1
|
Zhou C, Chang X, Zou Y, Zhao F, Zhou G, Ye K. The mechanism of Enterococcus faecium on the virulence of Listeria monocytogenes during the storage of fermented sausages by whole genome analysis. Int J Food Microbiol 2024; 422:110826. [PMID: 39024730 DOI: 10.1016/j.ijfoodmicro.2024.110826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/17/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
This study investigated the safety characteristics and potential probiotic properties of Enterococcus faecium by using whole genome analysis, and then explored the effect of this strain on the virulence of Listeria monocytogenes in vitro and during the storage of fermented sausages. Results showed that E. faecium B1 presented enterocin A, B, and P, enterolysin A, and UviB, and the exotoxin related genes and exoenzyme related genes were not detected in the genome of E. faecium B1. However, the adherence genes including acm and scm were present in this strain, which also positively correlated with characteristics related to probiotic potential. In addition, E. faecium could adapt to the condition of fermented sausages, and decrease the survival of L. monocytogenes in vitro and in vivo. The expression of the virulence genes (prfA, hly, inlA, and inlB) and sigB-related genes (prli42, rsbT, rsbU, rsbV, rsbW, and sigB) were all inhibited by E. faecium B1 to different extents during the storage of fermented sausages at 4 °C. Moreover, compared with the E. faecium B1 group, the expression level of entA, entB, and entP genes of E. faecium B1 in the co-culture of fermented sausages was increased during the storage, which may be the inhibition mechanism of E. faecium B1 on L. monocytogenes. These results demonstrated that E. faecium B1 could potentially be used as bio-protection to control L. monocytogenes in meat products.
Collapse
Affiliation(s)
- Cong Zhou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China
| | - Xiaochen Chang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China
| | - Yafang Zou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China
| | - Fanwen Zhao
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China
| | - Guanghong Zhou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China
| | - Keping Ye
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, 210095 Nanjing, Jiangsu, PR China.
| |
Collapse
|
2
|
Popović N, Veljović K, Radojević D, Brdarić E, Stevanović D, Živković M, Kojić M. Insight into the Probiogenomic Potential of Enterococcus faecium BGPAS1-3 and Application of a Potent Thermostable Bacteriocin. Foods 2024; 13:2637. [PMID: 39200563 PMCID: PMC11353538 DOI: 10.3390/foods13162637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/02/2024] Open
Abstract
This study aimed to investigate the probiogenomic features of artisanal bacteriocin-producing Enterococcus faecium BGPAS1-3 and the use of the improved pMALc5HisEk expression vector for overexpressing class II bacteriocins and the application of purified bacteriocin 31 in a milk model as a preservative against L. monocytogenes. The BGPAS1-3 strain was isolated from traditional fresh soft cheese manufactured in households on a small scale in rural locations surrounding Pale Mountain City in Bosnia and Herzegovina. The whole-genome sequencing approach and bioinformatics analyses revealed that the strain BGPAS1-3 was non-pathogenic to humans. The presence of bacteriocin operons suggested the ability of the isolate to suppress the growth of pathogens. Coding regions for three maturated bacteriocins (bacteriocin 31, bacteriocin 32, and enterocin P) produced by BGPAS1-3 were amplified and expressed in Escherichia coli ER2523 using the pMALc5HisEk system. All three bacteriocins were successfully overexpressed and purified after enterokinase cleavage but showed different antimicrobial activity. Bacteriocin 31 showed significantly stronger antimicrobial activity compared with bacteriocin 32. It was the only one that proved to be suitable for use as a food preservative against L. monocytogenes in a milk model.
Collapse
Affiliation(s)
- Nikola Popović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Katarina Veljović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Dušan Radojević
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Emilija Brdarić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Dušan Stevanović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Milica Živković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
| | - Milan Kojić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (D.R.); (E.B.); (D.S.); (M.Ž.)
- Department of Research and Development, Institute of Virology, Vaccines, and Sera “Torlak”, Vojvode Stepe 458, 11152 Belgrade, Serbia
| |
Collapse
|
3
|
Banić M, Butorac K, Čuljak N, Butorac A, Novak J, Pavunc AL, Rušanac A, Stanečić Ž, Lovrić M, Šušković J, Kos B. An Integrated Comprehensive Peptidomics and In Silico Analysis of Bioactive Peptide-Rich Milk Fermented by Three Autochthonous Cocci Strains. Int J Mol Sci 2024; 25:2431. [PMID: 38397111 PMCID: PMC10888711 DOI: 10.3390/ijms25042431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Bioactive peptides (BPs) are molecules of paramount importance with great potential for the development of functional foods, nutraceuticals or therapeutics for the prevention or treatment of various diseases. A functional BP-rich dairy product was produced by lyophilisation of bovine milk fermented by the autochthonous strains Lactococcus lactis subsp. lactis ZGBP5-51, Enterococcus faecium ZGBP5-52 and Enterococcus faecalis ZGBP5-53 isolated from the same artisanal fresh cheese. The efficiency of the proteolytic system of the implemented strains in the production of BPs was confirmed by a combined high-throughput mass spectrometry (MS)-based peptidome profiling and an in silico approach. First, peptides released by microbial fermentation were identified via a non-targeted peptide analysis (NTA) comprising reversed-phase nano-liquid chromatography (RP nano-LC) coupled with matrix-assisted laser desorption/ionisation-time-of-flight/time-of-flight (MALDI-TOF/TOF) MS, and then quantified by targeted peptide analysis (TA) involving RP ultrahigh-performance LC (RP-UHPLC) coupled with triple-quadrupole MS (QQQ-MS). A combined database and literature search revealed that 10 of the 25 peptides identified in this work have bioactive properties described in the literature. Finally, by combining the output of MS-based peptidome profiling with in silico bioactivity prediction tools, three peptides (75QFLPYPYYAKPA86, 40VAPFPEVFGK49, 117ARHPHPHLSF126), whose bioactive properties have not been previously reported in the literature, were identified as potential BP candidates.
Collapse
Affiliation(s)
- Martina Banić
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Katarina Butorac
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Nina Čuljak
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Ana Butorac
- BICRO Biocentre Ltd., Borongajska cesta 83H, 10000 Zagreb, Croatia; (A.B.); (Ž.S.); (M.L.)
| | - Jasna Novak
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Andreja Leboš Pavunc
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Anamarija Rušanac
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Željka Stanečić
- BICRO Biocentre Ltd., Borongajska cesta 83H, 10000 Zagreb, Croatia; (A.B.); (Ž.S.); (M.L.)
| | - Marija Lovrić
- BICRO Biocentre Ltd., Borongajska cesta 83H, 10000 Zagreb, Croatia; (A.B.); (Ž.S.); (M.L.)
| | - Jagoda Šušković
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| | - Blaženka Kos
- Laboratory for Antibiotic, Enzyme, Probiotic and Starter Culture Technologies, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (M.B.); (K.B.); (N.Č.); (J.N.); (A.L.P.); (A.R.); (J.Š.)
| |
Collapse
|
4
|
Landete JM, Montiel R, Rodríguez-Mínguez E, Arqués JL. Enterocins Produced by Enterococci Isolated from Breast-Fed Infants: Antilisterial Potential. CHILDREN (BASEL, SWITZERLAND) 2024; 11:261. [PMID: 38397373 PMCID: PMC10887673 DOI: 10.3390/children11020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024]
Abstract
Enterocins are bacteriocins synthesized by Enterococcus strains that show an interesting antimicrobial effectiveness against foodborne pathogens such as Listeria monocytogenes. The objectives of this study were to identify and analyze the expression of enterocin genes of Enterococcus isolated from breast-fed infants and evaluate their ability to inhibit three human isolates of virulent L. monocytogenes, as well as some probiotic bacteria. The susceptibility of the strains of L. monocytogenes to fifteen antibiotics was tested, detecting their resistance to cefoxitin (constitutively resistant), oxacillin, and clindamycin. The production of enterocins A, B, and P was observed in Enterococcus faecium isolates, while enterocin AS-48 was detected in an Enterococcus faecalis isolate. AS-48 showed antilisterial activity by itself, while the joint action of enterocins A and B or B and P was necessary for inhibiting L. monocytogenes, demonstrating the synergistic effect of those combinations. The presence of multiple enterocin genes does not assure the inhibition of L. monocytogenes strains. However, the expression of multiple enterocin genes showed a good correlation with the inhibition capacity of these strains. Furthermore, the potential beneficial strains of lactobacilli and bifidobacteria examined were not inhibited by any of the enterocins produced individually or in combination, with the exception of Bifidobacterium longum BB536, which was inhibited by enterocin AS-48 and the joint production of enterocins A and B or B and P. The enterocins studied here could be candidates for developing alternative treatments against antibiotic-resistant bacterial infections. Moreover, these selected enterocin-producing E. faecium strains isolated from breast-fed infants could be used as probiotic strains due to their antilisterial effect, as well as the absence of virulence factors.
Collapse
Affiliation(s)
| | | | | | - Juan L. Arqués
- Department of Food Technology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, INIA-CSIC, Carretera de La Coruña Km 7, 28040 Madrid, Spain; (J.M.L.); (R.M.); (E.R.-M.)
| |
Collapse
|
5
|
Popović N, Stevanović D, Radojević D, Veljović K, Đokić J, Golić N, Terzić-Vidojević A. Insight into the Postbiotic Potential of the Autochthonous Bacteriocin-Producing Enterococcus faecium BGZLM1-5 in the Reduction in the Abundance of Listeria monocytogenes ATCC19111 in a Milk Model. Microorganisms 2023; 11:2844. [PMID: 38137988 PMCID: PMC10745621 DOI: 10.3390/microorganisms11122844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
This study aimed to explore the probiogenomic characteristics of artisanal bacteriocin-producing Enterococcus faecium BGZLM1-5 and its potential application in reducing Listeria monocytogenes in a milk model. The BGZLM1-5 strain was isolated from raw cow's milk from households in the Zlatar Mountain region. The whole genome sequencing approach and bioinformatics analyses reveal that the strain BGZLM1-5 is non-pathogenic to humans. Bacteriocin-containing supernatant was thermally stable and antimicrobial activity retained 75% of the initial activity compared with that of the control after treatment at 90 °C for 30 min. Antimicrobial activity maintained relative stability at pH 3-11 and retained 62.5% of the initial activity compared with that of the control after treatment at pH 1, 2, and 12. The highest activity of the partially purified bacteriocin was obtained after precipitation at 40% saturation with ammonium sulfate and further purification by mixing with chloroform. Applying 3% and 5% (v/v) of the bacteriocin-containing supernatant and 0.5% (v/v) of the partially purified bacteriocin decreased the viable number of L. monocytogenes ATCC19111 after three days of milk storage by 23.5%, 63.5%, and 58.9%, respectively.
Collapse
Affiliation(s)
- Nikola Popović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (D.S.); (D.R.); (K.V.); (J.Đ.); (N.G.); (A.T.-V.)
| | | | | | | | | | | | | |
Collapse
|
6
|
Qi Y, Yu L, Tian F, Zhao J, Zhai Q. In vitro models to study human gut-microbiota interactions: Applications, advances, and limitations. Microbiol Res 2023; 270:127336. [PMID: 36871313 DOI: 10.1016/j.micres.2023.127336] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
In vitro models of the human gut help compensate for the limitations of animal models in studying the human gut-microbiota interaction and are indispensable in the clarification the mechanism of microbial action or in the high-throughput screening and functional evaluation of probiotics. The development of these models constitutes a rapidly developing field of research. From 2D1 to 3D2 and from simple to complex, several in vitro cell and tissue models have been developed and continuously improved. In this review, we categorized and summarized these models and described their development, applications, advances, and limitations by using specific examples. We also highlighted the best ways to select an appropriate in vitro model, and we also discussed which variables to consider when imitating microbial and human gut epithelial interactions.
Collapse
Affiliation(s)
- Yuli Qi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
7
|
Guo L, Yin X, Liu Q. Fecal microbiota transplantation reduces mouse mortality from Listeria monocytogenes infection. Microb Pathog 2023; 178:106036. [PMID: 36813004 DOI: 10.1016/j.micpath.2023.106036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023]
Abstract
Listeria monocytogenes (Lm) is a food bacterium with strong pathogenicity which causes infections via the gastrointestinal tract. Mechanisms by which gut microbiota (GM) resist microbial infections have received little attention. Eight-week-old mice were orally inoculated with wild-type Lm EGD-e and fecal microbiota transplantation (FMT) employed. GM richness and diversity of infected mice changed rapidly within 24h. Firmicutes class decreased and Bacteroidetes, Tenericutes and Ruminococcaceae increased significantly. Coprococcus, Blautia and Eubacterium also increased on the 3rd day post-infection. Moreover, GM transplanted from healthy mice reduced mortality of infected mice by approximately 32%. FMT treatment decreased production of TNFα, IFN-γ, IL-1β and IL-6 relative to PBS treatment. In summary, FMT has potential as a treatment against Lm infection and may be used for bacterial resistance management. Further work is required to elucidate the key GM effector molecules.
Collapse
Affiliation(s)
- Liang Guo
- Zaozhuang University, Shandong, 277160, China; School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | | | - Qing Liu
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China.
| |
Collapse
|
8
|
Zhong Y, Wang T, Luo R, Liu J, Jin R, Peng X. Recent advances and potentiality of postbiotics in the food industry: Composition, inactivation methods, current applications in metabolic syndrome, and future trends. Crit Rev Food Sci Nutr 2022; 64:5768-5792. [PMID: 36537328 DOI: 10.1080/10408398.2022.2158174] [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: 12/24/2022]
Abstract
Postbiotics are defined as "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics have unique advantages over probiotics, such as stability, safety, and wide application. Although postbiotics are research hotspots, the research on them is still very limited. This review provides comprehensive information on the scope of postbiotics, the preparation methods of inanimate microorganisms, and the application and mechanisms of postbiotics in metabolic syndrome (MetS). Furthermore, the application trends of postbiotics in the food industry are reviewed. It was found that postbiotics mainly include inactivated microorganisms, microbial lysates, cell components, and metabolites. Thermal treatments are the main methods to prepare inanimate microorganisms as postbiotics, while non-thermal treatments, such as ionizing radiation, ultraviolet light, ultrasound, and supercritical CO2, show great potential in postbiotic preparation. Postbiotics could ameliorate MetS through multiple pathways including the modulation of gut microbiota, the enhancement of intestinal barrier, the regulation of inflammation and immunity, and the modulation of hormone homeostasis. Additionally, postbiotics have great potential in the food industry as functional food supplements, food quality improvers, and food preservatives. In addition, the SWOT analyses showed that the development of postbiotics in the food industry exists both opportunities and challenges.
Collapse
Affiliation(s)
- Yujie Zhong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Tao Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Ruilin Luo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiayu Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Ruyi Jin
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
9
|
Liu J, Shi L, Tuo X, Ma X, Hou X, Jiang S, Lv J, Cheng Y, Guo D, Han B. Preparation, characteristic and anti-inflammatory effect of selenium nanoparticle-enriched probiotic strain Enterococcus durans A8-1. J Trace Elem Med Biol 2022; 74:127056. [PMID: 35939922 DOI: 10.1016/j.jtemb.2022.127056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/01/2022] [Accepted: 08/02/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Elemental selenium, a new type of selenium supplement, can be biosynthesized via microorganisms. This study is to characterize a patent probiotic bacteria Enterococcus durans A8-1, capable of reducing selenite (Se6+ or Se4+) to elemental selenium (Se0) with the formation of Se nanoparticles (SeNPs). METHODS The selenium nanoparticles synthesized from A8-1 were characterized using scanning electron microscopy (SEM), transmission electron microscopy (TEM), energy dispersive spectroscopy (EDS), and X-ray photoelectron energy (XPS). The Caco2 cells were used to investigate the effects of Se-enriched A8-1 on the viability, membrane integrity, and the regulation of cellular inflammation through MTT and ELISA assays. The selenium-enriched metabolic function of A8-1 was analyzed by transcriptome sequencing. RESULTS E. durans A8-1 has the ability to synthesize intracellular SeNPs that are incubated with 60 mg/L sodium selenite for 18 h at 37 °C with 7 % inoculum under aerobic conditions. The selenium-enriched transformation rate increased to 43.46 %. After selenium enrichment, there were no significant morphological changes in E. durans A8-1 cells. The cells also exhibited no cytotoxicity when incubated with Caco-2 cells, and increased cellular proliferation. Furthermore, Se-enriched A8-1 cells antagonize the adhesion of S. typhimurium ATCC14028 onto the surface of Caco-2 cells protecting cell membrane integrity and was assessed by measuring LDH and AKP activities (P <0.001, P <0.001). Moreover, Se-enriched A8-1 could protect Caco-2 cells from inflammation induced by lipopolysaccharide and help the cells alleviate the inflammation through the reduced expression of cytokine IL-8 (P = 0.0012, P <0.001) and TNF-α (P <0.001, P <0.001). Based on transcriptome sequencing in Se-enriched E. durans A8-1 cells, there were 485 up-regulated genes and 322 down-regulated genes (Padj < 0.05). There were 19 predicted up-regulated genes that are highly related to the potential selenium metabolism pathway, which focuses on the transportation of Na2SeO3 by membrane proteins, and gradually reduces Na2SeO3 to elemental selenium aggregates that are deposited onto the membrane surface via the intracellular redox response. CONCLUSION E. durans A8-1 could convert extracellular selenite into intracellular biological SeNPs via redox pathway with strong selenium-rich metabolism, and its biological SeNPs have anti-inflammatory properties, which have the potential for the development of composite selenium nanomaterials and can be further studied for the function of SeNPs with potential applications.
Collapse
Affiliation(s)
- Jin Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lu Shi
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaohong Tuo
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xinxin Ma
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xinyao Hou
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sijin Jiang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jia Lv
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yue Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China
| | - Dagang Guo
- School of Material Science and Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Bei Han
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China.
| |
Collapse
|
10
|
Thorakkattu P, Khanashyam AC, Shah K, Babu KS, Mundanat AS, Deliephan A, Deokar GS, Santivarangkna C, Nirmal NP. Postbiotics: Current Trends in Food and Pharmaceutical Industry. Foods 2022; 11:3094. [PMID: 36230169 PMCID: PMC9564201 DOI: 10.3390/foods11193094] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Postbiotics are non-viable bacterial products or metabolic byproducts produced by probiotic microorganisms that have biologic activity in the host. Postbiotics are functional bioactive compounds, generated in a matrix during anaerobic fermentation of organic nutrients like prebiotics, for the generation of energy in the form of adenosine triphosphate. The byproducts of this metabolic sequence are called postbiotics, these are low molecular weight soluble compounds either secreted by live microflora or released after microbial cell lysis. A few examples of widely studied postbiotics are short-chain fatty acids, microbial cell fragments, extracellular polysaccharides, cell lysates, teichoic acid, vitamins, etc. Presently, prebiotics and probiotics are the products on the market; however, postbiotics are also gaining a great deal of attention. The numerous health advantages of postbiotic components may soon lead to an increase in consumer demand for postbiotic supplements. The most recent research aspects of postbiotics in the food and pharmaceutical industries are included in this review. The review encompasses a brief introduction, classification, production technologies, characterization, biological activities, and potential applications of postbiotics.
Collapse
Affiliation(s)
- Priyamvada Thorakkattu
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS 66506, USA
| | | | - Kartik Shah
- Sargento Foods, 305 Pine Street, Elkhart Lake, WI 53020, USA
| | - Karthik Sajith Babu
- Department of Animal Sciences and Industry, Food Science Institute, Kansas State University, Manhattan, KS 66506, USA
| | - Anjaly Shanker Mundanat
- Department of Agriculture and Environmental Sciences, National Institute of Food Technology Entrepreneurship and Management (NIFTEM), Sonipat 131028, India
| | | | - Gitanjali S. Deokar
- Department of Quality Assurance, MET’s Institute of Pharmacy, Bhujbal Knowledge City, Nashik 422003, India
| | - Chalat Santivarangkna
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Nilesh Prakash Nirmal
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| |
Collapse
|
11
|
Wu J, Zhou B, Pang X, Song X, Gu Y, Xie R, Liu T, Xu X, Wang B, Cao H. Clostridium butyricum, a butyrate-producing potential probiotic, alleviates experimental colitis through epidermal growth factor receptor activation. Food Funct 2022; 13:7046-7061. [PMID: 35678197 DOI: 10.1039/d2fo00478j] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic inflammatory bowel disease. Modulation of gut microbiota with dietary and nutritional targets is a feasible strategy for the prevention and treatment of IBD. In this study, we focused on Clostridium butyricum Prazmowski (CB), a butyrate-producing potential probiotic. We found that CB feeding decreased the disease activity index, colon inflammation/injury score and cell apoptosis in an experimental colitis mouse model, as well as elevated the level of SCFAs in cecal feces. CB could also balance the inflammatory cytokines, protect tight junctions, and increase the number of goblet cells and MUC2 production in mice, accompanied by EGFR signaling activation triggered by heparin-binding epidermal growth factor (HB-EGF) and amphiregulin (AREG). From the perspective of mechanism, the CB supernatant (CBS) stimulated EGFR activation in colon epithelial cell lines in concentration-dependent and time-dependent manners. CBS reduced the damage of tight junctions induced by H2O2, and inhibition of EGFR could suppress the protective effect of CBS. In conclusion, CB could protect the gut barrier and alleviate experimental colitis through the transactivation of EGFR signaling in intestinal epithelial cells induced by ligands (HB-EGF and AREG). This study identified the potential efficacy of CB as a preventive strategy for IBD and showed the broad prospect of CB as a food supplement.
Collapse
Affiliation(s)
- Jingyi Wu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Bingqian Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Xiaoqi Pang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Xueli Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Runxiang Xie
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Xin Xu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
12
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Pérez-Cano FJ, Rodríguez-Lagunas MJ. Preventive Effect of a Postbiotic and Prebiotic Mixture in a Rat Model of Early Life Rotavirus Induced-Diarrhea. Nutrients 2022; 14:nu14061163. [PMID: 35334820 PMCID: PMC8954028 DOI: 10.3390/nu14061163] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
Rotavirus (RV) is the main cause of gastroenteritis in children. Prebiotics and, more recently, postbiotics are used for preventing and treating gastrointestinal infections. The aim of this study was to analyze the effects of a LactofidusTM, short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharides (lcFOS) mixture, and their combination on RV infection, in a rat model, for early life diarrhea. Fifteen litters of suckling rats were intragastrically administered daily with the vehicle, the prebiotic mixture, the postbiotic or the combination. The RV was inoculated on day 5 and then fecal samples were clinically evaluated daily. Viral shedding, intestinal permeability assay, in vitro blocking assay, immunoglobulin profiles, and anti-RV response were assessed at day 8 and 16 of life. Cecal microbiota composition, intestinal gene expression, and short chain fatty acids (SCFAs) were analyzed at day 16. The incidence and severity of diarrhea were significantly reduced by all the supplementations. Moreover, they showed blocking activity, changes in the immunoglobulin profiles, in gut microbiota, and in the intestinal gene expression. The prebiotic mixture reduced gut permeability and changed the SCFA profile, whereas the postbiotic enhanced the expression of Toll-like receptors (TLRs). The combination preserved most of the individual observed effects, and furthermore, complementary effects, such as an increase in white blood cells and lymphocytes recruitment, as well as upregulation of TLR7 and TLR9 gene expression.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (M.J.R.-L.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
13
|
Zhou C, Zou Y, Zhang Y, Teng S, Ye K. Involvement of CCN1 Protein and TLR2/4 Signaling Pathways in Intestinal Epithelial Cells Response to Listeria monocytogenes. Int J Mol Sci 2022; 23:ijms23052739. [PMID: 35269881 PMCID: PMC8911323 DOI: 10.3390/ijms23052739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 11/21/2022] Open
Abstract
CCN1 is well studied in terms of its functions in injury repair, cell adhesion survival and apoptosis, bacterial clearance and mediation of inflammation-related pathways, such as the TLR2/4 pathways. However, the role of CCN1 protein and its interaction with TLR2/4 pathways in intestinal epithelial cells was not elucidated after Listeria monocytogenes infection. The results of this study confirm that L. monocytogenes infection induced intestinal inflammation and increased the protein expression of CCN1, TLR2, TLR4 and p38, which followed a similar tendency in the expression of genes related to the TLR2/4 pathways. In addition, organoids infected by L. monocytogenes showed a significant increase in the expression of CCN1 and the activation of TLR2/4 pathways. Furthermore, pre-treatment with CCN1 protein to organoids infected by L. monocytogenes could increase the related genes of TLR2/4 pathways and up-regulate the expression of TNF, and increase the count of pathogens in organoids, which indicates that the interaction between the CCN1 protein and TLR2/4 signaling pathways in intestinal epithelial cells occurred after L. monocytogenes infection. This study will provide a novel insight of the role of CCN1 protein after L. monocytogenes infection in the intestine.
Collapse
|
14
|
Zhou Y, Shi L, Wang J, Yuan J, Liu J, Liu L, Da R, Cheng Y, Han B. Probiotic Potential Analysis and Safety Evaluation of Enterococcus durans A8-1 Isolated From a Healthy Chinese Infant. Front Microbiol 2022; 12:799173. [PMID: 34970251 PMCID: PMC8712863 DOI: 10.3389/fmicb.2021.799173] [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: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
To evaluate the probiotic characteristics and safety of Enterococcus durans isolate A8-1 from a fecal sample of a healthy Chinese infant, we determined the tolerance to low pH, survival in bile salts and NaCl, adhesion ability, biofilm formation, antimicrobial activity, toxin gene distribution, hemolysis, gelatinase activity, antibiotic resistance, and virulence to Galleria mellonella and interpreted the characters by genome resequencing. Phenotypically, E. durans A8-1 survived at pH 5.0 in 7.0% NaCl and 3% bile salt under aerobic and anaerobic condition. The bacterium had higher adhesion ability toward mucin, collagen, and Bovine Serum Albumin (BSA) in vitro and showed high hydrophobicity (79.2% in chloroform, 49.2% in xylene), auto-aggregation activity (51.7%), and could co-aggregate (66.2%) with Salmonella typhimurium. It had adhesion capability to intestinal epithelial Caco-2 cells (38.74%) with moderate biofilm production and antimicrobial activity against several Gram-positive pathogenic bacteria. A8-1 can antagonize the adhesion of S. typhimurium ATCC14028 on Caco-2 cells to protect the integrity of the cell membrane by detection of lactate dehydrogenase (LDH) and AKP activities. A8-1 also helps the cell relieve the inflammation induced by lipopolysaccharide by reducing the expression of cytokine IL-8 (P = 0.002) and TNF-α (P > 0.05), and increasing the IL-10 (P < 0.001). For the safety evaluation, A8-1 showed no hemolytic activity, no gelatinase activity, and had only asa1 positive in the seven detected virulence genes in polymerase chain reaction (PCR), whereas it was not predicted in the genome sequence. It was susceptible to benzylpenicillin, ampicillin, ciprofloxacin, levofloxacin, moxifloxacin, tigecycline, nitrofurantoin, linezolid, vancomycin, erythromycin, and quinupristin/dalofopine except clindamycin, which was verified by the predicted lasA, lmrB, lmrC, and lmrD genes contributing to the clindamycin resistance. The virulence test of G. mellonella showed that it had toxicity lower than 10% at 1 × 107 CFU. According to the results of these evaluated attributes, E. durans strain A8-1 could be a promising probiotic candidate for applications.
Collapse
Affiliation(s)
- Yi Zhou
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lu Shi
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Juan Wang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jia Yuan
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jin Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lijuan Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Rong Da
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yue Cheng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bei Han
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, China
| |
Collapse
|
15
|
Terzić-Vidojević A, Veljović K, Popović N, Tolinački M, Golić N. Enterococci from Raw-Milk Cheeses: Current Knowledge on Safety, Technological, and Probiotic Concerns. Foods 2021; 10:2753. [PMID: 34829034 PMCID: PMC8624194 DOI: 10.3390/foods10112753] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The present study is focused on the safety, technological characteristics, and probiotic evaluation of Enterococcus species from different artisanal raw milk dairy products, mainly cheeses with ripening. Apart from proteolytic and lipolytic activities, most enterococci show the ability to metabolize citrate and convert it to various aromatic compounds. Long-ripened cheeses therefore have a specific flavor that makes them different from cheeses produced from thermally treated milk with commercial starter cultures. In addition, enterococci are producers of bacteriocins effective against spoilage and pathogenic bacteria, so they can be used as food preservatives. However, the use of enterococci in the dairy industry should be approached with caution. Although originating from food, enterococci strains may carry various virulence factors and antibiotic-resistance genes and can have many adverse effects on human health. Still, despite their controversial status, the use of enterococci in the food industry is not strictly regulated since the existence of these so-called desirable and undesirable traits in enterococci is a strain-dependent characteristic. To be specific, the results of many studies showed that there are some enterococci strains that are safe for use as starter cultures or as probiotics since they do not carry virulence factors and antibiotic-resistance genes. These strains even exhibit strong health-promoting effects such as stimulation of the immune response, anti-inflammatory activity, hypocholesterolemic action, and usefulness in prevention/treatment of some diseases.
Collapse
Affiliation(s)
- Amarela Terzić-Vidojević
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (K.V.); (N.P.); (M.T.); (N.G.)
| | | | | | | | | |
Collapse
|
16
|
Brdarić E, Soković Bajić S, Đokić J, Đurđić S, Ruas-Madiedo P, Stevanović M, Tolinački M, Dinić M, Mutić J, Golić N, Živković M. Protective Effect of an Exopolysaccharide Produced by Lactiplantibacillus plantarum BGAN8 Against Cadmium-Induced Toxicity in Caco-2 Cells. Front Microbiol 2021; 12:759378. [PMID: 34790183 PMCID: PMC8591446 DOI: 10.3389/fmicb.2021.759378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/01/2021] [Indexed: 01/13/2023] Open
Abstract
Cadmium (Cd) ranks seventh on the list of most significant potential threats to human health based on its suspected toxicity and the possibility of exposure to it. It has been reported that some bacterial exopolysaccharides (EPSs) have the ability to bind heavy metal ions. We therefore investigated the capacity of eight EPS-producing lactobacilli to adsorb Cd in the present study, and Lactiplantibacillus plantarum BGAN8 was chosen as the best candidate. In addition, we demonstrate that an EPS derived from BGAN8 (EPS-AN8) exhibits a high Cd-binding capacity and prevents Cd-mediated toxicity in intestinal epithelial Caco-2 cells. Simultaneous use of EPS-AN8 with Cd treatment prevents inflammation, disruption of tight-junction proteins, and oxidative stress. Our results indicate that the EPS in question has a strong potential to be used as a postbiotic in combatting the adverse effects of Cd. Moreover, we show that higher concentrations of EPS-AN8 can alleviate Cd-induced cell damage.
Collapse
Affiliation(s)
- Emilija Brdarić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Svetlana Soković Bajić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Slađana Đurđić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias - Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Asturias, Spain
| | - Magdalena Stevanović
- Institute of Technical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Maja Tolinački
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miroslav Dinić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Mutić
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Nataša Golić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Milica Živković
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Dinić M, Jakovljević S, Đokić J, Popović N, Radojević D, Strahinić I, Golić N. Probiotic-mediated p38 MAPK immune signaling prolongs the survival of Caenorhabditis elegans exposed to pathogenic bacteria. Sci Rep 2021; 11:21258. [PMID: 34711881 PMCID: PMC8553853 DOI: 10.1038/s41598-021-00698-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
The host-microbiota cross-talk represents an important factor contributing to innate immune response and host resistance during infection. It has been shown that probiotic lactobacilli exhibit the ability to modulate innate immunity and enhance pathogen elimination. Here we showed that heat-inactivated probiotic strain Lactobacillus curvatus BGMK2-41 stimulates immune response and resistance of the Caenorhabditis elegans against Staphylococcus aureus and Pseudomonas aeruginosa. By employing qRT-PCR and western blot analysis we showed that heat-inactivated BGMK2-41 activated PMK-1/p38 MAPK immunity pathway which prolongs the survival of C. elegans exposed to pathogenic bacteria in nematode killing assays. The C. elegans pmk-1 mutant was used to demonstrate a mechanistic basis for the antimicrobial potential of BGMK2-41, showing that BGMK2-41 upregulated PMK-1/p38 MAPK dependent transcription of C-type lectins, lysozymes and tight junction protein CLC-1. Overall, this study suggests that PMK-1/p38 MAPK-dependent immune regulation by BGMK2-41 is essential for probiotic-mediated C. elegans protection against gram-positive and gram-negative bacteria and could be further explored for development of probiotics with the potential to increase resistance of the host towards pathogens.
Collapse
Affiliation(s)
- Miroslav Dinić
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia.
| | - Stefan Jakovljević
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| | - Jelena Đokić
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| | - Nikola Popović
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| | - Ivana Strahinić
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| | - Nataša Golić
- Laboratory for Molecular Microbiology (LMM), Institute of Molecular Genetics and Genetic Engineering (IMGGE), University of Belgrade, Belgrade, Serbia
| |
Collapse
|
18
|
Aljasir SF, D'Amico DJ. Probiotic potential of commercial dairy-associated protective cultures: In vitro and in vivo protection against Listeria monocytogenes infection. Food Res Int 2021; 149:110699. [PMID: 34600693 DOI: 10.1016/j.foodres.2021.110699] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 01/23/2023]
Abstract
Protective bacterial cultures (PCs) are commercially available to producers to control undesirable microbes in foods, including foodborne pathogens such as Listeria monocytogenes. They are generally recognized as safe for consumption and many are capable of producing bacteriocins. Yet their potential to act as probiotics and confer a health benefit on the host is not known. This study investigated the ability of three commercial PCs to survive human gastrointestinal conditions and exert anti-infective properties against L. monocytogenes. Counts of two PCs of Lactiplantibacillus plantarum remained unchanged after exposure to simulated gastrointestinal conditions, whereas counts of the PC Lactococcus lactis subsp. lactis were reduced by 5.3 log CFU/mL. Cultures of Lactiplantibacillus plantarum and Lactococcus lactis subsp. lactis adhered to human Caco-2 epithelial cells at ∼ 6 log CFU/mL. This pretreatment reduced subsequent L. monocytogenes adhesion and invasion by 1-1.6 log CFU/mL and 3.8-4.9 log CFU/mL, respectively, compared to control. L. monocytogenes-induced cytotoxicity was also reduced from 29.1% in untreated monolayers to ∼ 8% in those treated with PCs. Pretreatment of Caco-2 monolayers with Lactococcus lactis subsp. lactis and one PC of Lactiplantibacillus plantarum reduced L. monocytogenes translocation by ≥ 1.2 log CFU/mL compared to control (≥ 94.5% inhibition). All PCs significantly reduced DextranFITC permeability through Caco-2 monolayers to approximately half that of control. Pretreatment with PCs also reduced L. monocytogenes-induced mortality in Caenorhabditis elegans. These findings demonstrate the potential for commercially produced PCs to exert probiotic effects in the host through protection against L. monocytogenes infection, thus providing an additional benefit to food safety beyond inhibiting pathogen growth, survival, and virulence in foods.
Collapse
Affiliation(s)
- Sulaiman F Aljasir
- Department of Animal Science, University of Connecticut, Agricultural Biotechnology Laboratory, 1390 Storrs Road, U-4163, Storrs, CT 06269-4163, USA.
| | - Dennis J D'Amico
- Department of Animal Science, University of Connecticut, Agricultural Biotechnology Laboratory, 1390 Storrs Road, U-4163, Storrs, CT 06269-4163, USA.
| |
Collapse
|
19
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Effects of a Postbiotic and Prebiotic Mixture on Suckling Rats' Microbiota and Immunity. Nutrients 2021; 13:2975. [PMID: 34578853 PMCID: PMC8469903 DOI: 10.3390/nu13092975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023] Open
Abstract
Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
20
|
Wang J, Da R, Tuo X, Cheng Y, Wei J, Jiang K, Lv J, Adediji OM, Han B. Probiotic and Safety Properties Screening of Enterococcus faecalis from Healthy Chinese Infants. Probiotics Antimicrob Proteins 2021; 12:1115-1125. [PMID: 31845113 DOI: 10.1007/s12602-019-09625-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The aim of this study was to evaluate the probiotic characteristics and safety of seven Enterococcus faecalis isolates from fecal samples of healthy Chinese infants. We evaluated the isolates' tolerance to low pH, survival in bile salts and NaCl, adhesion ability, biofilm formation, antimicrobial activity, toxin gene distribution, hemolysis, gelatinase activity, antibiotic resistance, and virulence to Galleria mellonella. All strains survived at pH 5.0, in 7.0% NaCl, and in 3% bile salt. Adhesion to Caco-2 cells was above 10%. Strain A3-1 had higher adhesion ability toward mucin, collagen, and BSA in vitro, better antibacterial activity, and the strongest biofilm production. We detected seven virulence genes with a distribution of asa1 (100%), cylA (71.4%), esp (85.7%), hyl (14.3%), gelE (85.7%), ace (42.9%), and agg (71.4%). Although all strains were γ-hemolytic, none showed gelatinase activity based on physiological activity detection. All isolates were susceptible to benzylpenicillin, ampicillin, ciprofloxacin, levofloxacin, moxifloxacin, tigecycline, nitrofurantoin, linezolid, and vancomycin; they were not susceptible to erythromycin, quinupristin/dalofopine, and clindamycin. The virulence test of G. mellonella showed that, except for strains 106-1 and 113-1, the other strains had toxicity lower than 10%. Strain A3-1 may have the greatest potential to be developed as a probiotic.
Collapse
Affiliation(s)
- Juan Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Rong Da
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohong Tuo
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Yue Cheng
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jie Wei
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Kaichong Jiang
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Jia Lv
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Omolade Monisayo Adediji
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China
| | - Bei Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, P.O.44, NO.76 Yanta West Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
21
|
He Y, Liu X, Dong Y, Lei J, Ito K, Zhang B. Enterococcus faecium PNC01 isolated from the intestinal mucosa of chicken as an alternative for antibiotics to reduce feed conversion rate in broiler chickens. Microb Cell Fact 2021; 20:122. [PMID: 34182992 PMCID: PMC8240220 DOI: 10.1186/s12934-021-01609-z] [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: 02/06/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background The development and utilization of probiotics had many environmental benefits for replacing antibiotics in animal production. Bacteria in the intestinal mucosa have better adhesion to the host intestinal epithelial cells compared to bacteria in the intestinal contents. In this study, lactic acid bacteria were isolated from the intestinal mucosa of broiler chickens and investigated as the substitution to antibiotic in broiler production. Results In addition to acid resistance, high temperature resistance, antimicrobial sensitivity tests, and intestinal epithelial cell adhesion, Enterococcus faecium PNC01 (E. faecium PNC01) was showed to be non-cytotoxic to epithelial cells. Draft genome sequence of E. faecium PNC01 predicted that it synthesized bacteriocin to perform probiotic functions and bacteriocin activity assay showed it inhibited Salmonella typhimurium from invading intestinal epithelial cells. Diet supplemented with E. faecium PNC01 increased the ileal villus height and crypt depth in broiler chickens, reduced the relative length of the cecum at day 21, and reduced the relative length of jejunum and ileum at day 42. Diet supplemented with E. faecium PNC01 increased the relative abundance of Firmicutes and Lactobacillus, decreased the relative abundance of Bacteroides in the cecal microbiota. Conclusion E. faecium PNC01 replaced antibiotics to reduce the feed conversion rate. Furthermore, E. faecium PNC01 improved intestinal morphology and altered the composition of microbiota in the cecum to reduce feed conversion rate. Thus, it can be used as an alternative for antibiotics in broiler production to avoid the adverse impact of antibiotics by altering the gut microbiota. Graphic Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12934-021-01609-z.
Collapse
Affiliation(s)
- Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China.,College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Xuan Liu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Yuanyang Dong
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Jiaqi Lei
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Koichi Ito
- Department of Food and Physiological Models, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 3145 Ago, Kasama, Ibaraki, 319-0206, Japan
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China.
| |
Collapse
|
22
|
Abstract
Enterococci, being the common part of indigenous human microbiota and present in numerous fermented food products and probiotics, are often positioned as health-threatening bacterial pathogens. This makes medical community, especially part of it which is not involved in microbiology research, in an uncertainty regarding the legitimacy of using enterococcal probiotics in medical practice or even consuming them in foods such as cheese or salami. However, the difference between the clinical strains and strains used as probiotics is more than significant. The present paper tries to shed light on enterococci based on the contemporary knowledge of molecular microbiology.
Collapse
|
23
|
He Y, Yang Q, Tian L, Zhang Z, Qiu L, Tao X, Wei H. Protection of surface layer protein from Enterococcus faecium WEFA23 against Listeria monocytogenes CMCC54007 infection by modulating intestinal permeability and immunity. Appl Microbiol Biotechnol 2021; 105:4269-4284. [PMID: 33990856 DOI: 10.1007/s00253-021-11240-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Enterococcus faecium WEFA23 was previously found effectively against adherence and colonization of Listeria monocytogenes CMCC54007, which might be closely related to its surface layer protein (SLP). In this study, the protective of SLP of E. faecium WEFA23 against infection of L. monocytogenes CMCC54007 was systemically investigated. In vitro assay showed that SLP actively inhibited L. monocytogenes internalization into Caco-2 cell line, with decreasing mRNA level of pro-inflammation cytokines and virulence factors and restoring destroyed intestinal barrier. In vivo assay through excluding SLP of E. faecium WEFA23 by 5 M LiCl represented that SLP increased body weight, reduced mortality and cell counts of L. monocytogenes CMCC54007 in tissues of mice. Further researches showed that SLP protected against L. monocytogenes CMCC54007 infection by modulation of intestinal permeability and immunity, namely, it decreased fluorescein isothiocyanate (FITC)-Dextran in serum, ameliorated destroyed colon structure, and increased number of goblet cells and protein level of TJ protein (Claudin-1, Occludin, and ZO-1) in colon. For immunity, SLP decreased number of CD4+ and CD8+ T cells in liver, mRNA level, and content of pro-inflammatory factors IL-6, IL-1β, IFN-γ ,TNF-α, and NO, and restored the structure of liver and spleen. Key Points•SLP of E. faecium inhibited L. monocytogenes internalization and colonization•SLP of E. faecium ameliorated host intestinal barrier dysfunction•SLP of E. faecium decreased pro-inflammatory cytokines and cells.
Collapse
Affiliation(s)
- Yao He
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Qin Yang
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Linlin Tian
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Zhihong Zhang
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Liang Qiu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, People's Republic of China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, People's Republic of China
| | - Hua Wei
- Jiangxi-OAI Joint Research Institute, Nanchang University, 235 Nanjing Donglu, Nanchang, Jiangxi, 330047, People's Republic of China.
| |
Collapse
|
24
|
Raheem A, Liang L, Zhang G, Cui S. Modulatory Effects of Probiotics During Pathogenic Infections With Emphasis on Immune Regulation. Front Immunol 2021; 12:616713. [PMID: 33897683 PMCID: PMC8060567 DOI: 10.3389/fimmu.2021.616713] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In order to inhibit pathogenic complications and to enhance animal and poultry growth, antibiotics have been extensively used for many years. Antibiotics applications not only affect target pathogens but also intestinal beneficially microbes, inducing long-lasting changes in intestinal microbiota associated with diseases. The application of antibiotics also has many other side effects like, intestinal barrier dysfunction, antibiotics residues in foodstuffs, nephropathy, allergy, bone marrow toxicity, mutagenicity, reproductive disorders, hepatotoxicity carcinogenicity, and antibiotic-resistant bacteria, which greatly compromise the efficacy of antibiotics. Thus, the development of new antibiotics is necessary, while the search for antibiotic alternatives continues. Probiotics are considered the ideal antibiotic substitute; in recent years, probiotic research concerning their application during pathogenic infections in humans, aquaculture, poultry, and livestock industry, with emphasis on modulating the immune system of the host, has been attracting considerable interest. Hence, the adverse effects of antibiotics and remedial effects of probiotics during infectious diseases have become central points of focus among researchers. Probiotics are live microorganisms, and when given in adequate quantities, confer good health effects to the host through different mechanisms. Among them, the regulation of host immune response during pathogenic infections is one of the most important mechanisms. A number of studies have investigated different aspects of probiotics. In this review, we mainly summarize recent discoveries and discuss two important aspects: (1) the application of probiotics during pathogenic infections; and (2) their modulatory effects on the immune response of the host during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Abdul Raheem
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Lin Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Guangzhi Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Shangjin Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| |
Collapse
|
25
|
Host-commensal interaction promotes health and lifespan in Caenorhabditis elegans through the activation of HLH-30/TFEB-mediated autophagy. Aging (Albany NY) 2021; 13:8040-8054. [PMID: 33770762 PMCID: PMC8034897 DOI: 10.18632/aging.202885] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/13/2021] [Indexed: 11/25/2022]
Abstract
Gut homeostasis is maintained by the close interaction between commensal intestinal microbiota and the host, affecting the most complex physiological processes, such as aging. Some commensal bacteria with the potential to promote healthy aging arise as attractive candidates for the development of pro-longevity probiotics. Here, we showed that heat-inactivated human commensal Lactobacillus fermentum BGHV110 (BGHV110) extends the lifespan of Caenorhabditis elegans and improves age-related physiological features, including locomotor function and lipid metabolism. Mechanistically, we found that BGHV110 promotes HLH-30/TFEB-dependent autophagy to delay aging, as longevity assurance was completely abolished in the mutant lacking HLH-30, a major autophagy regulator in C. elegans. Moreover, we observed that BGHV110 partially decreased the content of lipid droplets in an HLH-30-dependent manner and, at the same time, slightly increased mitochondrial activity. In summary, this study demonstrates that specific factors from commensal bacteria can be used to exploit HLH-30/TFEB-mediated autophagy in order to promote longevity and fitness of the host.
Collapse
|
26
|
Antilisterial Potential of Lactic Acid Bacteria in Eliminating Listeria monocytogenes in Host and Ready-to-Eat Food Application. MICROBIOLOGY RESEARCH 2021. [DOI: 10.3390/microbiolres12010017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Listeriosis is a severe food borne disease with a mortality rate of up to 30% caused by pathogenic Listeria monocytogenes via the production of several virulence factors including listeriolysin O (LLO), transcriptional activator (PrfA), actin (Act), internalin (Int), etc. It is a foodborne disease predominantly causing infections through consumption of contaminated food and is often associated with ready-to-eat food (RTE) and dairy products. Common medication for listeriosis such as antibiotics might cause an eagle effect and antibiotic resistance if it is overused. Therefore, exploration of the use of lactic acid bacteria (LAB) with probiotic characteristics and multiple antimicrobial properties is increasingly getting attention for their capability to treat listeriosis, vaccine development, and hurdle technologies. The antilisterial gene, a gene coding to produce antimicrobial peptide (AMP), one of the inhibitory substances found in LAB, is one of the potential key factors in listeriosis treatment, coupled with the vast array of functions and strategies; this review summarizes the various strategies by LAB against L. monocytogenes and the prospect in development of a ‘generally regarded as safe’ LAB for treatment of listeriosis.
Collapse
|
27
|
Tight Junctions as a Key for Pathogens Invasion in Intestinal Epithelial Cells. Int J Mol Sci 2021; 22:ijms22052506. [PMID: 33801524 PMCID: PMC7958858 DOI: 10.3390/ijms22052506] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Tight junctions play a major role in maintaining the integrity and impermeability of the intestinal barrier. As such, they act as an ideal target for pathogens to promote their translocation through the intestinal mucosa and invade their host. Different strategies are used by pathogens, aimed at directly destabilizing the junctional network or modulating the different signaling pathways involved in the modulation of these junctions. After a brief presentation of the organization and modulation of tight junctions, we provide the state of the art of the molecular mechanisms leading to permeability breakdown of the gut barrier as a consequence of tight junctions’ attack by pathogens, including bacteria, viruses, fungi, and parasites.
Collapse
|
28
|
Maternal Linoleic Acid Overconsumption Alters Offspring Gut and Adipose Tissue Homeostasis in Young but Not Older Adult Rats. Nutrients 2020; 12:nu12113451. [PMID: 33187208 PMCID: PMC7697261 DOI: 10.3390/nu12113451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/26/2020] [Accepted: 11/09/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal n-6 polyunsaturated fatty acids (PUFA) consumption during gestation and lactation can predispose offspring to the development of metabolic diseases such as obesity later in life. However, the mechanisms underlying the potential programming effect of n-6 PUFA upon offspring physiology are not yet all established. Herein, we investigated the effects of maternal and weaning linoleic acid (LA)-rich diet interactions on gut intestinal and adipose tissue physiology in young (3-month-old) and older (6-month-old) adult offspring. Pregnant rats were fed a control diet (2% LA) or an LA-rich diet (12% LA) during gestation and lactation. At weaning, offspring were either maintained on the maternal diet or fed the other diet for 3 or 6 months. At 3 months of age, the maternal LA-diet favored low-grade inflammation and greater adiposity, while at 6 months of age, offspring intestinal barrier function, adipose tissue physiology and hepatic conjugated linoleic acids were strongly influenced by the weaning diet. The maternal LA-diet impacted offspring cecal microbiota diversity and composition at 3 months of age, but had only few remnant effects upon cecal microbiota composition at 6 months of age. Our study suggests that perinatal exposure to high LA levels induces a differential metabolic response to weaning diet exposure in adult life. This programming effect of a maternal LA-diet may be related to the alteration of offspring gut microbiota.
Collapse
|
29
|
Popović N, Brdarić E, Đokić J, Dinić M, Veljović K, Golić N, Terzić-Vidojević A. Yogurt Produced by Novel Natural Starter Cultures Improves Gut Epithelial Barrier In Vitro. Microorganisms 2020; 8:E1586. [PMID: 33076224 PMCID: PMC7602395 DOI: 10.3390/microorganisms8101586] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Yogurt is a traditional fermented dairy product, prepared with starter cultures containing Streptococcus thermophilus and Lactobacillus bulgaricus that has gained widespread consumer acceptance as a healthy food. It is widely accepted that yogurt cultures have been recognized as probiotics, due to their beneficial effects on human health. In this study, we have characterized technological and health-promoting properties of autochthonous strains S. thermophilus BGKMJ1-36 and L. bulgaricus BGVLJ1-21 isolated from artisanal sour milk and yogurt, respectively, in order to be used as functional yogurt starter cultures. Both BGKMJ1-36 and BGVLJ1-21 strains have the ability to form curd after five hours at 42 °C, hydrolyze αs1-, β-, and κ- casein, and to show antimicrobial activity toward Listeria monocytogenes. The strain BGKMJ1-36 produces exopolysaccharides important for rheological properties of the yogurt. The colonies of BGKMJ1-36 and BGVLJ1-21 strains that successfully survived transit of the yogurt through simulated gastrointestinal tract conditions have been tested for adhesion to intestinal epithelial Caco-2 cells. The results reveal that both strains adhere to Caco-2 cells and significantly upregulate the expression of autophagy-, tight junction proteins-, and anti-microbial peptides-related genes. Hence, both strains may be interesting for use as a novel functional starter culture for production of added-value yogurt with health-promoting properties.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Amarela Terzić-Vidojević
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.P.); (E.B.); (J.Đ.); (M.D.); (K.V.); (N.G.)
| |
Collapse
|
30
|
Terzić-Vidojević A, Veljović K, Tolinački M, Živković M, Lukić J, Lozo J, Fira Đ, Jovčić B, Strahinić I, Begović J, Popović N, Miljković M, Kojić M, Topisirović L, Golić N. Diversity of non-starter lactic acid bacteria in autochthonous dairy products from Western Balkan Countries - Technological and probiotic properties. Food Res Int 2020; 136:109494. [PMID: 32846575 DOI: 10.1016/j.foodres.2020.109494] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
The aim of this review was to summarize the data regarding diversity of non-starter lactic acid bacteria (NSLAB) isolated from various artisanal dairy products manufactured in Western Balkan Countries. The dairy products examined were manufactured from raw cow's, sheep's or goat's milk or mixed milk, in the traditional way without the addition of commercial starter cultures. Dairy products such as white brined cheese, fresh cheese, hard cheese, yogurt, sour cream and kajmak were sampled in the households of Serbia, Croatia, Slovenia, Bosnia and Herzegovina, Montenegro, and North Macedonia. It has been established that the diversity of lactic acid bacteria (LAB) from raw milk artisanal dairy products is extensive. In the reviewed literature, 28 LAB species and a large number of strains belonging to the Lactobacillus, Lactococcus, Enterococcus, Streptococcus, Pediococcus, Leuconostoc and Weissella genera were isolated from various dairy products. Over 3000 LAB strains were obtained and characterized for their technological and probiotic properties including: acidification and coagulation of milk, production of aromatic compounds, proteolytic activity, bacteriocins production and competitive exclusion of pathogens, production of exopolysaccharides, aggregation ability and immunomodulatory effect. Results show that many of the isolated NSLAB strains had one, two or more of the properties mentioned. The data presented emphasize the importance of artisanal products as a valuable source of NSLAB with unique technological and probiotic features important both as a base for scientific research as well as for designing novel starter cultures for functional dairy food.
Collapse
Affiliation(s)
- Amarela Terzić-Vidojević
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia.
| | - Katarina Veljović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Maja Tolinački
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Milica Živković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Jovanka Lukić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Jelena Lozo
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| | - Đorđe Fira
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| | - Branko Jovčić
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
| | - Ivana Strahinić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Jelena Begović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Nikola Popović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Marija Miljković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Milan Kojić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Ljubiša Topisirović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| | - Nataša Golić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade 152, Serbia
| |
Collapse
|
31
|
Burkholder KM, Fletcher DH, Gileau L, Kandolo A. Lactic acid bacteria decrease Salmonella enterica Javiana virulence and modulate host inflammation during infection of an intestinal epithelial cell line. Pathog Dis 2020; 77:5480463. [PMID: 31065694 DOI: 10.1093/femspd/ftz025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
Salmonella enterica Javiana is a leading cause of severe foodborne Salmonellosis. Despite its emergence as a major foodborne pathogen, little is known of how S. Javiana interacts with intestinal epithelial cells, or of potential methods for ameliorating the bacterial-host interaction. Using cell-based adhesion, invasion and lactate dehydrogenase release assays, we observed an invasive and cytotoxic effect of S. Javiana on intestinal epithelial cells. We assessed the effect of probiotic species of lactic acid bacteria (LAB) on the S. Javiana-host cell interaction, and hypothesized that LAB would reduce S. Javiana infectivity. Salmonella enterica Javiana invasion was significantly impaired in host cells pre-treated with live Lactobacillus acidophilus and Lactobacillus rhamnosus. In addition, pre-exposure of host cells to live L. acidophilus, L. rhamnosus and L. casei reduced S. Javiana-induced cytotoxicity, while heat-killed LAB cultures had no effect on S. Javiana invasion or cytotoxicity. qRT-PCR analysis revealed that S. Javiana exposed to L. acidophilus and L. rhamnosus exhibited reduced virulence gene expression. Moreover, pre-treating host cells with LAB prior to S. Javiana infection reduced host cell production of inflammatory cytokines. Data suggest a potential protective effect of L. acidophilus, L. rhamnosus and L. casei against intestinal epithelial infection and pathogen-induced damage caused by S. Javiana.
Collapse
Affiliation(s)
- Kristin M Burkholder
- University of New England, Department of Biology, 11 Hills Beach Rd, Biddeford, ME, USA 04005
| | - Dylan H Fletcher
- University of New England, Department of Biology, 11 Hills Beach Rd, Biddeford, ME, USA 04005
| | - Lauren Gileau
- University of New England, Department of Biology, 11 Hills Beach Rd, Biddeford, ME, USA 04005
| | - Arnold Kandolo
- University of New England, Department of Biology, 11 Hills Beach Rd, Biddeford, ME, USA 04005
| |
Collapse
|
32
|
Oikonomou G, Addis MF, Chassard C, Nader-Macias MEF, Grant I, Delbès C, Bogni CI, Le Loir Y, Even S. Milk Microbiota: What Are We Exactly Talking About? Front Microbiol 2020; 11:60. [PMID: 32117107 PMCID: PMC7034295 DOI: 10.3389/fmicb.2020.00060] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/13/2020] [Indexed: 12/17/2022] Open
Abstract
The development of powerful sequencing techniques has allowed, albeit with some biases, the identification and inventory of complex microbial communities that inhabit different body sites or body fluids, some of which were previously considered sterile. Notably, milk is now considered to host a complex microbial community with great diversity. Milk microbiota is now well documented in various hosts. Based on the growing literature on this microbial community, we address here the question of what milk microbiota is. We summarize and compare the microbial composition of milk in humans and in ruminants and address the existence of a putative core milk microbiota. We discuss the factors that contribute to shape the milk microbiota or affect its composition, including host and environmental factors as well as methodological factors, such as the sampling and sequencing techniques, which likely introduce distortion in milk microbiota analysis. The roles that milk microbiota are likely to play in the mother and offspring physiology and health are presented together with recent data on the hypothesis of an enteromammary pathway. At last, this fascinating field raises a series of questions, which are listed and commented here and which open new research avenues.
Collapse
Affiliation(s)
- Georgios Oikonomou
- Institute of Veterinary Science, University of Liverpool, Neston, United Kingdom
| | - Maria Filippa Addis
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Milan, Italy
| | | | | | - I Grant
- Institute of Veterinary Science, University of Liverpool, Neston, United Kingdom
| | - Celine Delbès
- Université Clermont Auvergne, INRAE, UMRF, Aurillac, France
| | - Cristina Inés Bogni
- Departamento de Microbiología e Inmunología, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Universidad Nacional de Río Cuarto, Río Cuarto, Argentina
| | - Yves Le Loir
- STLO, UMR 1253, INRAE, Agrocampus Ouest, Rennes, France
| | - Sergine Even
- STLO, UMR 1253, INRAE, Agrocampus Ouest, Rennes, France
| |
Collapse
|
33
|
Dempsey JL, Cui JY. Microbiome is a functional modifier of P450 drug metabolism. CURRENT PHARMACOLOGY REPORTS 2019; 5:481-490. [PMID: 33312848 PMCID: PMC7731899 DOI: 10.1007/s40495-019-00200-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Host cytochrome P450s (P450s) play important roles in the bioactivation and detoxification of numerous therapeutic drugs, environmental toxicants, dietary factors, as well as endogenous compounds. Gut microbiome is increasingly recognized as our "second genome" that contributes to the xenobiotic biotransformation of the host, and the first pass metabolism of many orally exposed chemicals is a joint effort between host drug metabolizing enzymes including P450s and gut microbiome. Gut microbiome contributes to the drug metabolism via two distinct mechanisms: direct mechanism refers to the metabolism of drugs by microbial enzymes, among which reduction and hydrolysis (or deconjugation) are among the most important reactions; whereas indirect mechanism refers to the influence of host receptors and signaling pathways by microbial metabolites. Many types of microbial metabolites, such as secondary bile acids (BAs), short chain fatty acids (SCFAs), and tryptophan metabolites, are known regulators of human diseases through modulating host xenobiotic-sensing receptors. To study the roles of gut microbiome in regulating host drug metabolism including P450s, several models including germ free mice, antibiotics or probiotics treatments, have been widely used. The present review summarized the current information regarding the interactions between microbiome and the host P450s in xenobiotic biotransformation organs such as liver, intestine, and kidney, highlighting the remote sensing mechanisms underlying gut microbiome mediated regulation of host xenobiotic biotransformation. In addition, the roles of bacterial, fungal, and other microbiome kingdom P450s, which is an understudied area of research in pharmacology and toxicology, are discussed.
Collapse
Affiliation(s)
- Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington
| |
Collapse
|