance, adhesive ability onto HeLa cells, hemolytic activity, antibiotic susceptibility, and autoaggregation ability. Then, the antimicrobial activity of Ld45E was determined using Ld45E culture, cell-free supernatant, and crude bacteriocin solution. Co-aggregation and competition ability assays against various pathogens were conducted. The immunoregulatory effects of Ld45E were analyzed by measuring the proinflammatory cytokine IL-17. A p-value less than 0.05 was considered statistical significance. RESULTS Ld45E is 3-5 mm in diameter and round with a flat-shaped colony. pH 4 and 4.5 were the most favorable range for Ld45E growth within 12 h of incubation. Ld45E showed a strong adhesion ability onto HeLa cells (86%) and negative hemolytic activities. Ld45E was also sensitive to ceftriaxone, cefuroxime, ciprofloxacin, and doxycycline. We found that it had a good autoaggregation ability of 80%. Regarding antagonistic properties, Ld45E culture showed strong antimicrobial activity against GBS, E. coli, and Klebsiella spp. but only a moderate effect on C. parapsilosis. Cell-free supernatant of Ld45E exerted the most potent inhibitory effects at 40 °C against all genital pathogens, whereas bacteriocin showed a robust inhibition at 37 °C and 40 °C. The highest co-aggregation affinity was observed with GBS (81%) and E. coli (40%). Competition ability against the adhesion of GBS (80%), E. coli (76%), Klebsiella (72%), and C. parapsilosis (58%) was found. Ld45E was able to reduce the induction of the proinflammatory protein IL-17. CONCLUSIONS Ld45E possessed antimicrobial and immunoregulatory properties, with better cell-on-cell activity than supernatant activity. Thus, Ld45E is a potential probiotic candidate for adjunct therapy to address vaginal infections.
Collapse
Affiliation(s)
- Ameda Abdullah Bnfaga
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department of Para-Clinic, Faculty of Medicine, Aden University, Aden, Yemen
| | - Kai Wei Lee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Syafinaz Amin-Nordin
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Hospital Sultan Abdul Aziz Shah, Universiti Putra Malaysia, Persiaran MARDI-UPM, 43400, Serdang, Malaysia.
| |
Collapse
|
27
|
Kemper L, Hensel A. Campylobacter jejuni: targeting host cells, adhesion, invasion, and survival. Appl Microbiol Biotechnol 2023; 107:2725-2754. [PMID: 36941439 PMCID: PMC10027602 DOI: 10.1007/s00253-023-12456-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/23/2023]
Abstract
Campylobacter jejuni, causing strong enteritis, is an unusual bacterium with numerous peculiarities. Chemotactically controlled motility in viscous milieu allows targeted navigation to intestinal mucus and colonization. By phase variation, quorum sensing, extensive O-and N-glycosylation and use of the flagellum as type-3-secretion system C. jejuni adapts effectively to environmental conditions. C. jejuni utilizes proteases to open cell-cell junctions and subsequently transmigrates paracellularly. Fibronectin at the basolateral side of polarized epithelial cells serves as binding site for adhesins CadF and FlpA, leading to intracellular signaling, which again triggers membrane ruffling and reduced host cell migration by focal adhesion. Cell contacts of C. jejuni results in its secretion of invasion antigens, which induce membrane ruffling by paxillin-independent pathway. In addition to fibronectin-binding proteins, other adhesins with other target structures and lectins and their corresponding sugar structures are involved in host-pathogen interaction. Invasion into the intestinal epithelial cell depends on host cell structures. Fibronectin, clathrin, and dynein influence cytoskeletal restructuring, endocytosis, and vesicular transport, through different mechanisms. C. jejuni can persist over a 72-h period in the cell. Campylobacter-containing vacuoles, avoid fusion with lysosomes and enter the perinuclear space via dynein, inducing signaling pathways. Secretion of cytolethal distending toxin directs the cell into programmed cell death, including the pyroptotic release of proinflammatory substances from the destroyed cell compartments. The immune system reacts with an inflammatory cascade by participation of numerous immune cells. The development of autoantibodies, directed not only against lipooligosaccharides, but also against endogenous gangliosides, triggers autoimmune diseases. Lesions of the epithelium result in loss of electrolytes, water, and blood, leading to diarrhea, which flushes out mucus containing C. jejuni. Together with the response of the immune system, this limits infection time. Based on the structural interactions between host cell and bacterium, the numerous virulence mechanisms, signaling, and effects that characterize the infection process of C. jejuni, a wide variety of targets for attenuation of the pathogen can be characterized. The review summarizes strategies of C. jejuni for host-pathogen interaction and should stimulate innovative research towards improved definition of targets for future drug development. KEY POINTS: • Bacterial adhesion of Campylobacter to host cells and invasion into host cells are strictly coordinated processes, which can serve as targets to prevent infection. • Reaction and signalling of host cell depend on the cell type. • Campylobacter virulence factors can be used as targets for development of antivirulence drug compounds.
Collapse
Affiliation(s)
- Leon Kemper
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Corrensstraße 48, 48149, Münster, Germany.
| |
Collapse
|
28
|
Hammouda ZK, Wasfi R, Abdeltawab NF. Hormonal drugs: Influence on growth, biofilm formation, and adherence of selected gut microbiota. Front Cell Infect Microbiol 2023; 13:1147585. [PMID: 36992682 PMCID: PMC10042233 DOI: 10.3389/fcimb.2023.1147585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 02/17/2023] [Indexed: 03/14/2023] Open
Abstract
Many studies have reported the influence of hormonal drugs on gut microbiota composition. However, the underlying mechanism of this interaction is still under study. Therefore, this study aimed to evaluate the possible in vitro changes in selected members of gut bacteria exposed to oral hormonal drugs used for years. Selected members of gut bacteria were Bifidobacterium longum, Limosilactobacillus reuteri, Bacteroides fragilis, and Escherichia coli representing the four main phyla in the gut. Selected hormonal drugs used for a long time were estradiol, progesterone, and thyroxine. The effect of intestinal concentrations of these drugs on the selected bacterial growth, biofilm formation, and adherence to Caco-2/HT-29 cell line was assessed. Short-chain fatty acids (SCFAs) have been included in host functions including the gut, immune and nervous functions; thus, the drug’s effects on their production were assayed using High- Performance Liquid Chromatography. Sex steroids significantly increased the growth of all tested bacteria except B. longum, similarly, thyroxine increased the growth of tested Gram-negative bacteria however reducing that of tested Gram-positive bacteria. The effect of drugs on biofilm formation and bacterial adherence to cell lines cocultures was variable. Progesterone decreased the biofilm formation of tested Gram-positive bacteria, it nevertheless increased L. reuteri adherence to Caco-2/HT-29 cell line cell lines coculture. By contrast, progesterone increased biofilm formation by Gram-negative bacteria and increased adherence of B. fragilis to the cell lines coculture. Moreover, thyroxine and estradiol exhibited antibiofilm activity against L. reuteri, while thyroxine increased the ability of E. coli to form a biofilm. Moreover, hormones affected bacterial adherence to cell lines independently of their effect on hydrophobicity suggesting other specific binding factors might contribute to this effect. Tested drugs affected SCFAs production variably, mostly independent of their effect on bacterial growth. In conclusion, our results showed that the microbiota signature associated with some hormonal drug consumption could be the result of the direct effect of these drugs on bacterial growth, and adherence to enterocytes besides the effect of these drugs on the host tissue targets. Additionally, these drugs affect the production of SCFAs which could contribute to some of the side effects of these drugs.
Collapse
Affiliation(s)
- Zainab K. Hammouda
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
| | - Reham Wasfi
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza, Egypt
- *Correspondence: Reham Wasfi,
| | - Nourtan F. Abdeltawab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
29
|
Grigaleviciute R, Planciuniene R, Prikockyte I, Radzeviciute-Valciuke E, Baleviciute A, Zelvys A, Zinkeviciene A, Zigmantaite V, Kucinskas A, Matusevicius P, Kavaliauskas P. The Influence of Feeding with Colostrum and Colostrum Replacer on Major Blood Biomarkers and Growth Performance in Dairy Calves. Vet Sci 2023; 10:vetsci10020128. [PMID: 36851432 PMCID: PMC9965887 DOI: 10.3390/vetsci10020128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Bovine colostrum (BC) is the first milk produced by lactating cows after parturition. BC is rich in various amino acids, proteins, and fats essential for the nutrition of the neonate calves. Despite the evident beneficial effect of BC on calves, the effect of BC on blood biomarkers is poorly understood. Calves that received BC showed significantly higher body mass at days 7 and 30 (38.54 kg and 43.42 kg, respectively) compared to the colostrum replacer group (p = 0.0064). BC induced greater quantities of blood neutrophils (0.27 × 109/L) and monocytes (4.76 × 109/L) in comparison to the colostrum replacer (0.08 and 0.06 × 109/L, respectively) (p = 0.0001). Animals that received BC showed higher levels of total serum protein (59.16 g/L) and albumin (29.96 g/L) in comparison to the colostrum replacer group (44.34 g/L and 31.58 g/L, respectively). In addition, BC induced greater intestinal mucus production in the Wistar rat model. Collectively, these results demonstrate that BC is important for the growth of calves and that it provides a significant beneficial effect on morphological and biochemical blood parameters.
Collapse
Affiliation(s)
- Ramune Grigaleviciute
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Str. 18/7, LT-47181 Kaunas, Lithuania
- Department of Animal Nutrition, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| | - Rita Planciuniene
- Institute of Microbiology and Virology, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| | - Ieva Prikockyte
- Kaunas Hospital, Lithuanian University of Health Sciences, Josvainiu Strg. 2, LT-47144 Kaunas, Lithuania
| | | | - Austeja Baleviciute
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solnavägen 1, 171 77 Solna, Sweden
| | - Augustinas Zelvys
- Department of Immunology, Centre for Innovative Medicine, Santariskiu Str. 5, LT-08410 Vilnius, Lithuania
| | - Aukse Zinkeviciene
- Department of Immunology, Centre for Innovative Medicine, Santariskiu Str. 5, LT-08410 Vilnius, Lithuania
| | - Vilma Zigmantaite
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Str. 18/7, LT-47181 Kaunas, Lithuania
| | - Audrius Kucinskas
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Str. 18/7, LT-47181 Kaunas, Lithuania
| | - Paulius Matusevicius
- Department of Animal Nutrition, Lithuanian University of Health Sciences, Tilzes Str. 18, LT-47181 Kaunas, Lithuania
| | - Povilas Kavaliauskas
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Str. 18/7, LT-47181 Kaunas, Lithuania
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell University, 1300 York Avenue, New York, NY 1109, USA
- Department of Microbiology and Immunology, University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birstono Str. 38A, LT-59116 Prienai, Lithuania
- Correspondence:
| |
Collapse
|
30
|
Wang P, Chen S, Liao C, Jia Y, Li J, Shang K, Chen J, Cao P, Li W, Li Y, Yu Z, Ding K. Probiotic Properties of Chicken-Derived Highly Adherent Lactic Acid Bacteria and Inhibition of Enteropathogenic Bacteria in Caco-2 Cells. Microorganisms 2022; 10:microorganisms10122515. [PMID: 36557770 PMCID: PMC9788042 DOI: 10.3390/microorganisms10122515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Lactic acid bacteria (LAB) as probiotic candidates have various beneficial functions, such as regulating gut microbiota, inhibiting intestinal pathogens, and improving gut immunity. The colonization of the intestine is a prerequisite for probiotic function. Therefore, it is necessary to screen the highly adherent LAB. In this study, the cell surface properties, such as hydrophobicity, auto-aggregation, co-aggregation, and adhesion abilities of the six chicken-derived LAB to Caco-2 cells were investigated. All six strains showed different hydrophobicity (21.18-95.27%), auto-aggregation (13.61-30.17%), co-aggregation with Escherichia coli ATCC 25922 (10.23-36.23%), and Salmonella enterica subsp. enterica serovar Typhimurium ATCC 13311 (11.71-39.35%), and adhesion to Caco-2 cells (8.57-26.37%). Pediococcus pentosaceus 2-5 and Lactobacillus reuteri L-3 were identified as the strains with strong adhesion abilities (26.37% and 21.57%, respectively). Moreover, these strains could survive in a gastric acid environment at pH 2, 3, and 4 for 3 h and in a bile salt environment at 0.1%, 0.2%, and 0.3% (w/v) concentration for 6 h. Furthermore, the cell-free supernatant of P. pentosaceus 2-5 and L. reuteri L-3 inhibited the growth of enteropathogenic bacteria and the strains inhibited the adhesion of these pathogens to Caco-2 cells. In this study, these results suggested that P. pentosaceus 2-5 and L. reuteri L-3, isolated from chicken intestines might be good probiotic candidates to be used as feed additives or delivery vehicles of biologically active substances.
Collapse
Affiliation(s)
- Pudi Wang
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Songbiao Chen
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Chengshui Liao
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Yanyan Jia
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Jing Li
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Ke Shang
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Jian Chen
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Pinghua Cao
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Wang Li
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Yuanxiao Li
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
| | - Zuhua Yu
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
- Correspondence: (Z.Y.); (K.D.)
| | - Ke Ding
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Luoyang 471003, China
- Laboratory of Functional Microbiology and Animal Health, Henan University of Science and Technology, Luoyang 471003, China
- Correspondence: (Z.Y.); (K.D.)
| |
Collapse
|
31
|
Mazzantini D, Calvigioni M, Celandroni F, Lupetti A, Ghelardi E. In vitro assessment of probiotic attributes for strains contained in commercial formulations. Sci Rep 2022; 12:21640. [PMID: 36517529 PMCID: PMC9751119 DOI: 10.1038/s41598-022-25688-z] [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: 08/06/2021] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Although probiotics are often indiscriminately prescribed, they are not equal and their effects on the host may profoundly differ. In vitro determination of the attributes of probiotics should be a primary concern and be performed even before clinical studies are designed. In fact, knowledge on the biological properties a microbe possesses is crucial for selecting the most suitable bacteriotherapy for each individual. Herein, nine strains (Bacillus clausii NR, OC, SIN, T, Bacillus coagulans ATCC 7050, Bifidobacterium breve DSM 16604, Limosilactobacillus reuteri DSM 17938, Lacticaseibacillus rhamnosus ATCC 53103, and Saccharomyces boulardii CNCM I-745) declared to be contained in six commercial formulations were tested for their ability to tolerate simulated intestinal conditions, adhere to mucins, and produce β-galactosidase, antioxidant enzymes, riboflavin, and D-lactate. With the exception of B. breve, all microbes survived in simulated intestinal fluid. L. rhamnosus was unable to adhere to mucins and differences in mucin adhesion were evidenced for L. reuteri and S. boulardii depending on oxygen levels. All microorganisms produced antioxidant enzymes, but only B. clausii, B. coagulans, B. breve, and L. reuteri synthesize β-galactosidase. Riboflavin secretion was observed for Bacillus species and L. rhamnosus, while D-lactate production was restricted to L. reuteri and L. rhamnosus. Our findings indicate that the analyzed strains possess different in vitro biological properties, thus highlighting the usefulness of in vitro tests as prelude for clinical research.
Collapse
Affiliation(s)
- Diletta Mazzantini
- grid.5395.a0000 0004 1757 3729Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marco Calvigioni
- grid.5395.a0000 0004 1757 3729Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Celandroni
- grid.5395.a0000 0004 1757 3729Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Antonella Lupetti
- grid.5395.a0000 0004 1757 3729Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- grid.5395.a0000 0004 1757 3729Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy ,grid.5395.a0000 0004 1757 3729Research Center Nutraceuticals and Food for Health-Nutrafood, University of Pisa, Pisa, Italy
| |
Collapse
|
32
|
Torres-Miranda A, Melis-Arcos F, Garrido D. Characterization and Identification of Probiotic Features in Lacticaseibacillus Paracasei Using a Comparative Genomic Analysis Approach. Probiotics Antimicrob Proteins 2022; 14:1211-1224. [PMID: 36201155 DOI: 10.1007/s12602-022-09999-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2022] [Indexed: 12/25/2022]
Abstract
Lacticaseibacillus paracasei species are widely used for their health-promoting properties in food and agricultural applications. These bacteria have been isolated from various habitats such as the oral cavity, cereals, vegetables, meats, and dairy products conferring them the ability to consume different carbohydrates. Two subspecies are recognized, Lacticaseibacillus paracasei subsp. paracasei and Lacticaseibacillus paracasei subsp. tolerans according to their acid production from carbohydrates. Some strains are currently used as probiotics. In this study, we performed a comparative genomic analysis of 181 genomes of the Lacticaseibacillus paracasei species to reveal genomic differences at the subspecies level and to reveal adaptive and probiotic features, and special emphasis is given to inulin consumption. No clear distinction at the subspecies level for L. paracasei was shown using a phylogenetic tree with orthologous genes from the core-genome set. In general, a good correlation was observed between genomic distance and isolation origin, suggesting that L. paracasei strains are adapted to their natural habitat, giving rise to genetic differences at the genomic level. A low frequency of undesirable characteristics such as plasmids, prophages, antibiotic resistance genes, absence of virulence factors, and frequent bacteriocin production supports these species being good candidates for use as probiotics. Lastly, we found that the inulin gene cluster in L. paracasei strains seems to differ slightly in the presence or absence of some genes but maintains a core defined by at least three fructose-PTS proteins, one hypothetical protein, and extracellular β-fructosidase. Finally, we conclude that further work has to be done for L. paracasei subspecies classification. Improving outgroup selection criteria is a key factor for their correct subspecies assignation.
Collapse
Affiliation(s)
- Alexis Torres-Miranda
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Catolica de Chile, Av. Vicuña Mackenna 4860, Santiago, Chile
| | - Felipe Melis-Arcos
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Catolica de Chile, Av. Vicuña Mackenna 4860, Santiago, Chile
| | - Daniel Garrido
- Department of Chemical and Bioprocess Engineering, School of Engineering, Pontificia Universidad Catolica de Chile, Av. Vicuña Mackenna 4860, Santiago, Chile.
| |
Collapse
|
33
|
Pang Y, Ermann Lundberg L, Mata Forsberg M, Ahl D, Bysell H, Pallin A, Sverremark-Ekström E, Karlsson R, Jonsson H, Roos S. Extracellular membrane vesicles from Limosilactobacillus reuteri strengthen the intestinal epithelial integrity, modulate cytokine responses and antagonize activation of TRPV1. Front Microbiol 2022; 13:1032202. [PMID: 36466671 PMCID: PMC9712456 DOI: 10.3389/fmicb.2022.1032202] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/24/2022] [Indexed: 09/05/2023] Open
Abstract
Bacterial extracellular membrane vesicles (MV) are potent mediators of microbe-host signals, and they are not only important in host-pathogen interactions but also for the interactions between mutualistic bacteria and their hosts. Studies of MV derived from probiotics could enhance the understanding of these universal signal entities, and here we have studied MV derived from Limosilactobacillus reuteri DSM 17938 and BG-R46. The production of MV increased with cultivation time and after oxygen stress. Mass spectrometry-based proteomics analyses revealed that the MV carried a large number of bacterial cell surface proteins, several predicted to be involved in host-bacteria interactions. A 5'-nucleotidase, which catalyze the conversion of AMP into the signal molecule adenosine, was one of these and analysis of enzymatic activity showed that L. reuteri BG-R46 derived MV exhibited the highest activity. We also detected the TLR2 activator lipoteichoic acid on the MV. In models for host interactions, we first observed that L. reuteri MV were internalized by Caco-2/HT29-MTX epithelial cells, and in a dose-dependent manner decreased the leakage caused by enterotoxigenic Escherichia coli by up to 65%. Furthermore, the MV upregulated IL-1β and IL-6 from peripheral blood mononuclear cells (PBMC), but also dampened IFN-γ and TNF-α responses in PBMC challenged with Staphylococcus aureus. Finally, we showed that MV from the L. reuteri strains have an antagonistic effect on the pain receptor transient receptor potential vanilloid 1 in a model with primary dorsal root ganglion cells from rats. In summary, we have shown that these mobile nanometer scale MV reproduce several biological effects of L. reuteri cells and that the production parameters and selection of strain have an impact on the activity of the MV. This could potentially provide key information for development of innovative and more efficient probiotic products.
Collapse
Affiliation(s)
- Yanhong Pang
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ludwig Ermann Lundberg
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia AB, Stockholm, Sweden
| | - Manuel Mata Forsberg
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - David Ahl
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Anton Pallin
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Eva Sverremark-Ekström
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Roger Karlsson
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Nanoxis Consulting AB, Gothenburg, Sweden
| | - Hans Jonsson
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia AB, Stockholm, Sweden
| |
Collapse
|
34
|
Trullàs C, Sewaka M, Rodkhum C, Chansue N, Boonanuntanasarn S, Kamble MT, Pirarat N. Effects of Jerusalem Artichoke ( Helianthus tuberosus) as a Prebiotic Supplement in the Diet of Red Tilapia ( Oreochromis spp.). Animals (Basel) 2022; 12:ani12202882. [PMID: 36290267 PMCID: PMC9598478 DOI: 10.3390/ani12202882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of the present study was to evaluate the effects of a Jerusalem artichoke-supplemented diet on the blood chemistry, growth performance, intestinal morphology, expression of antioxidant-related genes, and disease resistance against Aeromonas veronii challenge in juvenile red tilapia. A completely randomized design (CRD) was followed to feed red tilapias with three experimental diets: control, 5.0 g/kg JA-supplemented (JA5), or 10.0 g/kg JA-supplemented (JA10) diets in triplicates for 4 weeks. The results revealed that the growth performance, weight gain (WG), specific growth rate (SGR), and average daily gain (ADG) of fish fed diets JA5 and JA10 were significantly higher (p < 0.05) than those of fish fed the control diet. Fish fed the control diet had significantly higher T-bilirubin, D-bilirubin, and ALT in blood serum than fish fed JA5 and JA10, as well as higher BUN than fish fed JA5. The number of goblet cells in the proximal and distal parts of the intestine revealed that the number of acid, neutral, and double-staining mucous cells of fish fed diets JA5 and JA10 was significantly higher (p < 0.05) than in fish fed the control diet. The diets including the prebiotic (JA5 and JA10) were associated with a significant increase in the expression of gpx1 and gst antioxidant-related genes and disease resistance against A. veronii in juvenile red tilapia. Therefore, JA5 and JA10 can be employed as promising prebiotics for sustainable red tilapia farming.
Collapse
Affiliation(s)
- Clara Trullàs
- Wildlife, Exotic and Aquatic Animal Pathology Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Mariya Sewaka
- Faculty of Veterinary Science, Rajamangala University of Technology Srivijaya, Nakhon Si Thammarat 80240, Thailand
| | - Channarong Rodkhum
- Center of Excellence in Fish Diseases (CE FID), Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nantarika Chansue
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Surintorn Boonanuntanasarn
- Institute of Agricultural Technology, School of Animal Production Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Manoj Tukaram Kamble
- Wildlife, Exotic and Aquatic Animal Pathology Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nopadon Pirarat
- Wildlife, Exotic and Aquatic Animal Pathology Research Unit, Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence:
| |
Collapse
|
35
|
Subramanian DA, Langer R, Traverso G. Mucus interaction to improve gastrointestinal retention and pharmacokinetics of orally administered nano-drug delivery systems. J Nanobiotechnology 2022; 20:362. [PMID: 35933341 PMCID: PMC9356434 DOI: 10.1186/s12951-022-01539-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
Oral delivery of therapeutics is the preferred route of administration due to ease of administration which is associated with greater patient medication adherence. One major barrier to oral delivery and intestinal absorption is rapid clearance of the drug and the drug delivery system from the gastrointestinal (GI) tract. To address this issue, researchers have investigated using GI mucus to help maximize the pharmacokinetics of the therapeutic; while mucus can act as a barrier to effective oral delivery, it can also be used as an anchoring mechanism to improve intestinal residence. Nano-drug delivery systems that use materials which can interact with the mucus layers in the GI tract can enable longer residence time, improving the efficacy of oral drug delivery. This review examines the properties and function of mucus in the GI tract, as well as diseases that alter mucus. Three broad classes of mucus-interacting systems are discussed: mucoadhesive, mucus-penetrating, and mucolytic drug delivery systems. For each class of system, the basis for mucus interaction is presented, and examples of materials that inform the development of these systems are discussed and reviewed. Finally, a list of FDA-approved mucoadhesive, mucus-penetrating, and mucolytic drug delivery systems is reviewed. In summary, this review highlights the progress made in developing mucus-interacting systems, both at a research-scale and commercial-scale level, and describes the theoretical basis for each type of system.
Collapse
Affiliation(s)
- Deepak A Subramanian
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni Traverso
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Wu Y, Dong XY, Zhou XZ, Li ZS, Du YQ. Effects of probiotics on gastric microbiota and its precombination with quadruple regimen for Helicobacter pylori eradication. J Dig Dis 2022; 23:462-472. [PMID: 36208049 DOI: 10.1111/1751-2980.13138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVES This study aimed to assess the efficacy and safety of probiotics for Helicobacter pylori (H. pylori) eradication therapy as well as their effects on gastric microbiota. METHODS Patients who had failed H. pylori eradication therapy for at least twice were prospectively enrolled during March 2019 and March 2021. A 2-week administration of compound Lactobacillus acidophilus probiotic (1 g thrice daily) followed by a 10-day quadruple bismuth-containing H. pylori eradication therapy was administrated. Endoscopy was performed, and gastric biopsy samples were obtained for drug sensitivity testing and 16S rRNA gene sequencing before and after probiotic treatment. Eradication was evaluated by the 13 C-urea breath test at least 4 weeks after treatment completion. Adverse events (AEs) were recorded. RESULTS Thirty-seven patients were included. Probiotic supplementation had no effects on the diversity, community structure, and composition of gastric microbiota and no inhibition on H. pylori activity. However, it increased some of the beneficial bacteria such as Blautia, Dorea, and Roseburia (P < 0.05). The overall eradication rate was 97.3% (95% confidence interval [CI] 91.8%-100%). AEs, mainly dizziness, nausea, diarrhea, and chest distress, were detected in six individuals, all of which were resolved upon cessation of antibiotic administration. Dyspeptic symptoms were improved after probiotic supplementation and at treatment completion (both P < 0.001). CONCLUSIONS H. pylori-infected individuals might benefit from probiotics followed by a quadruple bismuth-containing eradication therapy. Further studies with large sample sizes are warranted.
Collapse
Affiliation(s)
- You Wu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiao Yang Dong
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xian Zhu Zhou
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhao Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yi Qi Du
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
37
|
Hossain ME, Akter N. Further insights into the prevention of pulmonary hypertension syndrome (ascites) in broiler: a 65-year review. WORLD POULTRY SCI J 2022. [DOI: 10.1080/00439339.2022.2090305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Md. Emran Hossain
- Department of Animal Science and Nutrition, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Zakir Hossain Road, Khulshi, Bangladesh
| | - Nasima Akter
- Department of Dairy and Poultry Science, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Zakir Hossain Road, Khulshi, Bangladesh
| |
Collapse
|
38
|
Lozano J, Fernández-Ciganda S, González Revello Á, Hirigoyen D, Martínez M, Scorza C, Zunino P. Probiotic potential of GABA-producing lactobacilli isolated from Uruguayan artisanal cheese starter cultures. J Appl Microbiol 2022; 133:1610-1619. [PMID: 35699653 DOI: 10.1111/jam.15664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/05/2022] [Accepted: 06/06/2022] [Indexed: 11/29/2022]
Abstract
AIMS In this study, we sought to identify and characterize a collection of 101 lactobacilli strains isolated from natural whey starters used in Uruguayan artisan cheese production, based on their capacity to produce gamma-aminobutyric acid (GABA) and their probiotic potential. METHODS AND RESULTS The probiotic potential was assessed using low pH and bile salt resistance assays; bacterial adhesion to intestinal mucus was also evaluated. Selected strains were then identified by 16S sequencing, and their GABA-producing potential was confirmed and quantified using a UHPLC-MS system. Twenty-five strains were identified and characterized as GABA-producing lactobacilli belonging to the phylogenetical groups Lactiplantibacillus (n = 19) and Lacticaseibacillus (n = 6). Fifteen strains of the Lactiplantibacillus group showed a significantly higher GABA production than the rest. They showed the predicted ability to survive the passage through the gastrointestinal tract, according to the in vitro assays. CONCLUSIONS A set of promising candidate strains was identified as potential probiotics with action on the gut-brain axis. Further studies are needed to assess their possible effects on behaviour using in vivo assay. SIGNIFICANCE AND IMPACT OF THE STUDY This study shows the potential of strains isolated from local natural whey starters as probiotics and for biotechnological use in functional GABA-enriched foods formulation.
Collapse
Affiliation(s)
- Joaquin Lozano
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Sofía Fernández-Ciganda
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.,Animal Health Research Platform, Estación Experimental La Estanzuela, Instituto Nacional de Investigación Agropecuaria, Colonia, Uruguay
| | - Álvaro González Revello
- Department of Food Science and Technology, Facultad de Veterinaria, UdelaR, Montevideo, Uruguay
| | - Darío Hirigoyen
- Department of Food Science and Technology, Facultad de Veterinaria, UdelaR, Montevideo, Uruguay
| | - Marcela Martínez
- Analytical Platform, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Cecilia Scorza
- Department of Experimental Neuropharmacology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Pablo Zunino
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
39
|
Sugajski M, Maślak E, Złoch M, Rafińska K, Pomastowski P, Białczak D, Buszewski B. New sources of lactic acid bacteria with potential antibacterial properties. Arch Microbiol 2022; 204:349. [PMID: 35616812 DOI: 10.1007/s00203-022-02956-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
In the face of the growing demand for functional food, the search for new sources of lactic acid bacteria (LAB) becomes a priority. In our research, we used multiplied culture conditions followed by identification via the matrix-assisted laser desorption ionization-time of flight mass spectrometry for seeking LAB strains in plant- and animal-derived sources. Furthermore, the selected LAB isolates were examined for their proteolytic activity as well as antimicrobial action against different bacterial pathogens. The applied method appeared to be useful tool for searching LAB strains within different types of the biological matrices. The best source of the LABs was from calf. Comparing properties of the two selected LABs, those isolated from calf demonstrated the greatest proteolytic and antibacterial properties suggesting that gastrointestinal microbiota are the most valuable LAB source. Nevertheless, second selected strain derived from pickled cucumber juice may be also treated as a promising source of potential probiotic strains.
Collapse
Affiliation(s)
- Mateusz Sugajski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str, 87-100, Toruń, Poland
| | - Ewelina Maślak
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str, 87-100, Toruń, Poland.,Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100, Toruń, Poland
| | - Michał Złoch
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str, 87-100, Toruń, Poland. .,Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100, Toruń, Poland.
| | - Katarzyna Rafińska
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100, Toruń, Poland
| | - Paweł Pomastowski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str, 87-100, Toruń, Poland
| | - Dorota Białczak
- Polmlek Grudziądz Sp. z o. o, Magazynowa 8, 86-302, Grudziądz, Poland
| | - Bogusław Buszewski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Wileńska 4 Str, 87-100, Toruń, Poland.,Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University in Toruń, Gagarina 7, 87-100, Toruń, Poland
| |
Collapse
|
40
|
Breugelmans T, Oosterlinck B, Arras W, Ceuleers H, De Man J, Hold GL, De Winter BY, Smet A. The role of mucins in gastrointestinal barrier function during health and disease. Lancet Gastroenterol Hepatol 2022; 7:455-471. [PMID: 35397245 DOI: 10.1016/s2468-1253(21)00431-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
Abstract
Mucins are the gatekeepers of the mucosal barrier of the gastrointestinal tract and are aberrantly expressed in various gastrointestinal pathologies, including pathogen infection, inflammation, and uncontrolled growth and spread of abnormal cells. Although several studies have emphasised the role of mucins in dysfunction of the gastrointestinal mucosal barrier, they are often still considered to be passive mediators of this barrier instead of regulators or modulators. In this Review, we discuss the interactions between mucins and gastrointestinal barrier function during health and disease. We will focus on the bidirectional relationship between mucins and the gut microbiota and will also address the molecular mechanisms involved in key cell signalling pathways, such as inflammation, cell interactions, and cell differentiation, proliferation, and survival. Additionally, we highlight the potential use of mucins in the diagnosis, follow-up, and treatment of gastrointestinal diseases, such as chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Tom Breugelmans
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium
| | - Baptiste Oosterlinck
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium
| | - Wout Arras
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium
| | - Hannah Ceuleers
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium
| | - Joris De Man
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium
| | - Georgina L Hold
- Microbiome Research Centre, St George and Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium; Department of Gastroenterology and Hepatology, University Hospital of Antwerp, Antwerp, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Infla-Med Research Consortium of Excellence, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
41
|
Xu H, Lao L, Ji C, Lu Q, Guo Y, Pan D, Wu Z. Anti-inflammation and adhesion enhancement properties of the multifunctional LPxTG-motif surface protein derived from the Lactobacillus reuteri DSM 8533. Mol Immunol 2022; 146:38-45. [PMID: 35421739 DOI: 10.1016/j.molimm.2022.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
LPxTG-motif protein (LMP) is one kind of a precursor protein that contains a conserved LPxTG-motif at the C-terminus, which can be recognized by sortase A (SrtA) and covalently bind to the bacterial peptidoglycan. In this study, LMP derived from Lactobacillus reuteri (L. reuteri) was heterologous expressed and the tolerance and intestinal colonization ability of the LMP on L. reuteri were analyzed in simulated gastrointestinal fluid. Meanwhile, the anti-inflammatory activity of LMP was also evaluated in the LPS-stimulated RAW 264.7 cell model. The results indicated that LMP can promote the intestinal survival rate and adhesion characteristics of L. reuteri and enhanced the autoinducer-2 (AI-2) signaling molecule of the Lactobacillus strains in quorum sensing. Furthermore, LMP can inhibit the expressions of inflammatory cytokine TNF-α and IL-1β via ERK-JNK related MAPK signaling cascades. These findings provide a better understanding of the multifunctional LPxTG-motif surface protein derived from L. reuteri in the gastrointestinal tract environment.
Collapse
Affiliation(s)
- Hai Xu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lifeng Lao
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chunyu Ji
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Qianqian Lu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Yuxing Guo
- School of Food Science & Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zhen Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
42
|
Leeuwendaal NK, Stanton C, O’Toole PW, Beresford TP. Fermented Foods, Health and the Gut Microbiome. Nutrients 2022; 14:nu14071527. [PMID: 35406140 PMCID: PMC9003261 DOI: 10.3390/nu14071527] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/12/2022] Open
Abstract
Fermented foods have been a part of human diet for almost 10,000 years, and their level of diversity in the 21st century is substantial. The health benefits of fermented foods have been intensively investigated; identification of bioactive peptides and microbial metabolites in fermented foods that can positively affect human health has consolidated this interest. Each fermented food typically hosts a distinct population of microorganisms. Once ingested, nutrients and microorganisms from fermented foods may survive to interact with the gut microbiome, which can now be resolved at the species and strain level by metagenomics. Transient or long-term colonization of the gut by fermented food strains or impacts of fermented foods on indigenous gut microbes can therefore be determined. This review considers the primary food fermentation pathways and microorganisms involved, the potential health benefits, and the ability of these foodstuffs to impact the gut microbiome once ingested either through compounds produced during the fermentation process or through interactions with microorganisms from the fermented food that are capable of surviving in the gastro-intestinal transit. This review clearly shows that fermented foods can affect the gut microbiome in both the short and long term, and should be considered an important element of the human diet.
Collapse
Affiliation(s)
| | - Catherine Stanton
- Teagasc Food Research Centre, P61 C996 Cork, Ireland; (N.K.L.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland;
| | - Paul W. O’Toole
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland;
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland
| | - Tom P. Beresford
- Teagasc Food Research Centre, P61 C996 Cork, Ireland; (N.K.L.); (C.S.)
- Correspondence:
| |
Collapse
|
43
|
Alkalbani NS, Osaili TM, Al-Nabulsi AA, Olaimat AN, Liu SQ, Shah NP, Apostolopoulos V, Ayyash MM. Assessment of Yeasts as Potential Probiotics: A Review of Gastrointestinal Tract Conditions and Investigation Methods. J Fungi (Basel) 2022; 8:jof8040365. [PMID: 35448596 PMCID: PMC9027893 DOI: 10.3390/jof8040365] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022] Open
Abstract
Probiotics are microorganisms (including bacteria, yeasts and moulds) that confer various health benefits to the host, when consumed in sufficient amounts. Food products containing probiotics, called functional foods, have several health-promoting and therapeutic benefits. The significant role of yeasts in producing functional foods with promoted health benefits is well documented. Hence, there is considerable interest in isolating new yeasts as potential probiotics. Survival in the gastrointestinal tract (GIT), salt tolerance and adherence to epithelial cells are preconditions to classify such microorganisms as probiotics. Clear understanding of how yeasts can overcome GIT and salt stresses and the conditions that support yeasts to grow under such conditions is paramount for identifying, characterising and selecting probiotic yeast strains. This study elaborated the adaptations and mechanisms underlying the survival of probiotic yeasts under GIT and salt stresses. This study also discussed the capability of yeasts to adhere to epithelial cells (hydrophobicity and autoaggregation) and shed light on in vitro methods used to assess the probiotic characteristics of newly isolated yeasts.
Collapse
Affiliation(s)
- Nadia S. Alkalbani
- Department of Food Science, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Tareq M. Osaili
- Department Clinical Nutrition and Dietetics, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates;
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Anas A. Al-Nabulsi
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid 22110, Jordan;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P. O. Box 330127, Zarqa 13133, Jordan;
| | - Shao-Quan Liu
- Department of Food Science and Technology, Faculty of Science, National University of Singapore, S14 Level 5, Science Drive 2, Singapore 117542, Singapore;
| | - Nagendra P. Shah
- Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Pokfulam, Hong Kong 999077, China;
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia;
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Mutamed M. Ayyash
- Department of Food Science, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Correspondence:
| |
Collapse
|
44
|
Zhang W, Wei Y, Jin X, Lv X, Liu Z, Ni L. Spoilage of tilapia by Pseudomonas putida with different adhesion abilities. Curr Res Food Sci 2022; 5:710-717. [PMID: 35479657 PMCID: PMC9035656 DOI: 10.1016/j.crfs.2022.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 01/17/2023] Open
Abstract
Four Pseudomonas putida strains isolated from spoiled tilapia were divided into three adhesion abilities—high, medium, and low—by an in vitro mucus model. Four strains had no significant difference in spoilage ability to the inoculated fish fillets. However, according to the in vivo experiment, the spoilage caused by the four P.putida was positively correlated with their adhesion abilities. High adhesion strains not only caused more TVB-N in chilled fish, but also activated the spoilage activity of intestinal flora. The diversity of intestinal flora and the changes in volatile components in fish were detected by high-throughput sequencing and SPME-GC/MS. The strains with high adhesion abilities significantly changed the intestinal flora, which led to a significant increase in low-grade aldehydes, indole, and esters in flesh of fish, as well as the production of a fishy and pungent odor. The intestinal adhesion ability of spoilage bacteria was considered the key factor in spoilage of chilled fish. A positive correlation between the intestinal adhesion ability of P.putida and the spoilage ability in vivo. P.putida affected the intestinal microflora and led to increase in fishy and pungent odor. The intestinal adhesion ability of P.putida was considered as a key factor in spoilage.
Collapse
|
45
|
Andersen-Civil AIS, Myhill LJ, Büdeyri Gökgöz N, Engström MT, Mejer H, Zhu L, Zeller WE, Salminen JP, Krych L, Lauridsen C, Nielsen DS, Thamsborg SM, Williams AR. Dietary proanthocyanidins promote localized antioxidant responses in porcine pulmonary and gastrointestinal tissues during Ascaris suum-induced type 2 inflammation. FASEB J 2022; 36:e22256. [PMID: 35333423 DOI: 10.1096/fj.202101603rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/16/2022] [Accepted: 03/07/2022] [Indexed: 11/11/2022]
Abstract
Proanthocyanidins (PAC) are dietary polyphenols with putative anti-inflammatory and immunomodulatory effects. However, whether dietary PAC can regulate type-2 immune function and inflammation at mucosal surfaces remains unclear. Here, we investigated if diets supplemented with purified PAC modulated pulmonary and intestinal mucosal immune responses during infection with the helminth parasite Ascaris suum in pigs. A. suum infection induced a type-2 biased immune response in lung and intestinal tissues, characterized by pulmonary granulocytosis, increased Th2/Th1 T cell ratios in tracheal-bronchial lymph nodes, intestinal eosinophilia, and modulation of genes involved in mucosal barrier function and immunity. Whilst PAC had only minor effects on pulmonary immune responses, RNA-sequencing of intestinal tissues revealed that dietary PAC significantly enhanced transcriptional responses related to immune function and antioxidant responses in the gut of both naïve and A. suum-infected animals. A. suum infection and dietary PAC induced distinct changes in gut microbiota composition, primarily in the jejunum and colon, respectively. Notably, PAC consumption substantially increased the abundance of Limosilactobacillus reuteri. In vitro experiments with porcine macrophages and intestinal epithelial cells supported a role for both PAC polymers and PAC-derived microbial metabolites in regulating oxidative stress responses in host tissues. Thus, dietary PAC may have distinct beneficial effects on intestinal health during infection with mucosal pathogens, while having a limited activity to modulate naturally-induced type-2 pulmonary inflammation. Our results shed further light on the mechanisms underlying the health-promoting properties of PAC-rich foods, and may aid in the design of novel dietary supplements to regulate mucosal inflammatory responses in the gastrointestinal tract.
Collapse
Affiliation(s)
| | - Laura J Myhill
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Marica T Engström
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Helena Mejer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ling Zhu
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Wayne E Zeller
- USDA-ARS, U.S. Dairy Forage Research Center, Madison, Wisconsin, USA
| | - Juha-Pekka Salminen
- Natural Chemistry Research Group, Department of Chemistry, University of Turku, Turku, Finland
| | - Lukasz Krych
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | | | - Dennis S Nielsen
- Department of Food Science, University of Copenhagen, Frederiksberg, Denmark
| | - Stig M Thamsborg
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Andrew R Williams
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
46
|
Novel Developments on Stimuli-Responsive Probiotic Encapsulates: From Smart Hydrogels to Nanostructured Platforms. FERMENTATION 2022. [DOI: 10.3390/fermentation8030117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Biomaterials engineering and biotechnology have advanced significantly towards probiotic encapsulation with encouraging results in assuring sufficient bioactivity. However, some major challenges remain to be addressed, and these include maintaining stability in different compartments of the gastrointestinal tract (GIT), favoring adhesion only at the site of action, and increasing residence times. An alternative to addressing such challenges is to manufacture encapsulates with stimuli-responsive polymers, such that controlled release is achievable by incorporating moieties that respond to chemical and physical stimuli present along the GIT. This review highlights, therefore, such emerging delivery matrices going from a comprehensive description of addressable stimuli in each GIT compartment to novel synthesis and functionalization techniques to currently employed materials used for probiotic’s encapsulation and achieving multi-modal delivery and multi-stimuli responses. Next, we explored the routes for encapsulates design to enhance their performance in terms of degradation kinetics, adsorption, and mucus and gut microbiome interactions. Finally, we present the clinical perspectives of implementing novel probiotics and the challenges to assure scalability and cost-effectiveness, prerequisites for an eventual niche market penetration.
Collapse
|
47
|
Lacticaseibacillus rhamnosus: A Suitable Candidate for the Construction of Novel Bioengineered Probiotic Strains for Targeted Pathogen Control. Foods 2022; 11:foods11060785. [PMID: 35327208 PMCID: PMC8947445 DOI: 10.3390/foods11060785] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics, with their associated beneficial effects, have gained popularity for the control of foodborne pathogens. Various sources are explored with the intent to isolate novel robust probiotic strains with a broad range of health benefits due to, among other mechanisms, the production of an array of antimicrobial compounds. One of the shortcomings of these wild-type probiotics is their non-specificity. A pursuit to circumvent this limitation led to the advent of the field of pathobiotechnology. In this discipline, specific pathogen gene(s) are cloned and expressed into a given probiotic to yield a novel pathogen-specific strain. The resultant recombinant probiotic strain will exhibit enhanced species-specific inhibition of the pathogen and its associated infection. Such probiotics are also used as vehicles to deliver therapeutic agents. As fascinating as this approach is, coupled with the availability of numerous probiotics, it brings a challenge with regard to deciding which of the probiotics to use. Nonetheless, it is indisputable that an ideal candidate must fulfil the probiotic selection criteria. This review aims to show how Lacticaseibacillus rhamnosus, a clinically best-studied probiotic, presents as such a candidate. The objective is to spark researchers’ interest to conduct further probiotic-engineering studies using L. rhamnosus, with prospects for the successful development of novel probiotic strains with enhanced beneficial attributes.
Collapse
|
48
|
Multiplex SYBR Green real-time PCR for Lactobacillus acidophilus group species targeting biomarker genes revealed by a pangenome approach. Microbiol Res 2022; 259:127013. [DOI: 10.1016/j.micres.2022.127013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 02/16/2022] [Accepted: 03/16/2022] [Indexed: 11/19/2022]
|
49
|
Zhang S, Wang T, Zhang D, Wang X, Zhang Z, Lim C, Lee S. Probiotic characterization of Lactiplantibacillus plantarum HOM3204 and its restoration effect on antibiotic-induced dysbiosis in mice. Lett Appl Microbiol 2022; 74:949-958. [PMID: 35231139 PMCID: PMC9315005 DOI: 10.1111/lam.13683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
The purpose of this study was to evaluate the probiotic characteristics of Lactiplantibacillus plantarum HOM3204 isolated from homemade pickled cabbage and to examine its restoration effect on antibiotic-induced dysbiosis in mice. Lact. plantarum HOM3204 tolerated simulated gastric and intestinal juices with a 99.38% survival rate. It also showed strong adhesion ability (3.45%) to Caco-2 cells and excellent antimicrobial activity against foodborne pathogens in vitro. For safety (antibiotic susceptibility) of this strain, it was susceptible to all the tested seven antibiotics. Lact. plantarum HOM3204 had good stability during storage, especially in cold and frozen conditions. Furthermore, Lact. plantarum HOM3204 significantly restored the gut microbiota composition by increasing the abundance of Lactobacilli and Bifidobacteria and decreasing Enterococci, and improved antioxidative function by raising the concentrations of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) in serum of antibiotic-induced dysbiosis in mice. These results suggest that Lact. plantarum HOM3204 could be a potential probiotic as a functional food ingredient.
Collapse
Affiliation(s)
- S Zhang
- Coree Beijing Co., Ltd, Beijing, China
| | - T Wang
- Beijing Hanmi pharmaceutical Co., Ltd, Beijing, China
| | - D Zhang
- Coree Beijing Co., Ltd, Beijing, China
| | - X Wang
- Beijing Hanmi pharmaceutical Co., Ltd, Beijing, China
| | - Z Zhang
- Beijing Hanmi pharmaceutical Co., Ltd, Beijing, China
| | - C Lim
- Coree Beijing Co., Ltd, Beijing, China.,Coree Pohang Co., Ltd, Pohang, Korea
| | - S Lee
- Coree Beijing Co., Ltd, Beijing, China.,Coree Pohang Co., Ltd, Pohang, Korea
| |
Collapse
|
50
|
Biagini F, Calvigioni M, De Maria C, Magliaro C, Montemurro F, Mazzantini D, Celandroni F, Mattioli-Belmonte M, Ghelardi E, Vozzi G. Study of the Adhesion of the Human Gut Microbiota on Electrospun Structures. Bioengineering (Basel) 2022; 9:bioengineering9030096. [PMID: 35324785 PMCID: PMC8945341 DOI: 10.3390/bioengineering9030096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/04/2022] Open
Abstract
Although the adhesion of bacteria on surfaces is a widely studied process, to date, most of the works focus on a single species of microorganisms and are aimed at evaluating the antimicrobial properties of biomaterials. Here, we describe how a complex microbial community, i.e., the human gut microbiota, adheres to a surface to form stable biofilms. Two electrospun structures made of natural, i.e., gelatin, and synthetic, i.e., polycaprolactone, polymers were used to study their ability to both promote the adhesion of the human gut microbiota and support microbial growth in vitro. Due to the different wettabilities of the two surfaces, a mucin coating was also added to the structures to decouple the effect of bulk and surface properties on microbial adhesion. The developed biofilm was quantified and monitored using live/dead imaging and scanning electron microscopy. The results indicated that the electrospun gelatin structure without the mucin coating was the optimal choice for developing a 3D in vitro model of the human gut microbiota.
Collapse
Affiliation(s)
- Francesco Biagini
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 55122 Pisa, Italy; (F.B.); (C.D.M.); (C.M.); (F.M.)
- Department of Information Engineering, University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
| | - Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 37, 56127 Pisa, Italy; (M.C.); (D.M.); (F.C.); (E.G.)
| | - Carmelo De Maria
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 55122 Pisa, Italy; (F.B.); (C.D.M.); (C.M.); (F.M.)
- Department of Information Engineering, University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
| | - Chiara Magliaro
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 55122 Pisa, Italy; (F.B.); (C.D.M.); (C.M.); (F.M.)
- Department of Information Engineering, University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
| | - Francesca Montemurro
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 55122 Pisa, Italy; (F.B.); (C.D.M.); (C.M.); (F.M.)
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 37, 56127 Pisa, Italy; (M.C.); (D.M.); (F.C.); (E.G.)
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 37, 56127 Pisa, Italy; (M.C.); (D.M.); (F.C.); (E.G.)
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Science—DISCLIMO Università Politecnica delle Marche, Via Tronto 10/A, 60126 Ancona, Italy;
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via San Zeno 37, 56127 Pisa, Italy; (M.C.); (D.M.); (F.C.); (E.G.)
| | - Giovanni Vozzi
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 55122 Pisa, Italy; (F.B.); (C.D.M.); (C.M.); (F.M.)
- Department of Information Engineering, University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Correspondence:
| |
Collapse
|