1
|
Ahmed ME, Aljarbou A, Mohammed HA, Khan RA. Bacteriocin isolated from Ralstonia mannitolilytica and bacteriocin-capped silver nanoparticles: Comparative effects on biofilm formation and LuxS Gene's expressions by Proteus mirabilis as an approach to counter MDR catheter infection. Microb Pathog 2025; 204:107558. [PMID: 40210139 DOI: 10.1016/j.micpath.2025.107558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 03/20/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Among undesirables in treating certain infections and diseases is the contamination of catheters, especially of the microbes' resistance to drugs. The situation has necessitated the search for alternative antimicrobial agents. Bacteriocin category, antibiotic-originate, peptide-natured, Ralstonia mannitolilytica microbes-produced, bacteriocin material, and the semi-pure bacteriocin capped silver metal nanoparticles (AgNPs) were used for combating the MDR (multi drug resistance) organism, Proteus mirabilis, which is the third-most common cause of UTI (urinary tract infection), and that is linked to catheter use, are being recommended for clinical use with certain development. The crude microbial product was isolated from the microbial entity, Ralstonia mannitolilytica, which grows in crude petroleum-contaminated soil, and was semi-purified for use in the synthesis of the bacteriocin-capped AgNPs. The prepared nanoparticles were characterized using X-ray diffraction, indicating the silver element's presence; field emission scanning electron microscopy, revealing near-spherical but irregular shapes of the bacteriocin-capped AgNPs with a size range of 34-46 nm; and atomic force microscopic analysis, which demonstrated the nanoparticles surface characteristics. The DLS analysis established the negative charge, and an average hydrodynamic size of 51 nm, while the UV-Vis spectroscopic analysis showed the absorption maxima (λmax) at 454 nm. The P. mirabilis isolates were numbered according to MDR detection by the VITEK 2 system (A to J), and the microbes appeared as a pale-coloured colony on MacConkey agar. The biofilm formation screening revealed the highest biofilm-producing isolates, identified as A, B, C, and D. The treatments with both bacteriocin and the bacteriocin-capped AgNPs showed that bacteriocin inhibited the biofilm formation for isolates A, B, and C, but isolate D was less affected, while bacteriocin capped AgNPs inhibited the film formation in isolates A, C, and D more than the bacteriocin alone. However, the activity level was low to moderate. In addition, the LuxS gene-down-regulating effects of bacteriocin and bacteriocin-capped AgNPs were also observed. The expression of the LuxS gene in P. mirabilis was lowered by bacteriocin-capped AgNPs during biofilm formation, while the isolates B and C lowered their expressions of the LuxS gene more effectively when the bacteriocin was used. The study finds the use of bacteriocin and bacteriocin-capped AgNPs of value for developing these products, especially bacteriocin-capped AgNPs, for managing the catheter infections. The products need further development and clinical testings.
Collapse
Affiliation(s)
- Mais E Ahmed
- Department of Biology, College of Science, University of Baghdad, Baghdad, Iraq.
| | - Ahmed Aljarbou
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
| | - Riaz A Khan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, 51452, Saudi Arabia
| |
Collapse
|
2
|
Huang T, Li Z, Qu X, Yao G, Kwok LY, He Q, Zhang H. Preliminary Purification and Partial Characterization of a Functional Bacteriocin of Lacticaseibacillus paracasei Zhang and Mining for its Gene Cluster. Probiotics Antimicrob Proteins 2025; 17:487-499. [PMID: 38748307 PMCID: PMC11926035 DOI: 10.1007/s12602-024-10249-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2024] [Indexed: 03/21/2025]
Abstract
Bacteriocins produced by lactic acid bacteria (LAB) have good potential for use as food biopreservatives. Lacticaseibacillus paracasei Zhang (L. paracasei Zhang) is both a food use and a probiotic bacterium. This study aimed to purify and preliminary characterize the active antibacterial metabolite of L. paracasei Zhang. The cell-free supernatant of L. paracasei Zhang was collected and purified by ultrafiltration and gel filtration chromatography. The 1-3 kDa active fraction could inhibit the growth of Staphylococcus aureus but not Escherichia coli. Further antibacterial activity assays revealed its capacity to suppress various foodborne and human opportunistic pathogens (including Staphylococcus aureus, Pseudomonas fluorescens, Pseudomonas aeruginosa, Listeria monocytogenes, and Bacillus cereus), but not fungi. The antibacterial activity showed good tolerance to heat (40 to 100 °C), acid-base (pH 2-3 and pH 6-10), and digestions by a number of industrial and animal/human enzymes (such as trypsin, pepsin, α-amylase, and protease K, except papain); these desired properties make it a suitable biopreservative to be used in harsh and complex industrial production processes. The high papain sensitivity suggested a proteinaceous/peptide nature of the bioactivity. Moreover, our genomic data mining for bacteriocin through BAGEL4 revealed an area of interest encoding a complete set of putative genes required for bacteriocin production. In conclusion, our study showed that L. paracasei Zhang can produce extracellular functional antibacterial metabolite, likely a class II bacteriocin. Our preliminary extraction and characterization of the active metabolite demonstrated that it has good potential to be used as a biopreservative or an agent for suppressing gastrointestinal infections.
Collapse
Affiliation(s)
- Tian Huang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Zhaojie Li
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, Shandong, China
- Qingdao Special Food Research Institute, QingdaoShandong, 266109, China
| | - Xinan Qu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, Shandong, China
- Qingdao Special Food Research Institute, QingdaoShandong, 266109, China
| | - Guoqiang Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Qiuwen He
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
- Department of College of Food Science and Engineering, Inner Mongolia Agricultural University, No. 306, Zhaowuda Road, Saihan District, Hohhot, 010018, Inner Mongolia, China.
| |
Collapse
|
3
|
Adeyemi DN, Ajide EO, Adebami GE, Abiala MA. Bacillus Genotypes Exhibit Antagonistic Effects on Lettuce-Based Enterobacter Pathotypes. Curr Microbiol 2025; 82:181. [PMID: 40057914 DOI: 10.1007/s00284-025-04163-8] [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: 07/09/2024] [Accepted: 02/25/2025] [Indexed: 03/18/2025]
Abstract
Globally, among ready-to-eat (RTE) vegetables, lettuce is well-known as a potential host for Enterobacteriaceae, which theatens public health. However, in addition to E. coli, pathogenic Enterobacter species associated with RTE lettuce is poorly investigated, as well as their genetic relationship with sources of contamination has not been determined. This study investigated the evolutionary relationship between Enterobacter species and their antibiotic resistance attributes in RTE lettuce, irrigation water, and the soil of lettuce farms. Enterobacter species had the highest occurrence in the irrigation water (38%), followed by both the RTE lettuce (31%) and the soil of the lettuce farm (31%). The 16S rRNA-identified the bacteria isolates as Enterobacter species. Phylogenetic analysis revealed that Enterobacter species from RTE lettuce and irrigation water maintained a strong evolutionary relationship in comparison with those from the soil of the lettuce farm. In addition, irrespective of the source of contamination, the isolates demonstrated resistance to 67% of the 12 antibiotics tested. To overcome these challenges, efforts have been made to identify novel antimicrobial agents, especially from eco-friendly soil bacteria. Among the 76 soil bacterial isolates that were assessed via the antagonist‒pathogen agar plug method, only two (IBT42 and VFK47) exhibited outstanding (P = 0.05) antagonism against > 50% of the Enterobacter pathotypes in comparison with the control. The potency of IBT42 and VFK47 were validated via agar well diffusion, the minimum inhibitory concentration, and the minimum bactericidal concentration. The 16S rRNA and phylogenetic analysis revealed that the identities of IBT42 and VFK47 were Bacillus mycoides and Bacillus cereus, respectively.
Collapse
Affiliation(s)
- Damilola N Adeyemi
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Esther O Ajide
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Gboyega E Adebami
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria
| | - Moses A Abiala
- Department of Biological Sciences, Mountain Top University, Ogun State, Prayer City, Nigeria.
| |
Collapse
|
4
|
Kalairaj A, Rajendran S, Karthikeyan R, Panda RC, Senthilvelan T. A Comprehensive Review on Preparation of Silver Nanoparticles from a Bacteriocin for the Natural Preservation of Food Products. Appl Biochem Biotechnol 2025; 197:1419-1452. [PMID: 39621224 DOI: 10.1007/s12010-024-05122-y] [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] [Accepted: 11/19/2024] [Indexed: 03/29/2025]
Abstract
Food preservation aims to maintain safe and nutritious food for extended periods by inhibiting microbial growth that causes spoilage and poses health risks. Traditional chemical preservatives like sodium sulfite, sodium nitrite, sodium benzoate, tBHQ and BHA have raised concerns due to potential carcinogenicity, genotoxicity and allergies with long-term consumption. As a natural alternative, bacteriocins have emerged for food preservation. These ribosomally synthesised antimicrobial peptides are produced by various microorganisms, including bacteria, fungi and yeast, typically during their stationary growth phase. Bacteriocins are categorised into four classes based on structure and function, with molecular weights averaging between 30 and 80 kDa. They exhibit antimicrobial activity against a range of bacteria, mediating complex interactions between bacterial species and enhancing competitiveness and survival of producer strains. Both gram-positive and gram-negative bacteria produce bacteriocins. Recent advancements have identified and optimized bacteriocins for applications in food technology, extending shelf life, managing foodborne illnesses and contributing to public health preservation. Their eco-friendly nature and safety profile make bacteriocins promising for future food preservation strategies without detrimental effects on humans or animals. The current review has mainly focused on the preservation of food products using bacteriocin.
Collapse
Affiliation(s)
- Ashmitha Kalairaj
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Swethashree Rajendran
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602105, Tamil Nadu, India
| | - R Karthikeyan
- Central Instrumentation Laboratory, National Institute of Food Technology, Entrepreneurship and Management (NIFTEM), Thanjavur (an Institute of National Importance, Formerly Indian Institute of Food Processing Technology (IIFPT)), Ministry of Food Processing Industries (MoFPI), Government of India, Pudukkottai Road, Thanjavur, 613005, Tamil Nadu, India
| | - Rames C Panda
- Chemical Engineering Division, Rajalakshmi Engineering College, Thandalam, Chennai, 602 105, Tamil Nadu, India
| | - T Senthilvelan
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, 602105, Tamil Nadu, India.
| |
Collapse
|
5
|
Sebe M, Senoura S, Miura K, Kobayashi W, Yano N, Yamauchi G, Harada K, Fukuyama Y, Kubo M, Murakami K. Antibacterial Activity of Banglene Extracted from Indonesian Ginger "Bangle" Against Porphyromonas gingivalis. Int J Mol Sci 2025; 26:1787. [PMID: 40076415 PMCID: PMC11898951 DOI: 10.3390/ijms26051787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Periodontitis is one of the most common diseases associated with the lifestyle habits of adults and is caused by the formation of biofilms, called dental plaques, in periodontal pockets by oral bacteria, such as Porphyromonas gingivalis. Bangle, Zingiber purpureum Rosc. (Indonesian ginger), a native Indonesian plant, has been traditionally consumed as food and medicine across Southeast Asia. The cis- and trans-banglenes, components of the rhizomes of Z. purpureum, have been reported to possess neurotrophic activity. Hexane extract of bangle exhibited antibacterial activity against P. gingivalis, with a minimum inhibitory concentration of 8 μg/mL. We isolated several compounds from the active fractions through the bioassay-guided isolation of hexane extract. Further, we found that c- and t-banglene inhibited the growth of P. gingivalis at 4 µg/mL; however, these compounds showed no antibacterial effects against oral microorganisms. We also observed that c- and t-banglenes resulted in 47% and 40% reductions in biofilm formation. In conclusion, our results suggest that banglene has specific antibacterial effects against the periodontopathogen P. gingivalis, with minimal impact on oral microorganisms. Thus, banglene has potential applications in the prevention of periodontitis without the risk of substituted microbisms.
Collapse
Affiliation(s)
- Mayu Sebe
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan; (M.S.)
| | - Satoka Senoura
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan; (M.S.)
| | - Kiyoshi Miura
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan; (M.S.)
| | - Wako Kobayashi
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan; (M.S.)
| | - Nagisa Yano
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Gaku Yamauchi
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Kenichi Harada
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Yoshiyasu Fukuyama
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Miwa Kubo
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima 770-8514, Japan
| | - Keiji Murakami
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan; (M.S.)
| |
Collapse
|
6
|
Liang Q, Zhou W, Peng S, Liang Z, Liu Z, Zhu C, Mou H. Current status and potential of bacteriocin-producing lactic acid bacteria applied in the food industry. Curr Res Food Sci 2025; 10:100997. [PMID: 39995467 PMCID: PMC11849202 DOI: 10.1016/j.crfs.2025.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/11/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Lactic acid bacteria (LAB) have been widely applied in the food industry and have brought many beneficial effects on food products, and some of those benefits are related to their metabolic product. Bacteriocins produced by LAB have attracted the attentions for application in the food industry as natural food bio-preservatives because of their antimicrobial activity against the food spoilage and pathogenic bacteria. With the increasing demands of consumers for more healthier food and investigations on natural food preservatives, the bioactivity of bacteriocins allows them to give the application values to the bacteriocin-producing LAB. Accordingly, the capacity of LAB to produce bacteriocin in the aspects of classifications, mode of action, biosynthesis mechanisms are introduced, which leads to further consideration of the current status and potential values of bacteriocin-producing LAB applied in the food industry. The comparation of guidelines of LAB and bacteriocins for food application are also proposed for better understanding their practical application promising. This review will be helpful for current and future researches on the application of bacteriocin-producing LAB in the food industry.
Collapse
Affiliation(s)
- Qingping Liang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| | - Wei Zhou
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| | - Siyuan Peng
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| | - Ziyu Liang
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Zhemin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| | - Changliang Zhu
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| | - Haijin Mou
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266404, China
| |
Collapse
|
7
|
Siamalube B, Ehinmitan E. Vibrio cholerae: Understanding a persistent pathogen in Sub-Saharan Africa and the East Mediterranean Region. Pathog Dis 2025; 83:ftaf004. [PMID: 40145130 PMCID: PMC11999019 DOI: 10.1093/femspd/ftaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 03/28/2025] Open
Abstract
Vibrio cholerae remains a significant public health threat in Sub-Saharan Africa and the East Mediterranean Region, where recurrent outbreaks are driven by inadequate water, sanitation, and hygiene infrastructure, climatic variability, and socio-political instability. This review explores the persistence of the pathogen in these regions, examining its epidemiology, environmental reservoirs, and genomic adaptations that enhance its survival and transmission. We highlight the impact of antimicrobial resistance and the role of climate change in cholera dynamics. Furthermore, we discuss current prevention and control strategies, including advancements in oral cholera vaccines, genomic surveillance, and microbiome-targeted interventions. Addressing these challenges requires a multifaceted approach that integrates sustainable sanitation improvements, strengthened disease surveillance, and innovative vaccination strategies. Understanding the persistence of V. cholerae in these high-risk regions is critical for developing effective, long-term mitigation strategies to reduce cholera morbidity and mortality.
Collapse
Affiliation(s)
- Beenzu Siamalube
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation, P.O. Box 62000-00200, Nairobi, Kenya
| | - Emmanuel Ehinmitan
- Department of Molecular Biology and Biotechnology, Pan African University Institute for Basic Sciences, Technology and Innovation, P.O. Box 62000-00200, Nairobi, Kenya
| |
Collapse
|
8
|
Fernández-Fernández R, Lozano C, Campaña-Burguet A, González-Azcona C, Álvarez-Gómez T, Fernández-Pérez R, Peña R, Zarazaga M, Carrasco J, Torres C. Bacteriocin-Producing Staphylococci and Mammaliicocci Strains for Agro-Food and Public Health Applications with Relevance of Micrococcin P1. Antibiotics (Basel) 2025; 14:97. [PMID: 39858382 PMCID: PMC11763047 DOI: 10.3390/antibiotics14010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/28/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Antimicrobial-producing strains and their bacteriocins hold great promise for the control of bacterial diseases, being an attractive alternative to antibiotics. Thus, the aim of this study was to evaluate the inhibitory activity of 15 bacteriocin-producing staphylococci and mammaliicocci (BP-S/M) strains and their pre-purified extracts with butanol (BT) against a collection of 27 harmful or zoonotic strains (including Gram-positive/-negative bacteria and molds) with relevance in the public health and agro-food fields. These indicators (excluding Gram-negative strains) were grouped into seven categories based on their potential application areas: dairy livestock mastitis, avian pathogen zoonoses, swine zoonoses, food safety, aquaculture, wine making, and mushroom cultivation. In addition, cross-immunity assays between the BP-S/M strains were carried out to identify potential strain combinations to enhance their activity against pathogens. Finally, the hemolytic and gelatinase activities were tested in the BP-S/M strains. A strong inhibitory capacity of the BP-S/M strains was verified against relevant Gram-positive indicators, such as methicillin-resistant Staphylococcus aureus, Listeria monocytogenes, and Clostridium perfringens, among others, while no activity was detected against Gram-negative ones. Interestingly, several BT extracts inhibited the two mold indicators included in this study as representants of mushroom pathogens. The Micrococcin P1 producer Staphylococcus hominis C5835 (>60% of indicators were intensively inhibited by all the methods) can be proposed as a potential candidate for the control of bacterial diseases in the aforementioned categories alone or in combination with other BP-S/M strains (mainly with Staphylococcus warneri X2969). In this regard, five potential combinations of BP-S/M strains that enhanced their activity against specific pathogens were detected.
Collapse
Affiliation(s)
- Rosa Fernández-Fernández
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Carmen Lozano
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Allelen Campaña-Burguet
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Carmen González-Azcona
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Tamara Álvarez-Gómez
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Rocío Fernández-Pérez
- Instituto de Ciencias de la Vid y del Vino (ICVV) (Universidad de La Rioja, Consejo Superior de Investigaciones Científicas (CSIC), Gobierno de La Rioja), 26007 Logroño, Spain;
| | - Raquel Peña
- Department of Microbiology and Parasitology, Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain;
| | - Myriam Zarazaga
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| | - Jaime Carrasco
- Department Ecology of Cultivated Mushrooms, Regional Institute for Agri-Food and Forest Research and Development (IRIAF), 16194 Cuenca, Spain;
| | - Carmen Torres
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006 Logroño, Spain; (R.F.-F.); (C.L.); (A.C.-B.); (C.G.-A.); (T.Á.-G.); (M.Z.)
| |
Collapse
|
9
|
Wang R, Yu YF, Yu WR, Sun SY, Lei YM, Li YX, Lu CX, Zhai JN, Bai FR, Ren F, Huang JQ, Chen J. Roles of Probiotics, Prebiotics, and Postbiotics in B-Cell-Mediated Immune Regulation. J Nutr 2025; 155:37-51. [PMID: 39551357 DOI: 10.1016/j.tjnut.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024] Open
Abstract
Probiotics, prebiotics, and postbiotics can significantly influence B-cell-related diseases through their immunomodulatory effects. They enhance the immune system's function, particularly affecting B cells, which originate in the bone marrow and are crucial for antibody production and immune memory. These substances have therapeutic potential in managing allergies, autoimmune diseases, and inflammatory conditions by regulating the gut microbiota, strengthening epithelial barriers, and directly interacting with various components of the innate and adaptive immune systems. The review highlights the critical need for further research into the precise mechanisms through which probiotics, prebiotics, and postbiotics modulate B cells. Gaining this understanding could facilitate the development of more effective treatments for B-cell-related diseases by harnessing the immunomodulatory properties of these dietary components.
Collapse
Affiliation(s)
- Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yifei F Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Weiru R Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Siyuan Y Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yumei M Lei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan X Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Chenxu X Lu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jianan N Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Feirong R Bai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jiaqiang Q Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| |
Collapse
|
10
|
Ismael M, Huang M, Zhong Q. The Bacteriocins Produced by Lactic Acid Bacteria and the Promising Applications in Promoting Gastrointestinal Health. Foods 2024; 13:3887. [PMID: 39682959 DOI: 10.3390/foods13233887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Bacteriocins produced by lactic acid bacteria (LAB) are promising bioactive peptides. Intriguingly, bacteriocins have health benefits to the host and may be applied safely in the food industry as bio-preservatives or as therapeutic interventions preventing intestinal diseases. In recent years, finding a safe alternative approach to conventional treatments to promote gut health is a scientific hotspot. Therefore, this review aimed to give insight into the promising applications of LAB-bacteriocins in preventing intestinal diseases, such as colonic cancer, Helicobacter pylori infections, multidrug-resistant infection-associated colitis, viral gastroenteritis, inflammatory bowel disease, and obesity disorders. Moreover, we highlighted the recent research on bacteriocins promoting gastrointestinal health. The review also provided insights into the proposed mechanisms, challenges and opportunities, trends and prospects. In addition, a SWOT analysis was conducted on the potential applications. Based on properties, biosafety, and health functions of LAB-bacteriocins, we conclude that the future applications of LAB-bacteriocins are promising in promoting gastrointestinal health. Further in vivo trials are needed to confirm these potential effects of LAB-bacteriocins interventions.
Collapse
Affiliation(s)
- Mohamedelfatieh Ismael
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Mingxin Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Qingping Zhong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
11
|
Arslan NP, Orak T, Ozdemir A, Altun R, Esim N, Eroglu E, Karaagac SI, Aktas C, Taskin M. Polysaccharides and Peptides With Wound Healing Activity From Bacteria and Fungi. J Basic Microbiol 2024; 64:e2400510. [PMID: 39410821 PMCID: PMC11609500 DOI: 10.1002/jobm.202400510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 12/13/2024]
Abstract
Bacteria and fungi are natural sources of metabolites exhibiting diverse bioactive properties such as wound healing, antioxidative, antibacterial, antifungal, anti-inflammatory, antidiabetic, and anticancer activities. Two important groups of bacteria or fungi-derived metabolites with wound-healing potential are polysaccharides and peptides. In addition to bacteria-derived cellulose and hyaluronic acid and fungi-derived chitin and chitosan, these organisms also produce different polysaccharides (e.g., exopolysaccharides) with wound-healing potential. The most commonly used bacterial peptides in wound healing studies are bacteriocins and lipopeptides. Bacteria or fungi-derived polysaccharides and peptides exhibit both the in vitro and the in vivo wound healing potency. In the in vivo models, including animals and humans, these metabolites positively affect wound healing by inhibiting pathogens, exhibiting antioxidant activity, modulating inflammatory response, moisturizing the wound environment, promoting the proliferation and migration of fibroblasts and keratinocytes, increasing collagen synthesis, re-epithelialization, and angiogenesis. Therefore, peptides and polysaccharides derived from bacteria and fungi have medicinal importance. This study aims to overview current literature knowledge (especially within the past 5 years) on the in vitro and in vivo wound repair potentials of polysaccharides and peptides obtained from bacteria (Actinobacteria, Bacteroidetes, Cyanobacteria, Firmicutes, and Proteobacteria) and fungi (yeasts, filamentous microfungi, and mushrooms).
Collapse
Affiliation(s)
| | - Tugba Orak
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Aysenur Ozdemir
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Ramazan Altun
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Nevzat Esim
- Department of Molecular Biology and Genetics, Science and Art FacultyBingol UniversityBingolTurkey
| | - Elvan Eroglu
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Sinem Ilayda Karaagac
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Cigdem Aktas
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| | - Mesut Taskin
- Department of Molecular Biology and Genetics, Science FacultyAtaturk UniversityErzurumTurkey
| |
Collapse
|
12
|
Nisar S, Shah AH, Nazir R. The clinical praxis of bacteriocins as natural anti-microbial therapeutics. Arch Microbiol 2024; 206:451. [PMID: 39476181 DOI: 10.1007/s00203-024-04152-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 11/10/2024]
Abstract
In recent decades, the excessive use of antibiotics has resulted in a rise in antimicrobial drug resistance (ADR). Annually, a significant number of human lives are lost due to resistant infectious diseases, leading to around 700,000 deaths, and it is estimated that by 2050, there could be up to 10 million casualties. Apart from their possible application as preservatives in the food sector, bacteriocins are gaining acknowledgment as potential clinical treatments. Not only this, these antimicrobial peptides have revealed in modulating the host immune system producing anti-inflammatory and anti-modulatory responses. At the same time, due to the ever-increasing global threat of antibiotic resistance, bacteriocins have gained attraction among researchers due to their potential clinical applications. Bacteriocins as antimicrobial peptides, represent one of the most important natural defense mechanisms among bacterial species, particularly lactic acid bacteria (LAB), that can fight against infection-causing pathogens. In this review, we are highlighting the potential of bacteriocins as novel therapeutics for inhibiting a wide range of clinically relevant and multi-drug-resistant pathogens (MDR). We also highlight the effectiveness and potential applications of current bacteriocin treatments in combating antimicrobial resistance (AMR), thereby promoting human health.
Collapse
Affiliation(s)
- Safura Nisar
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India
| | - Abdul Haseeb Shah
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| | - Ruqeya Nazir
- Centre of Research for Development (CORD), School of Biological Sciences, University of Kashmir, Hazratbal, Srinagar, 190006, India.
| |
Collapse
|
13
|
Guamán LP, Carrera-Pacheco SE, Zúñiga-Miranda J, Teran E, Erazo C, Barba-Ostria C. The Impact of Bioactive Molecules from Probiotics on Child Health: A Comprehensive Review. Nutrients 2024; 16:3706. [PMID: 39519539 PMCID: PMC11547800 DOI: 10.3390/nu16213706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Background: This review investigates the impact of bioactive molecules produced by probiotics on child health, focusing on their roles in modulating gut microbiota, enhancing immune function, and supporting overall development. Key metabolites, including short-chain fatty acids (SCFAs), bacteriocins, exopolysaccharides (EPSs), vitamins, and gamma-aminobutyric acid (GABA), are highlighted for their ability to maintain gut health, regulate inflammation, and support neurodevelopment. Objectives: The aim of this review is to examine the mechanisms of action and clinical evidence supporting the use of probiotics and postbiotics in pediatric healthcare, with a focus on promoting optimal growth, development, and overall health in children. Methods: The review synthesizes findings from clinical studies that investigate the effects of probiotics and their metabolites on pediatric health. The focus is on specific probiotics and their ability to influence gut health, immune responses, and developmental outcomes. Results: Clinical studies demonstrate that specific probiotics and their metabolites can reduce gastrointestinal disorders, enhance immune responses, and decrease the incidence of allergies and respiratory infections in pediatric populations. Additionally, postbiotics-bioactive compounds from probiotic fermentation-offer promising benefits, such as improved gut barrier function, reduced inflammation, and enhanced nutrient absorption, while presenting fewer safety concerns compared to live probiotics. Conclusions: By examining the mechanisms of action and clinical evidence, this review underscores the potential of integrating probiotics and postbiotics into pediatric healthcare strategies to promote optimal growth, development, and overall health in children.
Collapse
Affiliation(s)
- Linda P. Guamán
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Saskya E. Carrera-Pacheco
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Johana Zúñiga-Miranda
- Centro de Investigación Biomédica (CENBIO), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170527, Ecuador; (S.E.C.-P.); (J.Z.-M.)
| | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Cesar Erazo
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
| | - Carlos Barba-Ostria
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador; (E.T.); (C.E.)
- Instituto de Microbiología, Universidad San Francisco de Quito USFQ, Quito 170901, Ecuador
| |
Collapse
|
14
|
Seidler Y, Rimbach G, Lüersen K, Vinderola G, Ipharraguerre IR. The postbiotic potential of Aspergillus oryzae - a narrative review. Front Microbiol 2024; 15:1452725. [PMID: 39507340 PMCID: PMC11538067 DOI: 10.3389/fmicb.2024.1452725] [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: 06/21/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
The filamentous fungus Aspergillus oryzae has a long tradition in East Asian food processing. It is therefore not surprising that in recent years fermentation products of A. oryzae have attracted attention in the emerging field of postbiotics. This review aims to provide a comprehensive summary of the potential postbiotic effects of fermentation products from A. oryzae, by discussing possible mechanisms of action against the background of the molecular composition determined so far. In particular, cell wall constituents, enzymes, extracellular polymeric substances, and various metabolites found in A. oryzae fermentation preparations are described in detail. With reference to the generally assumed key targets of postbiotics, their putative beneficial bioactivities in modulating the microbiota, improving epithelial barrier function, influencing immune responses, metabolic reactions and signaling through the nervous system are assessed. Drawing on existing literature and case studies, we highlight A. oryzae as a promising source of postbiotics, particularly in the context of animal health and nutrition. Challenges and opportunities in quality control are also addressed, with a focus on the necessity for standardized methods to fully harness the potential of fungal-based postbiotics. Overall, this article sheds light on the emerging field of A. oryzae-derived postbiotics and emphasizes the need for further research to fully realize their therapeutic potential.
Collapse
Affiliation(s)
- Yvonne Seidler
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Kai Lüersen
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (CONICET-UNL), Faculty of Chemical Engineering, National University of Litoral, Santa Fe, Argentina
| | - Ignacio R. Ipharraguerre
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| |
Collapse
|
15
|
Niamah AK, Al-Sahlany STG, Verma DK, Shukla RM, Patel AR, Tripathy S, Singh S, Baranwal D, Singh AK, Utama GL, Chávez González ML, Alhilfi WAH, Srivastav PP, Aguilar CN. Emerging lactic acid bacteria bacteriocins as anti-cancer and anti-tumor agents for human health. Heliyon 2024; 10:e37054. [PMID: 39286220 PMCID: PMC11402949 DOI: 10.1016/j.heliyon.2024.e37054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Modern cancer diagnostics and treatment options have greatly improved survival rates; the illness remains a major cause of mortality worldwide. Current treatments for cancer, such as chemotherapy, are not cancer-specific and may cause harm to healthy cells; therefore, it is imperative that new drugs for cancer be developed that are both safe and effective. It has been found that lactic acid bacteria (LAB) have the potential to produce bacteriocins, which could potentially offer a promising alternative for cancer treatment. They have been shown in several studies to be effective against cancer cells while having no effect on healthy cells. More research is needed to fully understand the potential of LAB bacteriocins as anti-cancer medicines, to find the appropriate dose and delivery route, and to conduct clinical trials to evaluate the effectiveness and safety of the products in human patients, as is suggested by this work. Furthermore, LAB bacteriocins may evolve into a significant new class of anti-cancer drugs and food products. Patients with cancer may have a safe and effective alternative treatment option in the form of anti-cancer foods and drugs. Therefore, the aim of this study is to provide an in-depth analysis of the recent breakthroughs and potential future technical advancements of significant bacteriocins that are produced by LAB, how these bacteriocins function, and how these bacteriocins may be utilized as an anti-cancer agent. In addition, the current analysis emphasizes the significant constraints and boundaries that bacteriocins face when they are used as an anti-cancer factor.
Collapse
Affiliation(s)
- Alaa Kareem Niamah
- Department of Food Science, College of Agriculture, University of Basrah, Basra City, Iraq
| | | | - Deepak Kumar Verma
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Rakesh Mohan Shukla
- Processing and Food Engineering Division, College of Agricultural Engineering and Technology (CoAE&T), Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, 190 025, Jammu and Kashmir, India
| | - Ami R Patel
- Division of Dairy Microbiology, Mansinhbhai Institute of Dairy and Food Technology-MIDFT, Dudhsagar Dairy Campus, Mehsana-384 002, Gujarat, India
| | - Soubhagya Tripathy
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Smita Singh
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab 140401, India
| | - Deepika Baranwal
- Department of Home Science, Arya Mahila PG College, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India
| | - Amit Kumar Singh
- Department of Post Harvest Technology, College of Horticulture, Banda University of Agriculture and Technology, Banda 210 001, Uttar Pradesh, India
| | - Gemilang Lara Utama
- Faculty of Agro-Industrial Technology, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Center for Environment and Sustainability Science, Universitas Padjadjaran, Bandung 40132, Indonesia
| | - Mónica L Chávez González
- Bioprocesses and Bioproducts Group, Food Research Department, School of Chemistry, Universidad Autónoma de Coahuila, Saltillo Campus., 25280, Coahuila, Mexico
| | | | - Prem Prakash Srivastav
- Agricultural and Food Engineering Department, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Cristobal Noe Aguilar
- Bioprocesses and Bioproducts Group, Food Research Department, School of Chemistry, Universidad Autónoma de Coahuila, Saltillo Campus., 25280, Coahuila, Mexico
| |
Collapse
|
16
|
Wei L, Wong D, Jeoh T, Marco ML. Intestinal delivery of encapsulated bacteriocin peptides in cross-linked alginate microcapsules. Food Res Int 2024; 188:114473. [PMID: 38823837 DOI: 10.1016/j.foodres.2024.114473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/25/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
Oral delivery of larger bioactive peptides (>20 amino acids) to the small intestine remains a challenge due to their sensitivity to proteolytic degradation and chemical denaturation during gastrointestinal transit. In this study, we investigated the capacity of crosslinked alginate microcapsules (CLAMs) formed by spray drying to protect Plantaricin EF (PlnEF) (C-EF) in gastric conditions and to dissolve and release PlnEF in the small intestine. PlnEF is an unmodified, two-peptide (PlnE: 33 amino acids; PlnF: 34 amino acids) bacteriocin produced by Lactiplantibacillus plantarum with antimicrobial and gut barrier protective properties. After 2 h incubation in simulated gastric fluid (SGF) (pH 1.5), 43.39 % ± 8.27 % intact PlnEF was liberated from the CLAMs encapsulates, as determined by an antimicrobial activity assay. Transfer of the undissolved fraction to simulated intestinal fluid (SIF) (pH 7) for another 2 h incubation resulted in an additional release of 16.13 % ± 4.33 %. No active PlnEF was found during SGF or sequential SIF incubations when pepsin (2,000 U/ml) was added to the SGF. To test PlnEF release in C-EF contained in a food matrix, C-EF was mixed in peanut butter (PB) (0.15 g C-EF in 1.5 g PB). A total of 12.52 % ± 9.09 % active PlnEF was detected after incubation of PB + C-EF in SGF without pepsin, whereas no activity was found when pepsin was included. Transfer of the remaining PB + C-EF fractions to SIF yielded the recovery of 46.67 % ± 13.09 % and 39.42 % ± 11.53 % active PlnEF in the SIF following exposure to SGF and to SGF with pepsin, respectively. Upon accounting for the undissolved fraction after SIF incubation, PlnEF was fully protected in the CLAMs-PB mixture and there was not a significant reduction in active PlnEF when pepsin was present. These results show that CLAMs alone do not guard PlnEF bacteriocin peptides from gastric conditions, however, mixing them in PB protected against proteolysis and improved intestinal release.
Collapse
Affiliation(s)
- Lei Wei
- Department of Food Science and Technology, University of California, Davis, USA
| | - Dana Wong
- Department of Biological and Agricultural Engineering, University of California, Davis, USA
| | - Tina Jeoh
- Department of Biological and Agricultural Engineering, University of California, Davis, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, USA.
| |
Collapse
|
17
|
Fijan S, Kürti P, Rozman U, Šostar Turk S. A critical assessment of microbial-based antimicrobial sanitizing of inanimate surfaces in healthcare settings. Front Microbiol 2024; 15:1412269. [PMID: 38933019 PMCID: PMC11199901 DOI: 10.3389/fmicb.2024.1412269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
The global rise in antimicrobial resistance (AMR) poses a significant public health threat, especially in healthcare settings, where controlling the spread of antimicrobial genes is crucial. While person-to-person transmission remains the primary route for healthcare-associated infections (HAIs), hospital surfaces serve as key reservoirs for antimicrobial-resistant microorganisms. Regular cleaning and disinfection of these surfaces are essential. Microbial-based products for sanitizing hospital surfaces have emerged as promising tools to combat HAIs and AMR. However, a review of 32 publications found inconsistencies and potential risks. A total of 15 publications included hospital-based trials, while the rest were either in vitro or in situ assays, reviews, book chapters, or commentaries. In most of the hospital-based studies, specific strains of applied microorganisms were not identified, and the term "probiotic" was inaccurately used. These products mainly featured spores from Bacillus and Priestia genera, which was mainly hypothesized to work through competitive exclusion. Most hospital-based studies have shown that the application of microbial-based products resulted in a significant reduction in pathogens on surfaces, thereby contributing to a decrease in the incidence of healthcare-associated infections (HAIs). Further research is however needed to understand the effectiveness, mechanisms of action, and safety of microbial-based sanitizing agents. Strain-level identification is crucial for safety assessments, yet many reviewed products lacked this information. Consequently, there is a need for rigorous safety evaluations within existing regulatory frameworks to ensure the efficacy and safety of microbial-based cleaning products in healthcare settings.
Collapse
Affiliation(s)
- Sabina Fijan
- Faculty of Health Sciences, University of Maribor, Maribor, Slovenia
| | | | - Urška Rozman
- Faculty of Health Sciences, University of Maribor, Maribor, Slovenia
| | - Sonja Šostar Turk
- Faculty of Health Sciences, University of Maribor, Maribor, Slovenia
| |
Collapse
|
18
|
Mukherjee A, Breselge S, Dimidi E, Marco ML, Cotter PD. Fermented foods and gastrointestinal health: underlying mechanisms. Nat Rev Gastroenterol Hepatol 2024; 21:248-266. [PMID: 38081933 DOI: 10.1038/s41575-023-00869-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2023] [Indexed: 12/20/2023]
Abstract
Although fermentation probably originally developed as a means of preserving food substrates, many fermented foods (FFs), and components therein, are thought to have a beneficial effect on various aspects of human health, and gastrointestinal health in particular. It is important that any such perceived benefits are underpinned by rigorous scientific research to understand the associated mechanisms of action. Here, we review in vitro, ex vivo and in vivo studies that have provided insights into the ways in which the specific food components, including FF microorganisms and a variety of bioactives, can contribute to health-promoting activities. More specifically, we draw on representative examples of FFs to discuss the mechanisms through which functional components are produced or enriched during fermentation (such as bioactive peptides and exopolysaccharides), potentially toxic or harmful compounds (such as phytic acid, mycotoxins and lactose) are removed from the food substrate, and how the introduction of fermentation-associated live or dead microorganisms, or components thereof, to the gut can convey health benefits. These studies, combined with a deeper understanding of the microbial composition of a wider variety of modern and traditional FFs, can facilitate the future optimization of FFs, and associated microorganisms, to retain and maximize beneficial effects in the gut.
Collapse
Affiliation(s)
| | - Samuel Breselge
- Teagasc Food Research Centre, Moorepark, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Eirini Dimidi
- Department of Nutritional Sciences, King's College London, London, UK
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, CA, USA
| | - Paul D Cotter
- Teagasc Food Research Centre, Moorepark, Cork, Ireland.
- APC Microbiome Ireland, Cork, Ireland.
- VistaMilk, Cork, Ireland.
| |
Collapse
|
19
|
Todorov SD, Wachsman M, Tomé E, Vaz-Velho M, Ivanova IV. Plasmid-Associated Bacteriocin Produced by Pediococcus pentosaceus Isolated from Smoked Salmon: Partial Characterization and Some Aspects of his Mode of Action. Probiotics Antimicrob Proteins 2024; 16:394-412. [PMID: 36928486 DOI: 10.1007/s12602-023-10059-5] [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] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
Strain ST3Ha, isolated from commercially available smoked salmon, was identified as Pediococcus pentosaceus based on biochemical and physiological tests and 16S rRNA sequencing. Strain ST3Ha produces a class IIa bacteriocin active against lactic acid bacteria, Listeria monocytogenes and Enterococcus faecalis. The antimicrobial peptide was inactivated by proteolytic enzymes, confirming his proteinaceous nature, but was not affected when treated with α-amylase, SDS, Tween 20, Tween 80, urea, and EDTA. No change in activity was recorded after 2 h at pH values between 2.0 and 9.0 and after treatment at 100 °C for 120 min or 121 °C for 15 min. The mode of action against Listeria ivanovii subsp. ivanovii ATCC 19119 and E. faecalis ATCC 19443 was bactericidal, resulting in cell lyses and enzyme leakage. The highest level of activity (1.6 × 106 AU/mL) was recorded when cells were grown at 37 °C or 30 °C in MRS broth (pH 6.5). Antimicrobial peptide ST3Ha adsorbs at high levels to the sensitive test organisms on strain-specific manner and depending on incubation temperature, environmental pH, and presence of supplemented chemicals. Based on PCR analysis, P. pentosaceus ST3Ha harbor a 1044-bp plasmid-associated fragment corresponding in size to that recorded for pediocin PA-1. Sequencing of the fragment revealed a gene identical to pedB, reported for pediocin PA-1. The combined application of the low levels (below MIC) of ciprofloxacin and bacteriocin ST3Ha results in the synergetic effect in the inhibition of L. ivanovii subsp. ivanovii ATCC 19119. Expressed by P. pentosaceus ST3Ha, bacteriocin was characterized as low cytotoxic, a characteristic relevant for its application in food industry and/or in human and veterinary medical practices.
Collapse
Affiliation(s)
- Svetoslav Dimitrov Todorov
- ProBacLab, Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, 37554, Republic of Korea.
- ProBacLab, Laboratório de Microbiologia de Alimentos, Faculdade de Ciências Farmacêuticas, Departamento de Alimentos e Nutrição Experimental, Universidade de São Paulo, SP, São Paulo, Brazil.
- CISAS-Center for Research and Development in Agrifood Systems and Sustainability, Escola Superior de Tecnologia E Gestão, Instituto Politécnico de Viana Do Castelo, Viana Do Castelo, Portugal.
- Faculty of Biology, Department of General and Applied Microbiology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Blvd, 1164, Sofia, Bulgaria.
| | - Monica Wachsman
- Laboratorio de Virología, Facultad de Ciencias Exactas Y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Ciudad Universitaria, Pabellon 2, Piso 4, C1428EGA, Buenos Aires, Argentina
| | - Elisabetta Tomé
- Tecnologia de Alimentos, Universidad Metropolitana, Caracas, Venezuela
| | - Manuela Vaz-Velho
- CISAS-Center for Research and Development in Agrifood Systems and Sustainability, Escola Superior de Tecnologia E Gestão, Instituto Politécnico de Viana Do Castelo, Viana Do Castelo, Portugal
| | - Iskra Vitanova Ivanova
- Faculty of Biology, Department of General and Applied Microbiology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Blvd, 1164, Sofia, Bulgaria
| |
Collapse
|
20
|
Huidrom S, Ngashangva N, Khumlianlal J, Sharma KC, Mukherjee PK, Devi SI. Genomic insights from Lactiplantibacillus plantarum BRD3A isolated from Atingba, a traditional fermented rice-based beverage and analysis of its potential for probiotic and antimicrobial activity against Methicillin-resistant Staphylococcus aureus. Front Microbiol 2024; 15:1357818. [PMID: 38628861 PMCID: PMC11019378 DOI: 10.3389/fmicb.2024.1357818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Lactiplantibacillus plantarum BRD3A was isolated from Atingba, a traditional fermented rice-based beverage of Manipur. Its genomic sequence has 13 contigs and its genome size is 3,320,817 bp with a guanine-cytosine (GC) ratio of 44.6%. It comprises 3185 genes including 3112 coding sequences (CDSs), 73 RNAs (including 66 tRNAs and others), and one clustered regularly interspaced short palindromic repeat (CRISPR) array. A comparative and phylogenetic analysis with the Lp. plantarum genome shows that this strain has close similarity with other Lp. plantarum strains and about 99% average nucleotide identity. Functional annotation using evolutionary genealogy of genes-non-supervised orthologous groups (EggNOG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) reveals genes associated with various biological processes such as metabolism, genetic information processing, and transport functions. Furthermore, the strain harbors bacteriocins like plantaricin E, Plantaricin F, and Enterocin X categorized under class IIb by the BAGEL4 database, indicating its potential antimicrobial properties. Additionally, AntiSMASH web server predicted four secondary regions-T3PKS, terpene, cyclic lactone inducer, and ribosomally synthesized and post-translationally modified peptide (RiPP)-suggesting an even higher antimicrobial potential. We validated the antimicrobial activity of Lp. plantarum BRD3A through in vitro experiments in which it exhibited promising bactericidal effects on methicillin-resistant Staphylococcus aureus, inhibiting their biofilm growth. These findings indicate the potential of Lp. plantarum BRD3A to be used as an alternative to conventional antibiotics.
Collapse
Affiliation(s)
- Surmani Huidrom
- Microbial Resources Division, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | - Ng Ngashangva
- Microbial Resources Division, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
| | - Joshua Khumlianlal
- Microbial Resources Division, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Bhubaneswar, India
| | | | - Pulok Kumar Mukherjee
- Microbial Resources Division, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
| | - Sarangthem Indira Devi
- Microbial Resources Division, Institute of Bioresources and Sustainable Development (IBSD), Imphal, Manipur, India
| |
Collapse
|
21
|
Dong S, Li L, Hao F, Fang Z, Zhong R, Wu J, Fang X. Improving quality of poultry and its meat products with probiotics, prebiotics, and phytoextracts. Poult Sci 2024; 103:103287. [PMID: 38104412 PMCID: PMC10966786 DOI: 10.1016/j.psj.2023.103287] [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] [Received: 09/05/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Remarkable changes have occurred in poultry farming and meat processing in recent years, driven by advancements in breeding technology, feed processing technology, farming conditions, and management practices. The incorporation of probiotics, prebiotics, and phytoextracts has made significant contributions to the development of poultry meat products that promote both health and functionality throughout the growth phase and during meat processing. Poultry fed with these substances improve meat quality, while incorporating probiotics, prebiotics, and phytoextracts in poultry processing, as additives or supplements, inhibits pathogens and offers health benefits to consumers. However, it is vital to assess the safety of functional fermented meat products containing these compounds and their potential effects on consumer health. Currently, there's still uncertainty in these aspects. Additionally, research on utilizing next-generation probiotic strains and synergistic combinations of probiotics and prebiotics in poultry meat products is in its early stages. Therefore, further investigation is required to gain a comprehensive understanding of the beneficial effects and safety considerations of these substances in poultry meat products in the future. This review offered a comprehensive overview of the applications of probiotics and prebiotics in poultry farming, focusing on their effects on nutrient utilization, growth efficiency, and gut health. Furthermore, potential of probiotics, prebiotics, and phytoextracts in enhancing poultry meat production was explored for improved health benefits and functionality, and possible issues associated with the use of these substances were discussed. Moreover, the conclusions drawn from this review and potential future perspectives in this field are presented.
Collapse
Affiliation(s)
- Sashuang Dong
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512000, PR China
| | - Lanyin Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China
| | - Fanyu Hao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China
| | - Ziying Fang
- Weiran Food Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518000, PR China
| | - Ruimin Zhong
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512000, PR China
| | - Jianfeng Wu
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China.
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
22
|
Gao Y, Li D. Antibacterial mode of action of garviecin LG34 against Gram-negative bacterium Salmonella typhimurium. FEMS Microbiol Lett 2024; 371:fnae066. [PMID: 39138064 DOI: 10.1093/femsle/fnae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 08/15/2024] Open
Abstract
Garviecin LG34 produced by Lactococcus garvieae LG34 exhibits wide-spectrum antibacterial activity against both Gram-positive and Gram-negative bacteria. This work aimed at clarifying the antibacterial mode of action of garviecin LG34 against Gram-negative bacterium Salmonella typhimurium. To determine the concentration for the bacteriocin antimicrobial mode experiments, the minimum inhibitory concentration of garviecin LG34 against S. typhimurium CICC21484 was determined as 0.25 mg/ml. Garviecin LG34 decreased the viable count of S. typhimurium CICC21484 and its antibacterial activity was the dose and time dependant. Garviecin LG34 led to the dissipation of transmembrane potential, the rise in the extracellular conductivity, UV-absorbing material at 260 nm, and LDH level of S. typhimurium CICC21484. Scanning electron micrographs results shown that garviecin LG34 cause dramatic deformation and fragmentation including the flagellum shedding, pores formation in surface, and even completely breakage of S. typhimurium cell. Moreover, garviecin LG34 decreased the intracellular ATP level. The results of this study demonstrated that garviecin LG34 can destroy cell structure, increase membrane permeability of S. typhimurium, thereby might be used as biopreservative for treating food borne and salmonellosis resulting from Gram-negative bacterium S. typhimurium.
Collapse
Affiliation(s)
- Yurong Gao
- School of Biological and Environmental Engineering, Chaohu University, Bantang Road No.1, Chaohu Economic Development Zone, Hefei 238024, China
- Chaohu Regional Collaborative Technology Service Center for Rural Revitalization, Bantang Road No.1, Chaohu Economic Development Zone, Hefei 238024, China
| | - Dapeng Li
- School of Biological and Environmental Engineering, Chaohu University, Bantang Road No.1, Chaohu Economic Development Zone, Hefei 238024, China
- Chaohu Regional Collaborative Technology Service Center for Rural Revitalization, Bantang Road No.1, Chaohu Economic Development Zone, Hefei 238024, China
| |
Collapse
|
23
|
Gu Q, Yan J, Lou Y, Zhang Z, Li Y, Zhu Z, Liu M, Wu D, Liang Y, Pu J, Zhao X, Xiao H, Li P. Bacteriocins: Curial guardians of gastrointestinal tract. Compr Rev Food Sci Food Saf 2024; 23:e13292. [PMID: 38284593 DOI: 10.1111/1541-4337.13292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024]
Abstract
The human gastrointestinal (GI) tract microbiome secretes various metabolites that play pivotal roles in maintaining host physiological balance and influencing disease progression. Among these metabolites, bacteriocins-small, heat-stable peptides synthesized by ribosomes-are notably prevalent in the GI region. Their multifaceted benefits have garnered significant interest in the scientific community. This review comprehensively explores the methods for mining bacteriocins (traditional separation and purification, bioinformatics, and artificial intelligence), their effects on the stomach and intestines, and their complex bioactive mechanisms. These mechanisms include flora regulation, biological barrier restoration, and intervention in epithelial cell pathways. By detailing each well-documented bacteriocin, we reveal the diverse ways in which bacteriocins interact with the GI environment. Moreover, the future research direction is prospected. By further studying the function and interaction of intestinal bacteriocins, we can discover new pharmacological targets and develop drugs targeting intestinal bacteriocins to regulate and improve human health. It provides innovative ideas and infinite possibilities for further exploration, development, and utilization of bacteriocins. The inevitable fact is that the continuously exploration of bacteriocins is sure to bring the promising future for demic GI health understanding and interference strategy.
Collapse
Affiliation(s)
- Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Yan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yeqing Lou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zihao Zhang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yonglu Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zichun Zhu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Manman Liu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Danli Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ying Liang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Pu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodan Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
24
|
Baindara P, Mandal SM. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1732. [PMID: 38136766 PMCID: PMC10740742 DOI: 10.3390/antibiotics12121732] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Due to huge diversity and dynamic competition, the human gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that play an important role in human health. The gut microbiome has an important role in maintaining gut homeostasis by the AMPs and by interacting with other human organs via established connections such as the gut-lung, and gut-brain axis. Additionally, gut AMPs play a synergistic role with other gut microbiota and antimicrobials to maintain gut homeostasis by fighting against multi-antibiotic resistance (MAR) bacteria. Further, conventional antibiotics intake creates a synergistic evolutionary pressure for gut AMPs, where antibiotics and gut AMPs fight synergistically against MAR. Overall, gut AMPs are evolving under a complex and highly synergistic co-evolutionary pressure created by the various interactions between gut microbiota, gut AMPs, and antibiotics; however, the complete mechanism is not well understood. The current review explores the synergistic action of gut AMPs and antibiotics along with possibilities to fight against MAR bacteria.
Collapse
Affiliation(s)
- Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Santi M. Mandal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
25
|
Yang R, Ahmad S, Liu H, Xu Q, Yin C, Liu Y, Zhang H, Yan H. Biodegradation of Cholesterol by Enterococcus faecium YY01. Microorganisms 2023; 11:2979. [PMID: 38138122 PMCID: PMC10745435 DOI: 10.3390/microorganisms11122979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Cholesterol (CHOL) is one of the risk factors causing the blockage of the arterial wall, atherosclerosis, coronary heart disease, and other serious cardiovascular diseases. Here, a promising bacterial strain for biodegrading CHOL was successfully isolated from the gut of healthy individuals and identified as Enterococcus faecium YY01 with an analysis of the 16S rDNA sequence. An initial CHOL of 1.0 g/L was reduced to 0.5 g/L in 5 days, and glucose and beef extract were found to be optimal carbon and nitrogen sources for the rapid growth of YY01, respectively. To gain further insight into the mechanisms underlying CHOL biodegradation, the draft genome of YY01 was sequenced using Illumina HiSeq. Choloylglycine hydrolase, acyltransferase, and alkyl sulfatase was encoded by gene0586, gene1890, and gene2442, which play crucial roles in converting 3α, 7α, 12α-trihydroxy-5β-choranic acid to choline-CoA and then choline-CoA to bile acid. Notably, choloylglycine hydrolase was closely related to the biosynthesis of both primary and secondary bile acid. The findings of this study provide valuable insights into the metabolism pathway of CHOL biodegradation by YY01 and offer a potential avenue for the development of bacterioactive drugs against hypercholesterolemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hai Yan
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; (R.Y.); (S.A.); (H.L.); (Q.X.); (C.Y.); (Y.L.); (H.Z.)
| |
Collapse
|
26
|
Martinelli S, Lamminpää I, Dübüş EN, Sarıkaya D, Niccolai E. Synergistic Strategies for Gastrointestinal Cancer Care: Unveiling the Benefits of Immunonutrition and Microbiota Modulation. Nutrients 2023; 15:4408. [PMID: 37892482 PMCID: PMC10610426 DOI: 10.3390/nu15204408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Gastrointestinal (GI) cancers are a group of highly prevalent malignant tumors affecting the gastrointestinal tract. Globally, one in four cancer cases and one in three cancer deaths are estimated to be GI cancers. They can alter digestive and absorption functions, leading to severe malnutrition which may worsen the prognosis of the patients. Therefore, nutritional intervention and monitoring play a fundamental role in managing metabolic alterations and cancer symptoms, as well as minimizing side effects and increasing the effectiveness of chemotherapy. In this scenario, the use of immunonutrients that are able to modulate the immune system and the modification/regulation of the gut microbiota composition have gained attention as a possible strategy to improve the conditions of these patients. The complex interaction between nutrients and microbiota might contribute to maintaining the homeostasis of each individual's immune system; therefore, concurrent use of specific nutrients in combination with traditional cancer treatments may synergistically improve the overall care of GI cancer patients. This work aims to review and discuss the role of immunonutrition and microbiota modulation in improving nutritional status, postoperative recovery, and response to therapies in patients with GI cancer.
Collapse
Affiliation(s)
- Serena Martinelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Ingrid Lamminpää
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| | - Eda Nur Dübüş
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Dilara Sarıkaya
- Department of Nutrition and Dietetics, Gazi University, 06560 Ankara, Turkey; (E.N.D.); (D.S.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Firenze, Italy; (S.M.); (I.L.)
| |
Collapse
|
27
|
Koizumi J, Nakase K, Noguchi N, Nakaminami H. Avidumicin, a novel cyclic bacteriocin, produced by Cutibacterium avidum shows anti-Cutibacterium acnes activity. J Antibiot (Tokyo) 2023; 76:511-521. [PMID: 37264118 DOI: 10.1038/s41429-023-00635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/07/2023] [Accepted: 05/12/2023] [Indexed: 06/03/2023]
Abstract
The prevalence of antimicrobial-resistant Cutibacterium acnes in acne patients has increased owing to inappropriate antimicrobial use. Commensal skin bacteria may play an important role in maintaining the balance of the skin microbiome by producing antimicrobial substances. Inhibition of Cu. acnes overgrowth can prevent the development and exacerbation of acne vulgaris. Here, we evaluated skin bacteria with anti-Cu. acnes activity. Growth inhibition activity against Cu. acnes was tested using 122 strains isolated from the skin of healthy volunteers and acne patients. Comparative genomic analysis of the bacterium with or without anti-Cu. acnes activity was conducted. The anti-Cu. acnes activity was confirmed by cloning an identified gene cluster and chemically synthesized peptides. Cu. avidum ATCC25577 and 89.7% of the Cu. avidum clinical isolates (26/29 strains) inhibited Cu. acnes growth. The growth inhibition activity was also found against other Cutibacterium, Lactiplantibacillus, and Corynebacterium species, but not against Staphylococcus species. The genome sequence of Cu. avidum showed a gene cluster encoding a novel bacteriocin named avidumicin. The precursor protein encoded by avdA undergoes post-translational modifications, supposedly becoming a circular bacteriocin. The anti-Cu. acnes activity of avidumicin was confirmed by Lactococcus lactis MG1363 carrying avdA. The C-terminal region of the avidumicin may be essential for anti-Cu. acnes activity. A commensal skin bacterium, Cu. avidum, producing avidumicin has anti-Cu. acnes activity. Therefore, avidumicin is a novel cyclic bacteriocin with a narrow antimicrobial spectrum. These findings suggest that Cu. avidum and avidumicin represent potential alternative agents in antimicrobial therapy for acne vulgaris.
Collapse
Affiliation(s)
- Juri Koizumi
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Keisuke Nakase
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Norihisa Noguchi
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Hidemasa Nakaminami
- Department of Clinical Microbiology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
28
|
Bu Y, Liu Y, Liu Y, Cao J, Zhang Z, Yi H. Protective Effects of Bacteriocin-Producing Lactiplantibacillus plantarum on Intestinal Barrier of Mice. Nutrients 2023; 15:3518. [PMID: 37630708 PMCID: PMC10459803 DOI: 10.3390/nu15163518] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Bacteriocins are crucial metabolites of probiotics that display beneficial functions. The intestinal barrier is an important target on which probiotics exert their intestinal health activity. However, the impacts of bacteriocin-producing probiotics on the intestinal barrier are unclear. In this study, the effects of bacteriocin-producing Lactiplantibacillus plantarum Q7 and L. plantarum F3-2 on the intestinal barrier of mice were explored. It was shown that L. plantarum Q7 promoted the expression of mucin MUC2 to enhance the protection provided by the intestinal mucus layer. L. plantarum Q7 up-regulated the gene expression of intestinal tight junction proteins ZO-1 and JAM-1 significantly, and L. plantarum F3-2 up-regulated ZO-1 and Claudin-1 markedly, which exhibited tight junction intestinal barrier function. The two strains promoted the release of IgA and IgG at varying degrees. The antimicrobial peptide gene RegIIIγ was up-regulated markedly, and the gene expression of inflammatory cytokines appeared to exhibit an upward trend with L. plantarum Q7 treatment, so as to enhance intestinal immune regulation function. Furthermore, L. plantarum Q7 and L. plantarum F3-2 increased the abundance of the beneficial bacteria Muribaculaceae, inhibited the growth of the harmful bacteria Parabacteroides, and facilitated the synthesis of total short-chain fatty acids (SCFAs), which seemed to favor the prevention of metabolic diseases. Our results suggested that L. plantarum Q7 and L. plantarum F3-2 showed strain specificity in their protective effects on the intestinal chemical, physical, immunological and biological barriers of mice, which provided theoretical support for the selective utilization of bacteriocin-producing strains to regulate host health.
Collapse
Affiliation(s)
- Yushan Bu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yisuo Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yinxue Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
| | - Jiayuan Cao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
| | - Zhe Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (J.C.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| |
Collapse
|
29
|
Bu Y, Liu Y, Zhang T, Liu Y, Zhang Z, Yi H. Bacteriocin-Producing Lactiplantibacillus plantarum YRL45 Enhances Intestinal Immunity and Regulates Gut Microbiota in Mice. Nutrients 2023; 15:3437. [PMID: 37571374 PMCID: PMC10421436 DOI: 10.3390/nu15153437] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Bacteriocins production is one of important beneficial characteristics of probiotics, which has antibacterial property against intestinal pathogens and is helpful for regulating intestinal flora. To investigate the impact of bacteriocin-producing probiotics on gut microecology, bacteriocin-producing Lactiplantibacillus plantarum YRL45 was orally administered to mice. The results revealed that it promoted the release of cytokines and improved the phagocytic activity of peritoneal macrophages to activate the immune regulation system. L. plantarum YRL45 was conducive to maintaining the morphology of colon tissue without inflammation and increasing the ratio of villus height to crypt depth in the ileum. The gene expression levels of Muc2, ZO-1 and JAM-1 were significantly up-regulated in the ileum and colon, and the gene expression of Cramp presented an upward trend with L. plantarum YRL45 intervention. Moreover, L. plantarum YRL45 remarkably enhanced the levels of immunoglobulins sIgA, IgA and IgG in the intestine of mice. The 16S rRNA gene analysis suggested that L. plantarum YRL45 administration up-regulated the relative abundance of the beneficial bacteria Muribaculaceae and Akkermansia, down-regulated the abundance of the pathogenic bacteria Lachnoclostridium, and promoted the production of acetic acid, propionic acid and total short-chain fatty acids (SCFAs) in mice feces. Our findings indicated that L. plantarum YRL45 had the potential to be developed as a novel probiotic to regulate the intestinal barrier by altering gut microbiota to enhance intestinal immunity and ameliorate intestinal flora balance.
Collapse
Affiliation(s)
- Yushan Bu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yisuo Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Tai Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yinxue Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
| | - Zhe Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China; (Y.B.); (Y.L.); (T.Z.); (Y.L.)
- Food Laboratory of Zhongyuan, Luohe 462300, China
| |
Collapse
|
30
|
González-Orozco BD, Kosmerl E, Jiménez-Flores R, Alvarez VB. Enhanced probiotic potential of Lactobacillus kefiranofaciens OSU-BDGOA1 through co-culture with Kluyveromyces marxianus bdgo-ym6. Front Microbiol 2023; 14:1236634. [PMID: 37601389 PMCID: PMC10434783 DOI: 10.3389/fmicb.2023.1236634] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Due to the increasing consumer demand for the development and improvement of functional foods containing probiotics, new probiotic candidates need to be explored as well as novel means to enhance their beneficial effects. Lactobacillus kefiranofaciens OSU-BDGOA1 is a strain isolated from kefir grains that has demonstrated probiotic traits. This species is the main inhabitant of kefir grains and is responsible for the production of an exopolysaccharide (EPS) whit vast technological applications and potential bioactivities. Research has shown that interkingdom interactions of yeast and lactic acid bacteria can enhance metabolic activities and promote resistance to environmental stressors. Methods Comparative genomic analyses were performed to distinguish OSU-BDGOA1 from other strains of the same species, and the genome was mined to provide molecular evidence for relevant probiotic properties. We further assessed the cumulative effect on the probiotic properties of OSU-BDGOA1 and Kluyveromyces marxianus bdgo-ym6 yeast co-culture compared to monocultures. Results Survival during simulated digestion assessed by the INFOGEST digestion model showed higher survival of OSU-BDGOA1 and bdgo-ym6 in co-culture. The adhesion to intestinal cells assessed with the Caco-2 intestinal cell model revealed enhanced adhesion of OSU-BDGOA1 in co-culture. The observed increase in survival during digestion could be associated with the increased production of EPS during the late exponential and early stationary phases of co-culture that, by enhancing co-aggregation between the yeast and the bacterium, protects the microorganisms from severe gastrointestinal conditions as observed by SEM images. Immune modulation and barrier function for recovery and prevention of flagellin-mediated inflammation by Salmonella Typhimurium heat-killed cells (HKSC) in Caco-2 cells were also measured. OSU-BDGOA1 in mono- and co-culture regulated inflammation through downregulation of pro-inflammatory cytokine expression and increased membrane barrier integrity assessed by TEER, FD4 permeability, and expression of tight junctions. Discussion The results of the study warrant further research into the application of co-cultures of yeast and LAB in functional probiotic products and the potential to increase EPS production by co-culture strategies.
Collapse
Affiliation(s)
| | | | | | - Valente B. Alvarez
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
31
|
Nicholas-Haizelden K, Murphy B, Hoptroff M, Horsburgh MJ. Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products. Microorganisms 2023; 11:1899. [PMID: 37630459 PMCID: PMC10456854 DOI: 10.3390/microorganisms11081899] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Bioprospecting is the discovery and exploration of biological diversity found within organisms, genetic elements or produced compounds with prospective commercial or therapeutic applications. The human skin is an ecological niche which harbours a rich and compositional diversity microbiome stemming from the multifactorial interactions between the host and microbiota facilitated by exploitable effector compounds. Advances in the understanding of microbial colonisation mechanisms alongside species and strain interactions have revealed a novel chemical and biological understanding which displays applicative potential. Studies elucidating the organismal interfaces and concomitant understanding of the central processes of skin biology have begun to unravel a potential wealth of molecules which can exploited for their proposed functions. A variety of skin-microbiome-derived compounds display prospective therapeutic applications, ranging from antioncogenic agents relevant in skin cancer therapy to treatment strategies for antimicrobial-resistant bacterial and fungal infections. Considerable opportunities have emerged for the translation to personal care products, such as topical agents to mitigate various skin conditions such as acne and eczema. Adjacent compound developments have focused on cosmetic applications such as reducing skin ageing and its associated changes to skin properties and the microbiome. The skin microbiome contains a wealth of prospective compounds with therapeutic and commercial applications; however, considerable work is required for the translation of in vitro findings to relevant in vivo models to ensure translatability.
Collapse
Affiliation(s)
- Keir Nicholas-Haizelden
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| | - Barry Murphy
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Michael Hoptroff
- Unilever Research & Development, Port Sunlight, Wirral CH63 3JW, UK; (B.M.); (M.H.)
| | - Malcolm J. Horsburgh
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK;
| |
Collapse
|
32
|
Thoda C, Touraki M. Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment. Microorganisms 2023; 11:1898. [PMID: 37630458 PMCID: PMC10456921 DOI: 10.3390/microorganisms11081898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Colorectal cancer (CRC) is a multifactorial disease with increased morbidity and mortality rates globally. Despite advanced chemotherapeutic approaches for the treatment of CRC, low survival rates due to the regular occurrence of drug resistance and deleterious side effects render the need for alternative anticancer agents imperative. Accumulating evidence supports that gut microbiota imbalance precedes the establishment of carcinogenesis, subsequently contributing to cancer progression and response to anticancer therapy. Manipulation of the gut microbiota composition via the administration of probiotic-derived bioactive compounds has gradually attained the interest of scientific communities as a novel therapeutic strategy for CRC. These compounds encompass miscellaneous metabolic secreted products of probiotics, including bacteriocins, short-chain fatty acids (SCFAs), lactate, exopolysaccharides (EPSs), biosurfactants, and bacterial peptides, with profound anti-inflammatory and antiproliferative properties. This review provides a classification of postbiotic types and a comprehensive summary of the current state of research on their biological role against CRC. It also describes how their intricate interaction with the gut microbiota regulates the proper function of the intestinal barrier, thus eliminating gut dysbiosis and CRC development. Finally, it discusses the future perspectives in precision-medicine approaches as well as the challenges of their synthesis and optimization of administration in clinical studies.
Collapse
Affiliation(s)
| | - Maria Touraki
- Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece;
| |
Collapse
|
33
|
Laya A, Wangso H, Fernandes I, Djakba R, Oliveira J, Carvalho E. Bioactive Ingredients in Traditional Fermented Food Condiments: Emerging Products for Prevention and Treatment of Obesity and Type 2 Diabetes. J FOOD QUALITY 2023; 2023:1-26. [DOI: 10.1155/2023/5236509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Obesity and type 2 diabetes (T2D) are severe metabolic diseases due to inappropriate lifestyle and genetic factors and their prevention/treatment cause serious problems. Therefore, searching for effective and safe approaches to control obesity and T2D is an essential challenge. This study presents the knowledge regarding the possible use of traditional fermented condiments (TFC), a known major source of bioactive compounds (BACs), as an adjuvant treatment for obesity and T2D. Data on antiobesity, antidiabetic, and different mechanisms of BACs action of TFC were collected using a methodical search in PubMed, Scopus databases, Web of Science, SciELO, and the Cochrane Library. We discuss the mechanisms by which BCs prevent or treat obesity and T2D. The effects of TFC on obesity and T2D have been found both in animal, human, and clinical studies. The findings demonstrated that BACs in TFC confer potential promising antiobesity and antidiabetic effects. Because of the potential therapeutic significance of bioactive ingredients, the consumption of TFC could be recommended as a functional condiment. Nevertheless, further investigation is required in more clinical studies of TFC to support the formulation of functional fermented condiments and nutraceutical and pharmaceutical applications.
Collapse
Affiliation(s)
- Alphonse Laya
- Department of Biological Sciences, Faculty of Science, University of Maroua, Maroua, Cameroon
- Center for Neuroscience and Cell Biology, Faculdade de Medicina, University of Coimbra, Rua Larga, Polo I, 1º Andar, Coimbra 3004-504, Portugal
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão, Rua Dom Francisco de Lemos, Coimbra 3030-789, Portugal
- APDP-Portuguese Diabetes Association, Lisbon 1250-189, Portugal
| | - Honoré Wangso
- Department of Biological Sciences, Faculty of Science, University of Maroua, Maroua, Cameroon
| | - Iva Fernandes
- Laboratório Associado para a Química Verde-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, Porto 4169-007, Portugal
| | - Raphaël Djakba
- Department of Biological Sciences, Faculty of Science, University of Maroua, Maroua, Cameroon
| | - Joana Oliveira
- Laboratório Associado para a Química Verde-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, Porto 4169-007, Portugal
| | - Eugenia Carvalho
- Center for Neuroscience and Cell Biology, Faculdade de Medicina, University of Coimbra, Rua Larga, Polo I, 1º Andar, Coimbra 3004-504, Portugal
- Instituto de Investigação Interdisciplinar, University of Coimbra, Casa Costa Alemão, Rua Dom Francisco de Lemos, Coimbra 3030-789, Portugal
- APDP-Portuguese Diabetes Association, Lisbon 1250-189, Portugal
| |
Collapse
|
34
|
Oftedal TF, Diep DB. Flow cytometric detection of vancomycin-resistant Enterococcus faecium in urine using fluorescently labelled enterocin K1. Sci Rep 2023; 13:10930. [PMID: 37414859 PMCID: PMC10325980 DOI: 10.1038/s41598-023-38114-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 07/03/2023] [Indexed: 07/08/2023] Open
Abstract
A urinary tract infection (UTI) occurs when bacteria enter and multiply in the urinary system. The infection is most often caused by enteric bacteria that normally live in the gut, which include Enterococcus faecium. Without antibiotic treatment, UTIs can progress to life-threatening septic shock. Early diagnosis and identification of the pathogen will reduce antibiotic use and improve patient outcomes. In this work, we develop and optimize a cost-effective and rapid (< 40 min) method for detecting E. faecium in urine. The method uses a fluorescently labelled bacteriocin enterocin K1 (FITC-EntK1) that binds specifically to E. faecium and is then detected using a conventional flow cytometer. Using this detection assay, urine containing E. faecium was identified by an increase in the fluorescent signals by 25-73-fold (median fluorescence intensity) compared to control samples containing Escherichia coli or Staphylococcus aureus. The method presented in this work is a proof of concept showing the potential of bacteriocins to act as specific probes for the detection of specific bacteria, such as pathogens, in biological samples.
Collapse
Affiliation(s)
- Thomas F Oftedal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway.
| | - Dzung B Diep
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
35
|
Ye J, Fugaban JII, Dioso CM, Vazquez Bucheli JE, Choi GH, Kim B, Holzapfel WH, Todorov SD. Bacillus Strains Isolated from Korean Fermented Food Products with Antimicrobial Activity Against Staphylococci, an Alternative for Control of Antibiotic-Resistant Pathogens. Foodborne Pathog Dis 2023; 20:279-293. [PMID: 37366658 DOI: 10.1089/fpd.2023.0019] [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: 06/28/2023] Open
Abstract
Multidrug resistance in foodborne and clinical pathogens is a worldwide health problem. The urgent need for new alternatives to the existing antibiotics is emerging. Bacteriocin-like inhibitory substances can be considered part of the new generation of antimicrobials, which can be potentially applied in the food industry and health care practices. This study aimed to select Bacillus strains with antimicrobial activity against Staphylococcus spp. with future application in the formulation of pharmaceutical antimicrobial preparations. Putative antimicrobial agent-producing strains, previously isolated and preidentified as Bacillus spp. were profiled by repetitive element sequence-based polymerase chain reaction (rep-PCR) and 16s rRNA sequencing identified the strains as Bacillus tequilensis ST1962CD with 99.47% identity confidence and as Bacillus subtilis subsp. stercoris ST2056CD with 98.45% identity confidence. Both the selected Bacillus strains were evaluated via biomolecular and physiological approaches related to their safety and virulence, beneficial properties, enzyme production profile, and presence of corresponding genes for the production of antimicrobials and virulence. Both strains were confirmed to harbor srfa and sbo genes and be free of hemolysin binding component (B) and two lytic components (L1 and L2) [BL] and nonhemolytic enterotoxin-associated genes. Produced antimicrobial agents by strains ST1962CD and ST2056CD were partially purified through the combination of ammonium sulfate precipitation and hydrophobic-based chromatography on SepPakC18 and evaluated regarding their cytotoxicity. The dynamics of bacterial growth, pH change, accumulation of produced antimicrobials, and the mode of action were evaluated. Obtained results were pointing to the potential application of safe B. tequilensis ST1962CD and B. subtilis subsp. stercoris ST2056CD strains as functional beneficial microbial cultures that are putative producers of surfactin and/or subtilosin, as potent antimicrobials, for the treatment of some staphylococcal-associated infections. Expressed antimicrobials were shown to be not cytotoxic, and appropriate biotechnological approaches need to be developed for cost-effective production, isolation, and purification of expressed antimicrobials by studied strains.
Collapse
Affiliation(s)
- Jeemin Ye
- ProBacLab, Advanced Convergence, Handong Global University, Pohang, Republic of Korea
| | - Joanna Ivy Irorita Fugaban
- ProBacLab, Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Clarizza May Dioso
- Institute of Biological Sciences, University of the Philippines Los Baños, Laguna, Philippines
| | | | - Gee-Hyeun Choi
- ProBacLab, Advanced Convergence, Handong Global University, Pohang, Republic of Korea
| | - Bobae Kim
- HEM Pharma, Pohang, Republic of Korea
| | | | - Svetoslav Dimitrov Todorov
- ProBacLab, Advanced Convergence, Handong Global University, Pohang, Republic of Korea
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), Departamento de Alimentos e Nutrição Experimental, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Lazar V, Oprea E, Ditu LM. Resistance, Tolerance, Virulence and Bacterial Pathogen Fitness-Current State and Envisioned Solutions for the Near Future. Pathogens 2023; 12:pathogens12050746. [PMID: 37242416 DOI: 10.3390/pathogens12050746] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The current antibiotic crisis and the global phenomena of bacterial resistance, inherited and non-inherited, and tolerance-associated with biofilm formation-are prompting dire predictions of a post-antibiotic era in the near future. These predictions refer to increases in morbidity and mortality rates as a consequence of infections with multidrug-resistant or pandrug-resistant microbial strains. In this context, we aimed to highlight the current status of the antibiotic resistance phenomenon and the significance of bacterial virulence properties/fitness for human health and to review the main strategies alternative or complementary to antibiotic therapy, some of them being already clinically applied or in clinical trials, others only foreseen and in the research phase.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| | - Eliza Oprea
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| | - Lia-Mara Ditu
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Street, 060101 Bucharest, Romania
| |
Collapse
|
37
|
Soleimanifar H, Mahmoodzadeh Hosseini H, Samavarchi Tehrani S, Mirhosseini SA. The Anti-Adhesion Effect of Nisin as a Robust Lantibiotic on the Colorectal Cancer Cells. Adv Biomed Res 2023; 12:113. [PMID: 37288013 PMCID: PMC10241620 DOI: 10.4103/abr.abr_267_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 06/09/2023] Open
Abstract
Background Bacteriocins are a type of antimicrobial peptide that are produced by probiotics. They have been studied as possible therapeutic drugs and have been used to suppress bacterial development in foods. Nisin is a potent bacteriocin having the anti-microbial and anti-cancer characteristics produced by Lactococcus lactis. The aim of the present paper is to evaluate the influence of Nisin on cell adhesion and its two related genes, mmp-2 and mmp-9, in the colorectal cancer cell line. Materials and Methods For this purpose, HT-29 cells were treated with various concentrations of Nisin and the cell cytotoxicity, cell adhesion, and gene expression were evaluated using the MTT assay, cell adhesion assay, and real-time PCR. Results Our findings showed that 32 to 1024 μg/ml of Nisin resulted in a significant reduction in cell viability (P < 0.05). Furthermore, 128 and 256 μg/ml of Nisin significantly reduced the cell adhesion, and mmp-2 and mmp-9 gene expressions (P < 0.05). Conclusion Our findings suggested that Nisin could prevent metastasis and cancer progression.
Collapse
Affiliation(s)
- Hesam Soleimanifar
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamideh Mahmoodzadeh Hosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirhosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Zommiti M, Ferchichi M, Feuilloley MGJ. "Beneficial Microbes: Food, Mood and Beyond"-Editorial and the Perspectives of Research. Microorganisms 2023; 11:microorganisms11041014. [PMID: 37110437 PMCID: PMC10145506 DOI: 10.3390/microorganisms11041014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Amongst the list of beneficial microbes, lactic acid bacteria (LAB), Bifidobacterium sp [...].
Collapse
Affiliation(s)
- Mohamed Zommiti
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
- Normandie University, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Mounir Ferchichi
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia
| | - Marc G J Feuilloley
- Research Unit Bacterial Communication and Anti-infectious Strategies (CBSA, UR4312), University of Rouen Normandie, 27000 Evreux, France
| |
Collapse
|
39
|
Thoda C, Touraki M. Immunomodulatory Properties of Probiotics and Their Derived Bioactive Compounds. APPLIED SCIENCES 2023; 13:4726. [DOI: 10.3390/app13084726] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Immune system modulation is an intriguing part of scientific research. It is well established that the immune system plays a crucial role in orchestrating cellular and molecular key mediators, thus establishing a powerful defense barrier against infectious pathogens. Gut microbiota represent a complex community of approximately a hundred trillion microorganisms that live in the mammalian gastrointestinal (GI) tract, contributing to the maintenance of gut homeostasis via regulation of the innate and adaptive immune responses. However, impairment in the crosstalk between intestinal immunity and gut microbiota may reflect on detrimental health issues. In this context, many studies have indicated that probiotics and their bioactive compounds, such as bacteriocins and short chain fatty acids (SCFAs), display distinct immunomodulatory properties through which they suppress inflammation and enhance the restoration of microbial diversity in pathological states. This review highlights the fundamental features of probiotics, bacteriocins, and SCFAs, which make them ideal therapeutic agents for the amelioration of inflammatory and autoimmune diseases. It also describes their underlying mechanisms on gut microbiota modulation and emphasizes how they influence the function of immune cells involved in regulating gut homeostasis. Finally, it discusses the future perspectives and challenges of their administration to individuals.
Collapse
Affiliation(s)
- Christina Thoda
- Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece
| | - Maria Touraki
- Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece
| |
Collapse
|
40
|
Sadeghi A, Katouzian I, Ebrahimi M, Assadpour E, Tan C, Jafari SM. Bacteriocin-like inhibitory substances as green bio-preservatives; nanoliposomal encapsulation and evaluation of their in vitro/in situ anti-Listerial activity. Food Control 2023. [DOI: 10.1016/j.foodcont.2023.109725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
41
|
Kumari R, Singhvi N, Sharma P, Choudhury C, Shakya R. Virtual screening of gut microbiome bacteriocins as potential inhibitors of stearoyl-CoA desaturase 1 to regulate adipocyte differentiation and thermogenesis to combat obesity. J Biomol Struct Dyn 2023; 41:12632-12642. [PMID: 36644882 DOI: 10.1080/07391102.2023.2167121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
The gut bacterial strains and their metabolites have been shown to play a significant role in obesity, but the molecular mechanisms underlying this association are largely unresolved. Obesity is a multifactorial problem and is controlled by various mechanisms and pathways to produce and store fat cells. Bacteriocins are secondary metabolites produced by gut bacteria to defend themselves against their competitors. Recently, they have gained great attention due to their role in metabolic disorders, including obesity. Stearoyl-CoA desaturase 1 (SCD1) is a key enzyme involved in the differentiation of adipocytes. The aim of this study is to show the regulation of SCD1 by bacteriocins and thus their importance in obesity control. We screened the human gut bacteriome for the presence of bacteriocins, predicted their structures, and showed their inhibitory role by molecular docking with SCD1. Further, to confirm the docking results, MDS of six top scoring SCD1-bacteriocin complexes were carried out for 100 ns. These six bacteriocins namely, Plantaricin S-beta, Carnolysin, Lactococcin B, Bacteriocin Iic, Plantaricin N, and Thermophilin A, with strong binding affinities, are primarily produced by bacterial strains from the Lactobacillaeacea family. These findings can be the basis of further experiments for enhanced understanding of the underlying mechanisms for obesity control, specifically bacteriocins driven regulation of the SCD1 enzyme. In addition, a consortium of bacterial strains producing these bacteriocins can be developed and used as probiotics for the amelioration of obesity and other metabolic complications.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rekha Kumari
- Molecular Microbiology and Bioinformatics Laboratory, Department of Zoology, Miranda House, University of Delhi, Delhi, India
| | - Nirjara Singhvi
- School of Allied Sciences, Dev Bhoomi Uttarakhand University, Dehradun, Uttarakhand, India
| | - Poonam Sharma
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Chinmayee Choudhury
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Rashmi Shakya
- Department of Botany, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|
42
|
Wang Y, Gu Z, Zhang S, Li P. Complete Genome Sequencing Revealed the Potential Application of a Novel Weizmannia coagulans PL-W Production with Promising Bacteriocins in Food Preservative. Foods 2023; 12:216. [PMID: 36613432 PMCID: PMC9818457 DOI: 10.3390/foods12010216] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/01/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
Weizmannia coagulans is an important potential probiotic with dual characteristics of Bacillus and Lactobacillus. This study describes a novel Weizmannia coagulans PL-W with excellent antibacterial activity isolated from Mongolian traditional cheese, in which safety and probiotic potential were evaluated by complete genome sequencing. The crude bacteriocins of W. coagulans PL-W showed antibacterial activity against various foodborne pathogens, including Listeria monocytogenes CMCC 54,004, Bacillus cereus ATCC 14,579, and Staphylococcus aureus ATCC 25,923. Moreover, the crude bacteriocins have outstanding stability against pH, temperature, surfactants, and are sensitive to protease. The complete genome sequencing revealed W. coagulans PL-W consists of 3,666,052-base pair (bp) circular chromosomes with a GC content of 46.24% and 3485 protein-coding genes. It contains 84 tRNA, 10 23S rRNA, 10 16S rRNA, and 10 5S rRNA. In addition, no risk-related genes such as acquired antibiotic resistance genes, virulence, and pathogenic factors were identified, demonstrating that W. coagulans PL-W is safe to use. Furthermore, the presence of gene clusters involved in bacteriocin synthesis, adhesion-related genes, and genes contributing to acid and bile tolerance indicate that W. coagulans PL-W is a potential candidate probiotic. Thus, antimicrobial activity and genome characterization of W. coagulans PL-W demonstrate that it has extensive potential applications as a food protective culture.
Collapse
Affiliation(s)
| | | | | | - Pinglan Li
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| |
Collapse
|
43
|
The Discovery of Oropharyngeal Microbiota with Inhibitory Activity against Pathogenic Neisseria gonorrhoeae and Neisseria meningitidis: An In Vitro Study of Clinical Isolates. Microorganisms 2022; 10:microorganisms10122497. [PMID: 36557750 PMCID: PMC9787740 DOI: 10.3390/microorganisms10122497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
With increasing incidence of pathogenic Neisseria infections coupled with emerging resistance to antimicrobials, alternative approaches to limit the spread are sought. We investigated the inhibitory effect of oropharyngeal microbiota on the growth of N. gonorrhoeae and N. meningitidis and the impact of the essential oil-based mouthwash Listerine Cool Mint® (Listerine). Oropharyngeal swabs from 64 men who have sex with men (n = 118) from a previous study (PReGo study) were analysed (ClinicalTrials.gov, NCT03881007). These included 64 baseline and 54 samples following three months of daily use of Listerine. Inhibition was confirmed by agar overlay assay, and inhibitory bacteria isolated using replica plating and identified using MALDI-TOF. The number of inhibitory isolates were compared before and after Listerine use. Thirty-one pharyngeal samples (26%) showed inhibitory activity against N. gonorrhoeae and/or N. meningitidis, and 62 inhibitory isolates were characterised. Fourteen species belonging to the genera Streptococci and Rothia were identified. More inhibitory isolates were observed following Listerine use compared to baseline, although this effect was not statistically significant (p = 0.073). This study isolated and identified inhibitory bacteria against pathogenic Neisseria spp. and established that daily Listerine use did not decrease their prevalence. These findings could provide a new approach for the prevention and treatment of pharyngeal Neisseria infections.
Collapse
|
44
|
Zhu H, Guo L, Yu D, Du X. New insights into immunomodulatory properties of lactic acid bacteria fermented herbal medicines. Front Microbiol 2022; 13:1073922. [DOI: 10.3389/fmicb.2022.1073922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
The COVID-19 pandemic has brought more attention to the immune system, the body’s defense against infectious diseases. The immunomodulatory ability of traditional herbal medicine has been confirmed through clinical trial research, and has obvious advantages over prescription drugs due to its high number of potential targets and low toxicity. The active compounds of herbal drugs primarily include polysaccharides, saponins, flavonoids, and phenolics and can be modified to produce new active compounds after lactic acid bacteria (LAB) fermentation. LAB, primary source of probiotics, can produce additional immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. Moreover, several compounds from herbal medicines can promote the growth and production of LAB-based immune active metabolites. Thus, LAB-mediated fermentation of herbal medicines has become a novel strategy for regulating human immune responses. The current review discusses the immunomodulatory properties and active compounds of LAB fermented herbal drugs, the interaction between LAB and herbal medicines, and changes in immunoregulatory components that occur during fermentation. This study also discusses the mechanisms by which LAB-fermented herbal medicines regulate the immune response, including activation of the innate or adaptive immune system and the maintenance of intestinal immune homeostasis.
Collapse
|
45
|
Teng K, Huang F, Liu Y, Wang Y, Xia T, Yun F, Zhong J. Food and gut originated bacteriocins involved in gut microbe-host interactions. Crit Rev Microbiol 2022:1-13. [PMID: 35713699 DOI: 10.1080/1040841x.2022.2082860] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The gut microbes interact with each other as well as host, influencing human health and some diseases. Many gut commensals and food originated bacteria produce bacteriocins which can inhibit pathogens and modulate gut microbiota. Bacteriocins have comparable narrow antimicrobial spectrum and are attractive potentials for precision therapy of gut disorders. In this review, the bacteriocins from food and gut microbiomes and their involvement in the interaction between producers and gut ecosystem, along with their characteristics, types, biosynthesis, and functions are described and discussed. Bacteriocins are produced by many intestinal commensals and food microbes among which lactic acid bacteria (many are probiotics) has been paid more attention. Bacteriocin production has been generally regarded as a probiotic trait. They give a competitive advantage to bacteria, enabling their colonization in human gut, and mediating the interaction between the producers and host ecosystem. They fight against unwanted bacteria and pathogens without significant impact on the composition of commensal microbiota. Bacteriocins assist the producers to survive and colonize in the gut microbial populations. There is a great need to evaluate and utilize the potential of bacteriocins for improved therapeutic implications for intestinal health.
Collapse
Affiliation(s)
- Kunling Teng
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Fuqing Huang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yayong Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yudong Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Tianqi Xia
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Fangfei Yun
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jin Zhong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,School of Life Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
46
|
Bu Y, Liu Y, Liu Y, Wang S, Liu Q, Hao H, Yi H. Screening and Probiotic Potential Evaluation of Bacteriocin-Producing Lactiplantibacillus plantarum In Vitro. Foods 2022; 11:foods11111575. [PMID: 35681325 PMCID: PMC9180163 DOI: 10.3390/foods11111575] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Probiotics are gaining attention due to their functions of regulating the intestinal barrier and promoting human health. The production of bacteriocins is one of the important factors for probiotics to exert beneficial properties. This study aimed to screen bacteriocin-producing Lactiplantibacillus plantarum and evaluate the probiotic potential in vitro. It was found that L. plantarum Q7, L. plantarum F3-2 and L. plantarum YRL45 could produce bacteriocins and inhibit common intestinal pathogens. These three strains had probiotic potential with tolerance to the gastrointestinal environmental and colonization in the gut, and exhibited various degrees of anti-inflammatory activity and tight junction function in the intestinal barrier. Particularly, L. plantarum YRL45 could significantly (p < 0.05) reduce the increase in nitric oxide (NO), prostaglandin E2 (PGE2), necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) induced by lipopolysaccharide (LPS), thereby easing inflammatory response. L. plantarum F3-2 could remarkably (p < 0.05) up-regulate the expression levels of ZO-1, Occludin and Claudin-1 in intestinal epithelial injured cells, which was conducive to protecting the intestinal barrier. These findings provided fundamental information about the probiotic properties of bacteriocin-producing L. plantarum, which suggested that L. plantarum Q7, L. plantarum F3-2 and L. plantarum YRL45 had the potential to be used as novel probiotic strains.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huaxi Yi
- Correspondence: ; Tel.: +86-0532-13792497030
| |
Collapse
|
47
|
Tiwari SK. Bacteriocin-Producing Probiotic Lactic Acid Bacteria in Controlling Dysbiosis of the Gut Microbiota. Front Cell Infect Microbiol 2022; 12:851140. [PMID: 35651753 PMCID: PMC9149203 DOI: 10.3389/fcimb.2022.851140] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/16/2022] [Indexed: 11/20/2022] Open
Abstract
Several strains of lactic acid bacteria are potent probiotics and can cure a variety of diseases using different modes of actions. These bacteria produce antimicrobial peptides, bacteriocins, which inhibit or kill generally closely related bacterial strains and other pathogenic bacteria such as Listeria, Clostridium, and Salmonella. Bacteriocins are cationic peptides that kill the target cells by pore formation and the dissipation of cytosolic contents, leading to cell death. Bacteriocins are also known to modulate native microbiota and host immunity, affecting several health-promoting functions of the host. In this review, we have discussed the ability of bacteriocin-producing probiotic lactic acid bacteria in the modulation of gut microbiota correcting dysbiosis and treatment/maintenance of a few important human disorders such as chronic infections, inflammatory bowel diseases, obesity, and cancer.
Collapse
|
48
|
Ma K, Chen W, Yan SQ, Liu ZZ, Lin XQ, Zhang JB, Gao Y, Wang T, Zhang JG, Yang YJ. Purification, Characterization, Mode of Action, and Application of Jileicin, a Novel Antimicrobial from Paenibacillus jilinensis YPG26. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5570-5578. [PMID: 35499918 PMCID: PMC9104118 DOI: 10.1021/acs.jafc.2c01458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Antimicrobial compounds from the commensal gut microbiota have gained much attention due to their multifunctionality in maintaining good health in the host and killing multidrug-resistant bacteria. Our previous study showed that Paenibacillus jilinensis YPG26 isolated from chicken intestine can antagonize multiple pathogens. Herein, we characterized a bacteriocin-like inhibitory substance, jileicin, purified from P. jilinensis YPG26. Mass spectrometry analysis revealed that jileicin was a protein consisting of 211 amino acids, which showed 88.98% identity to the SIMPL domain-containing protein. The jileicin showed a relatively broad-spectrum antibacterial ability, especially against enterococci. Additionally, the jileicin exhibited good stability after various treatments, no detectable resistance, no significant cytotoxicity, and very low levels of hemolytic activity. The mode of action against Enterococcus faecium demonstrated that jileicin could destroy cell membrane integrity, increase cell membrane permeability, and eventually lead to cell death. Furthermore, jileicin was efficient in controlling the growth of E. faecium in milk. In conclusion, jileicin, as a newly identified antibacterial agent, is expected to be a promising candidate for application in the food, pharmaceutical, and biomedical industries.
Collapse
|
49
|
Domínguez Rubio AP, D’Antoni CL, Piuri M, Pérez OE. Probiotics, Their Extracellular Vesicles and Infectious Diseases. Front Microbiol 2022; 13:864720. [PMID: 35432276 PMCID: PMC9006447 DOI: 10.3389/fmicb.2022.864720] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics have been shown to be effective against infectious diseases in clinical trials, with either intestinal or extraintestinal health benefits. Even though probiotic effects are strain-specific, some "widespread effects" include: pathogen inhibition, enhancement of barrier integrity and regulation of immune responses. The mechanisms involved in the health benefits of probiotics are not completely understood, but these effects can be mediated, at least in part, by probiotic-derived extracellular vesicles (EVs). However, to date, there are no clinical trials examining probiotic-derived EVs health benefits against infectious diseases. There is still a long way to go to bridge the gap between basic research and clinical practice. This review attempts to summarize the current knowledge about EVs released by probiotic bacteria to understand their possible role in the prevention and/or treatment of infectious diseases. A better understanding of the mechanisms whereby EVs package their cargo and the process involved in communication with host cells (inter-kingdom communication), would allow further advances in this field. In addition, we comment on the potential use and missing knowledge of EVs as therapeutic agents (postbiotics) against infectious diseases. Future research on probiotic-derived EVs is needed to open new avenues for the encapsulation of bioactives inside EVs from GRAS (Generally Regarded as Safe) bacteria. This could be a scientific novelty with applications in functional foods and pharmaceutical industries.
Collapse
Affiliation(s)
- A. Paula Domínguez Rubio
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Cecilia L. D’Antoni
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Piuri
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Oscar E. Pérez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
50
|
Bozzetti V, Senger S. Organoid technologies for the study of intestinal microbiota–host interactions. Trends Mol Med 2022; 28:290-303. [PMID: 35232671 PMCID: PMC8957533 DOI: 10.1016/j.molmed.2022.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
Postbiotics have recently emerged as critical effectors of the activity of probiotics and, because of their safety profile, they are considered potential therapeutics for the treatment of fragile patients. Here, we present recent studies on probiotics and postbiotics in the context of novel discovery tools, such as organoids and organoid-based platforms, and nontransformed preclinical models, that can be generated from intestinal stem cells. The implementation of organoid-related techniques is the next gold standard for unraveling the effect of microbial communities on homeostasis, inflammation, idiopathic diseases, and cancer in the gut. We also summarize recent studies on biotics in organoid-based models and offer our perspective on future directions.
Collapse
|