1
|
Maddineni G, Obulareddy SJ, Paladiya RD, Korsapati RR, Jain S, Jeanty H, Vikash F, Tummala NC, Shetty S, Ghazalgoo A, Mahapatro A, Polana V, Patel D. The role of gut microbiota augmentation in managing non-alcoholic fatty liver disease: an in-depth umbrella review of meta-analyses with grade assessment. Ann Med Surg (Lond) 2024; 86:4714-4731. [PMID: 39118769 PMCID: PMC11305784 DOI: 10.1097/ms9.0000000000002276] [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: 02/19/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Background and aim Currently, there are no authorized medications specifically for non-alcoholic fatty liver disease (NAFLD) treatment. Studies indicate that changes in gut microbiota can disturb intestinal balance and impair the immune system and metabolism, thereby elevating the risk of developing and exacerbating NAFLD. Despite some debate, the potential benefits of microbial therapies in managing NAFLD have been shown. Methods A systematic search was undertaken to identify meta-analyses of randomized controlled trials that explored the effects of microbial therapy on the NAFLD population. The goal was to synthesize the existing evidence-based knowledge in this field. Results The results revealed that probiotics played a significant role in various aspects, including a reduction in liver stiffness (MD: -0.38, 95% CI: [-0.49, -0.26]), hepatic steatosis (OR: 4.87, 95% CI: [1.85, 12.79]), decrease in body mass index (MD: -1.46, 95% CI: [-2.43, -0.48]), diminished waist circumference (MD: -1.81, 95% CI: [-3.18, -0.43]), lowered alanine aminotransferase levels (MD: -13.40, 95% CI: [-17.02, -9.77]), decreased aspartate aminotransferase levels (MD: -13.54, 95% CI: [-17.85, -9.22]), lowered total cholesterol levels (MD: -15.38, 95% CI: [-26.49, -4.26]), decreased fasting plasma glucose levels (MD: -4.98, 95% CI: [-9.94, -0.01]), reduced fasting insulin (MD: -1.32, 95% CI: [-2.42, -0.21]), and a decline in homeostatic model assessment of insulin resistance (MD: -0.42, 95% CI: [-0.72, -0.11]) (P<0.05). Conclusion Overall, the results demonstrated that gut microbiota interventions could ameliorate a wide range of indicators including glycemic profile, dyslipidemia, anthropometric indices, and liver injury, allowing them to be considered a promising treatment strategy.
Collapse
Affiliation(s)
| | | | | | | | - Shika Jain
- MVJ Medical College and Research Hospital, Bengaluru, Karnataka, India
| | | | - Fnu Vikash
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx
| | - Nayanika C. Tummala
- Gitam Institute of Medical Sciences and Research, Visakhapatnam, Andhra Pradesh
| | | | - Arezoo Ghazalgoo
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | - Dhruvan Patel
- Drexel University College of Medicine, Philadelphia, Pennsylvania, PA
| |
Collapse
|
2
|
Amini-Salehi E, Hassanipour S, Keivanlou MH, Shahdkar M, Orang Goorabzarmakhi M, Vakilpour A, Joukar F, Hashemi M, Sattari N, Javid M, Mansour-Ghanaei F. The impact of gut microbiome-targeted therapy on liver enzymes in patients with nonalcoholic fatty liver disease: an umbrella meta-analysis. Nutr Rev 2024; 82:815-830. [PMID: 37550264 DOI: 10.1093/nutrit/nuad086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is considered the leading cause of chronic liver disease worldwide. To date, no confirmed medication is available for the treatment of NAFLD. Previous studies showed the promising effects of gut microbiome-targeted therapies; however, the results were controversial and the strength of the evidence and their clinical significance remained unclear. OBJECTIVES This umbrella study summarizes the results of meta-analyses investigating the effects of probiotics, prebiotics, and synbiotics on liver enzymes in the NAFLD population. DATA SOURCE A comprehensive search of the PubMed, Scopus, Web of Science, and Cochrane Library databases was done up to December 20, 2022, to find meta-analyses on randomized control trials reporting the effects of gut microbial therapy on patients with NAFLD. DATA EXTRACTION Two independent investigators extracted data on the characteristics of meta-analyses, and any discrepancies were resolved by a third researcher. The AMSTAR2 checklist was used for evaluating the quality of studies. DATA ANALYSIS A final total of 15 studies were included in the analysis. Results showed that microbiome-targeted therapies could significantly reduce levels of alanine aminotransferase (ALT; effect size [ES], -10.21; 95% confidence interval [CI], -13.29, -7.14; P < 0.001), aspartate aminotransferase (AST; ES, -8.86; 95%CI, -11.39, -6.32; P < 0.001), and γ-glutamyltransferase (ES, -5.56; 95%CI, -7.92, -3.31; P < 0.001) in patients with NAFLD. Results of subgroup analysis based on intervention showed probiotics could significantly reduce levels of AST (ES, -8.69; 95%CI, -11.01, -6.37; P < 0.001) and ALT (ES, -9.82; 95%CI, -11.59, -8.05; P < 0.001). Synbiotics could significantly reduce levels of AST (ES, -11.40; 95%CI, -13.91, -8.88; P < 0.001) and ALT (ES, -11.87; 95%CI, -13.80, -9.95; P < 0.001). Prebiotics had no significant effects on AST and ALT levels (ES, -2.96; 95%CI, -8.12, 2.18, P = 0.259; and ES, -4.69; 95%CI, -13.53, 4.15, P = 0.299, respectively). CONCLUSION Gut microbiome-targeted therapies could be a promising therapeutic approach in the improvement of hepatic damage in patients with NAFLD. However, more studies are needed to better determine the best bacterial strains, duration of treatment, and optimum dosage of gut microbiome-targeted therapies in the treatment of the NAFLD population. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022346998.
Collapse
Affiliation(s)
- Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Milad Shahdkar
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Azin Vakilpour
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Li X, Wang S, Wu P, Nan D, Chen D, Suo L, Lu X, Rao C, Li Q, Yue J, Xiang Y, Mao X, Yan J. Effect of O antigen glycosyl isomerase gene mutation on biological property and pathogenicity of Burkholderia pseudomallei strain BPC006. Int J Biol Macromol 2024; 258:128922. [PMID: 38141699 DOI: 10.1016/j.ijbiomac.2023.128922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Burkholderia pseudomallei, an intracellular pathogen, is responsible for melioidosis, a zoonotic disease. Its pathogenesis involves several virulence factors, among which lipopolysaccharide (LPS) plays a crucial role. Our research reveals that the O antigen present within the LPS significantly regulates the host immune response. In a previous study, we obtained a B. pseudomallei mutant strain ΔwbiI. Here, the purification of LPS from ΔwbiI and a gas chromatography-mass spectrometry (GC-MS) analysis were conducted. The results confirmed the absence of specific sugar 6-deoxy-Talp, which is a typical component of the O antigen in the wild type B. pseudomallei. Our findings underscore the potent impact the O antigen exerts on the virulence of B. pseudomallei. The ΔwbiI strain displayed significantly increased invasiveness and cytotoxicity in vitro. This enhanced cytotoxicity seems to be related to the exposure of lipid A and an increased cell membrane hydrophobicity resulting from the deletion of the O antigen. Additionally, in mouse models, the ΔwbiI strain resulted in a heightened host lethality and an excessive inflammatory response in mice. These findings indicate that the O-antigenic polysaccharide moiety of B. pseudomallei plays a role in its pathogenicity in vitro and in vivo.
Collapse
Affiliation(s)
- Xiao Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shiwei Wang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Pan Wu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dongqi Nan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dan Chen
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Liangpeng Suo
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaoxue Lu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chenglong Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Juanjuan Yue
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yang Xiang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Jingmin Yan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
4
|
Liu H, Yu Y, Dong A, Elsabahy M, Yang Y, Gao H. Emerging strategies for combating Fusobacterium nucleatum in colorectal cancer treatment: Systematic review, improvements and future challenges. EXPLORATION (BEIJING, CHINA) 2024; 4:20230092. [PMID: 38854496 PMCID: PMC10867388 DOI: 10.1002/exp.20230092] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 11/16/2023] [Indexed: 06/11/2024]
Abstract
Colorectal cancer (CRC) is generally characterized by a high prevalence of Fusobacterium nucleatum (F. nucleatum), a spindle-shaped, Gram-negative anaerobe pathogen derived from the oral cavity. This tumor-resident microorganism has been closely correlated with the occurrence, progression, chemoresistance and immunosuppressive microenvironment of CRC. Furthermore, F. nucleatum can specifically colonize CRC tissues through adhesion on its surface, forming biofilms that are highly resistant to commonly used antibiotics. Accordingly, it is crucial to develop efficacious non-antibiotic approaches to eradicate F. nucleatum and its biofilms for CRC treatment. In recent years, various antimicrobial strategies, such as natural extracts, inorganic chemicals, organic chemicals, polymers, inorganic-organic hybrid materials, bacteriophages, probiotics, and vaccines, have been proposed to combat F. nucleatum and F. nucleatum biofilms. This review summarizes the latest advancements in anti-F. nucleatum research, elucidates the antimicrobial mechanisms employed by these systems, and discusses the benefits and drawbacks of each antimicrobial technology. Additionally, this review also provides an outlook on the antimicrobial specificity, potential clinical implications, challenges, and future improvements of these antimicrobial strategies in the treatment of CRC.
Collapse
Affiliation(s)
- Hongyu Liu
- State Key Laboratory of Separation Membranes and Membrane ProcessesSchool of Materials Science and EngineeringTiangong UniversityTianjinP. R. China
| | - Yunjian Yu
- State Key Laboratory of Separation Membranes and Membrane ProcessesSchool of Materials Science and EngineeringTiangong UniversityTianjinP. R. China
| | - Alideertu Dong
- College of Chemistry and Chemical EngineeringInner Mongolia UniversityHohhotP. R. China
| | - Mahmoud Elsabahy
- Department of PharmaceuticsFaculty of PharmacyAssiut UniversityAssiutEgypt
| | - Ying‐Wei Yang
- International Joint Research Laboratory of Nano‐Micro Architecture ChemistryCollege of ChemistryJilin UniversityChangchunP. R. China
| | - Hui Gao
- State Key Laboratory of Separation Membranes and Membrane ProcessesSchool of Materials Science and EngineeringTiangong UniversityTianjinP. R. China
| |
Collapse
|
5
|
Khataybeh B, Jaradat Z, Ababneh Q. Anti-bacterial, anti-biofilm and anti-quorum sensing activities of honey: A review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116830. [PMID: 37400003 DOI: 10.1016/j.jep.2023.116830] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Man has used honey to treat diseases since ancient times, perhaps even before the history of medicine itself. Several civilizations have utilized natural honey as a functional and therapeutic food to ward off infections. Recently, researchers worldwide have been focusing on the antibacterial effects of natural honey against antibiotic-resistant bacteria. AIM OF THE STUDY This review aims to summarize research on the use of honey properties and constituents with their anti-bacterial, anti-biofilm, and anti-quorum sensing mechanisms of action. Further, honey's bacterial products, including probiotic organisms and antibacterial agents which are produced to curb the growth of other competitor microorganisms is addressed. MATERIALS AND METHODS In this review, we have provided a comprehensive overview of the antibacterial, anti-biofilm, and anti-quorum sensing activities of honey and their mechanisms of action. Furthermore, the review addressed the effects of antibacterial agents of honey from bacterial origin. Relevant information on the antibacterial activity of honey was obtained from scientific online databases such as Web of Science, Google Scholar, ScienceDirect, and PubMed. RESULTS Honey's antibacterial, anti-biofilm, and anti-quorum sensing activities are mostly attributed to four key components: hydrogen peroxide, methylglyoxal, bee defensin-1, and phenolic compounds. The performance of bacteria can be altered by honey components, which impact their cell cycle and cell morphology. To the best of our knowledge, this is the first review that specifically summarizes every phenolic compound identified in honey along with their potential antibacterial mechanisms of action. Furthermore, certain strains of beneficial lactic acid bacteria such as Bifidobacterium, Fructobacillus, and Lactobacillaceae, as well as Bacillus species can survive and even grow in honey, making it a potential delivery system for these agents. CONCLUSION Honey could be regarded as one of the best complementary and alternative medicines. The data presented in this review will enhance our knowledge of some of honey's therapeutic properties as well as its antibacterial activities.
Collapse
Affiliation(s)
- Batool Khataybeh
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ziad Jaradat
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Qutaiba Ababneh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
6
|
Liu Y, Liu Q, Zhang C, Zhao J, Zhang H, Chen W, Zhai Q. Strain-specific effects of Akkermansia muciniphila on the regulation of intestinal barrier. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
7
|
Saeed A, Yasmin A, Baig M, Ahmed MA, Farooqi ZUR. Streptococcus lactarius MB622 and Streptococcus salivarius MB620 isolated from human milk reduce chemokine IL-8 production in response to TNF-α in Caco-2 cell line, an exploratory study. Cytokine 2023; 168:156232. [PMID: 37224578 DOI: 10.1016/j.cyto.2023.156232] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023]
Abstract
Streptococci are a predominant genera of the human milk microbiome. Among different lactic acid bacteria (LAB) a few Streptococcal strains are also considered as probiotics. Probiotic bacteria are reported to modulate immunity when consumed in adequate amount and bacterial hydrophobicity can be considered as a preliminary experiment for the adhesive capability of probiotic bacteria to the epithelial cells. The present study aimed to investigate the probiotic, hydrophobic and immune modulation property of Streptococcus lactarius MB622 and Streptococcus salivarius MB620, isolated from human milk. S. lactarius MB622 and S. salivarius MB620 displayed higher hydrophobicity (78 % and 59 % respectively) in addition to intrinsic probiotic properties such as gram positive classification, catalase negative activity, resistance to artificially stimulated gastric juice and gastrointestinal bile salt concentration. In conclusion Streptococcus lactarius MB622 and Streptococcus salivarius MB620 isolated from human milk when administered in sufficient amount and for certain duration could be used to reduce inflammation inside the colon by reducing the production of inflammatory booster (IL-8) in diseased state.
Collapse
Affiliation(s)
- Ayesha Saeed
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University Rawalpindi, Pakistan.
| | - Azra Yasmin
- Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University Rawalpindi, Pakistan
| | - Mehreen Baig
- Surgical Unit II, Foundation University Islamabad, Pakistan
| | | | | |
Collapse
|
8
|
Niu X, Yin X, Wu X, Zhang Q, Jiang Y, He J, Zhao Y, Zhang C, Ren Y, Lai M, Sang Y, Wang R. Heat-Killed Bifidobacterium longum BBMN68 in Pasteurized Yogurt Alleviates Mugwort Pollen-Induced Allergic Airway Responses through Gut Microbiota Modulation in a Murine Model. Foods 2023; 12:2049. [PMID: 37238867 PMCID: PMC10217734 DOI: 10.3390/foods12102049] [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: 04/12/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Many probiotic bacteria have been proven to prevent allergic airway responses through immunomodulation. This study was conducted to evaluate the effects of heat-killed Bifidobacterium longum BBMN68 (BBMN68) in pasteurized yogurt on the alleviation of mugwort pollen (MP)-induced allergic inflammation. BALB/c mice aged 5-6 weeks were randomly assigned and fed pasteurized yogurt containing heat-killed BBMN68 for 27 days, followed by allergic sensitization and challenge with MP extract. The allergic mice that received pasteurized yogurt containing heat-killed BBMN68 had improved immune status, including a lower serum IgE level, decreased serum interleukin (IL)-4, IL-5, and IL-13 concentrations, and alleviated airway inflammation manifested by increased macrophage and decreased eosinophil and neutrophil counts in BALF, as well as airway remodeling and suppressed peribronchial cellular infiltration. Moreover, oral administration of pasteurized yogurt containing heat-killed BBMN68 significantly modulated gut microbiota composition by influencing the proportion of beneficial genera associated with inflammation and immunity, such as Lactobacillus, Candidatus_Saccharimonas, Odoribacter, and Parabacteroides, which also negatively correlated with serum IgE and Th2 cytokine levels. These results demonstrated that pasteurized yogurt containing heat-killed BBMN68 had mitigative effects on allergic airway inflammation, likely through maintaining the systemic Th1/Th2 immune balance by altering the structure and function of the gut microbiota.
Collapse
Affiliation(s)
- Xiaokang Niu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xindi Yin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xiuying Wu
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yunyun Jiang
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Jingjing He
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yuyang Zhao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chao Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yimei Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Mengxuan Lai
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Yue Sang
- Hebei Engineering Research Center of Animal Product, Sanhe 065200, China
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| |
Collapse
|
9
|
Niu X, Yin X, Wu X, Zhang Q, Jiang Y, He J, Zhao Y, Zhang C, Ren Y, Lai M, Sang Y, Wang R. Heat-Killed Bifidobacterium longum BBMN68 in Pasteurized Yogurt Alleviates Mugwort Pollen-Induced Allergic Airway Responses through Gut Microbiota Modulation in a Murine Model. Foods 2023; 12:2049. [PMID: 37238867 DOI: 10.3390/foods12102049if:] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 07/26/2024] Open
Abstract
Many probiotic bacteria have been proven to prevent allergic airway responses through immunomodulation. This study was conducted to evaluate the effects of heat-killed Bifidobacterium longum BBMN68 (BBMN68) in pasteurized yogurt on the alleviation of mugwort pollen (MP)-induced allergic inflammation. BALB/c mice aged 5-6 weeks were randomly assigned and fed pasteurized yogurt containing heat-killed BBMN68 for 27 days, followed by allergic sensitization and challenge with MP extract. The allergic mice that received pasteurized yogurt containing heat-killed BBMN68 had improved immune status, including a lower serum IgE level, decreased serum interleukin (IL)-4, IL-5, and IL-13 concentrations, and alleviated airway inflammation manifested by increased macrophage and decreased eosinophil and neutrophil counts in BALF, as well as airway remodeling and suppressed peribronchial cellular infiltration. Moreover, oral administration of pasteurized yogurt containing heat-killed BBMN68 significantly modulated gut microbiota composition by influencing the proportion of beneficial genera associated with inflammation and immunity, such as Lactobacillus, Candidatus_Saccharimonas, Odoribacter, and Parabacteroides, which also negatively correlated with serum IgE and Th2 cytokine levels. These results demonstrated that pasteurized yogurt containing heat-killed BBMN68 had mitigative effects on allergic airway inflammation, likely through maintaining the systemic Th1/Th2 immune balance by altering the structure and function of the gut microbiota.
Collapse
Affiliation(s)
- Xiaokang Niu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xindi Yin
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Xiuying Wu
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Qi Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yunyun Jiang
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Jingjing He
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yuyang Zhao
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chao Zhang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yimei Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Mengxuan Lai
- Inner Mongolia Mengniu Dairy (Group) Co., Ltd., Hohhot 011500, China
| | - Yue Sang
- Hebei Engineering Research Center of Animal Product, Sanhe 065200, China
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| |
Collapse
|
10
|
Neidhöfer C, Rathore K, Parčina M, Sieber MA. ESKAPEE Pathogen Biofilm Control on Surfaces with Probiotic Lactobacillaceae and Bacillus species. Antibiotics (Basel) 2023; 12:871. [PMID: 37237774 PMCID: PMC10215598 DOI: 10.3390/antibiotics12050871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Combatting the rapidly growing threat of antimicrobial resistance and reducing prevalence and transmission of ESKAPEE pathogens in healthcare settings requires innovative strategies, one of which is displacing these pathogens using beneficial microorganisms. Our review comprehensively examines the evidence of probiotic bacteria displacing ESKAPEE pathogens, with a focus on inanimate surfaces. A systematic search was conducted using the PubMed and Web of Science databases on 21 December 2021, and 143 studies were identified examining the effects of Lactobacillaceae and Bacillus spp. cells and products on the growth, colonization, and survival of ESKAPEE pathogens. While the diversity of study methods limits evidence analysis, results presented by narrative synthesis demonstrate that several species have the potential as cells or their products or supernatants to displace nosocomial infection-causing organisms in a variety of in vitro and in vivo settings. Our review aims to aid the development of new promising approaches to control pathogen biofilms in medical settings by informing researchers and policymakers about the potential of probiotics to combat nosocomial infections. More targeted studies are needed to assess safety and efficacy of different probiotic formulations, followed by large-scale studies to assess utility in infection control and medical practice.
Collapse
Affiliation(s)
- Claudio Neidhöfer
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Kamni Rathore
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, 53757 Sankt Augustin, Germany
| | - Marijo Parčina
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Martin A. Sieber
- Institute for Functional Gene Analytics, Bonn-Rhein-Sieg University of Applied Sciences, 53757 Sankt Augustin, Germany
| |
Collapse
|
11
|
Assandri MH, Malamud M, Trejo FM, Serradell MDLA. S-layer proteins as immune players: tales from pathogenic and non-pathogenic bacteria. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 4:100187. [PMID: 37064268 PMCID: PMC10102220 DOI: 10.1016/j.crmicr.2023.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
In bacteria, as in other microorganisms, surface compounds interact with different pattern recognition receptors expressed by host cells, which usually triggers a variety of cellular responses that result in immunomodulation. The S-layer is a two-dimensional macromolecular crystalline structure formed by (glyco)-protein subunits that covers the surface of many species of Bacteria and almost all Archaea. In Bacteria, the presence of S-layer has been described in both pathogenic and non-pathogenic strains. As surface components, special attention deserves the role that S-layer proteins (SLPs) play in the interaction of bacterial cells with humoral and cellular components of the immune system. In this sense, some differences can be predicted between pathogenic and non-pathogenic bacteria. In the first group, the S-layer constitutes an important virulence factor, which in turn makes it a potential therapeutic target. For the other group, the growing interest to understand the mechanisms of action of commensal microbiota and probiotic strains has prompted the studies of the role of the S-layer in the interaction between the host immune cells and bacteria bearing this surface structure. In this review, we aim to summarize the main latest reports and the perspectives of bacterial SLPs as immune players, focusing on those from pathogenic and commensal/probiotic most studied species.
Collapse
|
12
|
Mirzabekyan S, Harutyunyan N, Manvelyan A, Malkhasyan L, Balayan M, Miralimova S, Chikindas ML, Chistyakov V, Pepoyan A. Fish Probiotics: Cell Surface Properties of Fish Intestinal Lactobacilli and Escherichia coli. Microorganisms 2023; 11:microorganisms11030595. [PMID: 36985169 PMCID: PMC10052099 DOI: 10.3390/microorganisms11030595] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/19/2023] [Accepted: 02/20/2023] [Indexed: 03/03/2023] Open
Abstract
The properties of intestinal bacteria/probiotics, such as cell surface hydrophobicity (CSH), auto-aggregation, and biofilm formation ability, play an important role in shaping the relationship between the bacteria and the host. The current study aimed to investigate the cell surface properties of fish intestinal bacteria and probiotics. Microbial adhesion to hydrocarbons was tested according to Kos and coauthors. The aggregation abilities of the investigated strains were studied as described by Collado and coauthors. The ability of bacterial isolates to form a biofilm was determined by performing a qualitative analysis using crystal violet staining based on the attachment of bacteria to polystyrene. These studies prove that bacterial cell surface hydrophobicity (CSH) is associated with the growth medium, and the effect of the growth medium on CSH is species-specific and likely also strain-specific. Isolates of intestinal lactobacilli from fish (Salmo ischchan) differed from isolates of non-fish/shrimp origin in the relationship between auto-aggregation and biofilm formation. Average CSH levels for fish lactobacilli and E. coli might were lower compared to those of non-fish origin, which may affect the efficiency of non-fish probiotics use in fisheries due to the peculiarities of the hosts’ aquatic lifestyles.
Collapse
Affiliation(s)
- Susanna Mirzabekyan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
| | - Natalya Harutyunyan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
| | - Anahit Manvelyan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
| | - Lilit Malkhasyan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
| | - Marine Balayan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
| | - Shakhlo Miralimova
- Institute of Microbiology, Academy of Sciences of the Republic of Uzbekistan, Tashkent 100125, Uzbekistan
| | - Michael L. Chikindas
- Health Promoting Natural Laboratory, Rutgers State University, New Brunswick, NJ 08901, USA
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Bolshaya Pirogovskaya Str., 19/1, 119146 Moscow, Russia
| | - Vladimir Chistyakov
- Center for Agrobiotechnology, Don State Technical University, 344002 Rostov-on-Don, Russia
- D.I. Ivanovsky Academy of Biology and Biotechnology, Southern Federal University, Prosp. Stachky 194/1, 344090 Rostov-on-Don, Russia
| | - Astghik Pepoyan
- Division of Food Safety and Biotechnology, Armenian National Agrarian University, Yerevan 0009, Armenia
- The International Scientific-Educational Center of the National Academy of Sciences of the Republic of Armenia, Yerevan 0019, Armenia
- Correspondence: or or ; Tel.: +374-91-432490
| |
Collapse
|
13
|
Helmy YA, Taha-Abdelaziz K, Hawwas HAEH, Ghosh S, AlKafaas SS, Moawad MMM, Saied EM, Kassem II, Mawad AMM. Antimicrobial Resistance and Recent Alternatives to Antibiotics for the Control of Bacterial Pathogens with an Emphasis on Foodborne Pathogens. Antibiotics (Basel) 2023; 12:274. [PMID: 36830185 PMCID: PMC9952301 DOI: 10.3390/antibiotics12020274] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/21/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Antimicrobial resistance (AMR) is one of the most important global public health problems. The imprudent use of antibiotics in humans and animals has resulted in the emergence of antibiotic-resistant bacteria. The dissemination of these strains and their resistant determinants could endanger antibiotic efficacy. Therefore, there is an urgent need to identify and develop novel strategies to combat antibiotic resistance. This review provides insights into the evolution and the mechanisms of AMR. Additionally, it discusses alternative approaches that might be used to control AMR, including probiotics, prebiotics, antimicrobial peptides, small molecules, organic acids, essential oils, bacteriophage, fecal transplants, and nanoparticles.
Collapse
Affiliation(s)
- Yosra A. Helmy
- Department of Veterinary Science, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Khaled Taha-Abdelaziz
- Department of Animal and Veterinary Sciences, Clemson University, Clemson, SC 29634, USA
| | - Hanan Abd El-Halim Hawwas
- Department of Zoonoses, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Soumya Ghosh
- Department of Genetics, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Samar Sami AlKafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31511, Egypt
| | | | - Essa M. Saied
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, 12489 Berlin, Germany
| | - Issmat I. Kassem
- Centre for Food Safety, Department of Food Science and Technology, University of Georgia, Griffin, GA 30609, USA
| | - Asmaa M. M. Mawad
- Department of Biology, College of Science, Taibah University, Madinah 42317, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
14
|
Yadav AK, Varikuti SR, Kumar A, Kumar M, Debanth N, Rajkumar H. Expression of heterologous heparan sulphate binding protein of Helicobacter pylori on the surface of Lactobacillus rhamnosus GG. 3 Biotech 2023; 13:19. [PMID: 36568501 PMCID: PMC9768065 DOI: 10.1007/s13205-022-03428-4] [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: 05/16/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Helicobacter pylori (H. pylori) is one of most commonly found pathogen in the stomach. In spite of emergence of different treatment strategies, H. pylori infection remains difficult to treat. The bioengineered probiotic lactobacilli that could displace H. pylori and simultaneously present immunogenic peptides such as heparan sulphate binding protein (Hsbp) to elicit immune response could emerge as a potential therapeutic agent. The aim of this study was to discover the anti-H. pylori activities and faster exclusion of H. pylori from host cells by the recombinant strain of Lactobacillus expressing the immunogenic Hsbp protein. The results were promising and showed a 65% reduction in H. pylori adhesion after two hours of pre-incubation with recombinant-LGG and HeLa S3 cells, followed by the adhesion of H. pylori pathogen (P < 0.002). Additionally, 36% and 39% reduction were examined in co-incubation and post-incubation with recombinant-LGG, respectively. When challenged with H. pylori, the proinflammatory cytokine expression was also down regulated in recombinant-LGG treated HeLa S3 cells. This promising result provides a new insight of bioengineered probiotic lactobacilli which could displace H. pylori and simultaneously has immunogenic properties thereby may be useful to prevent H. pylori infection. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03428-4.
Collapse
Affiliation(s)
- Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Distt., Samba, 181143 Jammu and Kashmir India
- Department of Microbiology and Immunology, ICMR-National Institute of Nutrition, Hyderabad, 500007 Telangana India
| | - Sudarshan Reddy Varikuti
- Department of Microbiology and Immunology, ICMR-National Institute of Nutrition, Hyderabad, 500007 Telangana India
| | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendergarh, Jant-Pali, 123031 Haryana India
| | - Manoj Kumar
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Nabendu Debanth
- Centre for Molecular Biology, Central University of Jammu, Distt., Samba, 181143 Jammu and Kashmir India
| | - Hemalatha Rajkumar
- Department of Microbiology and Immunology, ICMR-National Institute of Nutrition, Hyderabad, 500007 Telangana India
| |
Collapse
|
15
|
Updates on the Role of Probiotics against Different Health Issues: Focus on Lactobacillus. Int J Mol Sci 2022; 24:ijms24010142. [PMID: 36613586 PMCID: PMC9820606 DOI: 10.3390/ijms24010142] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
This review article is built on the beneficial effects of Lactobacillus against different diseases, and a special focus has been made on its effects against neurological disorders, such as depression, multiple sclerosis, Alzheimer's, and Parkinson's disease. Probiotics are live microbes, which are found in fermented foods, beverages, and cultured milk and, when administered in an adequate dose, confer health benefits to the host. They are known as "health-friendly bacteria", normally residing in the human gut and involved in maintaining homeostatic conditions. Imbalance in gut microbiota results in the pathophysiology of several diseases entailing the GIT tract, skin, immune system, inflammation, and gut-brain axis. Recently, the use of probiotics has gained tremendous interest, because of their profound effects on the management of these disease conditions. Recent findings suggest that probiotics enrichment in different human and mouse disease models showed promising beneficial effects and results in the amelioration of disease symptoms. Thus, this review focuses on the current probiotics-based products, different disease models, variable markers measured during trials, and evidence obtained from past studies on the use of probiotics in the prevention and treatment of different diseases, covering the skin to the central nervous system diseases.
Collapse
|
16
|
Lactococcus lactis' Effect on the Intestinal Microbiota of Streptococcus agalactiae-Infected Zebrafish (Danio rerio). Microbiol Spectr 2022; 10:e0112822. [PMID: 36214699 PMCID: PMC9604069 DOI: 10.1128/spectrum.01128-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Streptococcus agalactiae is a common pathogen in aquaculture that disrupts the balance of the intestinal microbiota and threatens fish health, causing enormous losses to the aquaculture industry. In this study, we isolated and screened a Lactococcus lactis KUST48 (LLK48) strain with antibacterial effect against S. agalactiae in vitro and used it as a potential probiotic to explore its therapeutic effect on zebrafish (Danio rerio) infected with S. agalactiae. This study divided zebrafish into 3 groups: control group, injected with phosphate-buffered saline; infection group, injected with S. agalactiae; and treatment group, treated with LLK48 after S. agalactiae injection. Then, the 16S rRNA gene sequences of the intestinal microbiota of these 3 groups were sequenced using Illumina high-throughput sequencing technology. The results showed that the relative abundance of intestinal bacteria was significantly decreased in the infection group, and a high relative abundance of S. agalactiae was observed. The relative abundance of the intestinal microbiota was increased in the treatment group, with a decrease in the relative abundance of S. agalactiae compared to that in the control group. In the Cluster of Orthologous Groups of proteins function classification, the relative abundance of each biological function in the infection group was significantly lower than that of the control and treatment groups, showing that LLK48 has a positive therapeutic effect on zebrafish infected with S. agalactiae. This study provides a foundation for exploring the pathogenic mechanism of S. agalactiae on fish and their intestinal symbionts, and also presents a new approach for the treatment of S. agalactiae infections in fish aquaculture systems. IMPORTANCE L. lactis KUST48 (LLK48) with a bacteriostatic effect against S. agalactiae was isolated from tilapia intestinal tracts. S. agalactiae infection significantly reduced the relative abundance of intestinal bacteria and various physiological functions in zebrafish intestines. LLK48 demonstrated infection and subsequent therapeutic effects on the S. agalactiae infection in the zebrafish intestine. Therefore, the potential probiotic LLK48 can be considered as a therapeutic treatment for S. agalactiae infections in aquaculture, which can reduce the use of antibiotics and help maintain fish health.
Collapse
|
17
|
Xu X, Duarte ME, Kim SW. Postbiotic effects of Lactobacillus fermentate on intestinal health, mucosa-associated microbiota, and growth efficiency of nursery pigs challenged with F18+Escherichia coli. J Anim Sci 2022; 100:6603433. [PMID: 35666999 PMCID: PMC9387594 DOI: 10.1093/jas/skac210] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
This study determined the supplemental effects of Lactobacillus fermentate (LBF, Adare Biome, France) on intestinal health and prevention of postweaning diarrhea caused by F18+Escherichia coli in nursery pigs. Sixty-four weaned pigs (6.6 ± 0.7 kg body weight) were allotted in a randomized complete block design to four treatments: NC: no challenge/no supplement; PC: E. coli challenge/no supplement; AGP: E. coli challenge/bacitracin (30 g/t feed); and PBT: E. coli challenge/LBF (2 kg/t feed). Bacitracin methylene disalicylate (BMD) was used as a source of bacitracin. On day 7, challenged groups were orally inoculated with F18+E. coli (2.4 × 1010 CFU), whereas NC received sterile saline solution. Growth performance was analyzed weekly, and pigs were euthanized at the end of 28 d feeding to analyze intestinal health. Data were analyzed using the Mixed procedure of SAS 9.4. During the post-challenge period, PC tended to decrease (P = 0.067) average daily gain (ADG) when compared with NC, whereas AGP increased (P < 0.05) when compared with PC; PBT tended to increase (P = 0.081) ADG when compared with PC. The PC increased fecal score (P < 0.05) during day 7 to 14 when compared with NC, whereas AGP decreased it (P < 0.05) during day 14 to 21 when compared with PC. The PC increased (P < 0.05) protein carbonyl, crypt cell proliferation, and the relative abundance of Helicobacter rodentium when compared with NC. However, AGP decreased (P < 0.05) crypt cell proliferation and H. rodentium and increased (P < 0.05) villus height, Bifidobacterium boum, Pelomonas spp., and Microbacterium ginsengisoli when compared with PC. The PBT reduced (P < 0.05) crypt cell proliferation and H. rodentium and increased (P < 0.05) Lactobacillus salivarius and Propionibacterium acnes when compared with PC. At the genus level, AGP and PBT increased (P < 0.05) the alpha diversity of jejunal mucosa-associated microbiota in pigs estimated with Chao1 richness estimator when compared with PC. Collectively, F18+E. coli reduced growth performance by adversely affecting microbiota and intestinal health. The LBF and BMD improved growth performance, and it was related to the enhanced intestinal health and increased diversity and abundance of beneficial microbiota in pigs challenged with F18+E. coli.
Collapse
Affiliation(s)
- Xiangyi Xu
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review updates recent findings about Escherichia coli O157:H7 virulence factors and its bovine reservoir. This Shiga toxin (Stx)-producing E. coli belongs to the Enterohemorrhagic E. coli (EHEC) pathotype causing hemorrhagic colitis. Its low infectious dose makes it an efficient, severe, foodborne pathogen. Although EHEC remains in the intestine, Stx can translocate systemically and is cytotoxic to microvascular endothelial cells, especially in the kidney and brain. Disease can progress to life-threatening hemolytic uremic syndrome (HUS) with hemolytic anemia, acute kidney failure, and thrombocytopenia. Young children, the immunocompromised, and the elderly are at the highest risk for HUS. Healthy ruminants are the major reservoir of EHEC and cattle are the primary source of human exposure. RECENT FINDINGS Advances in understanding E. coli O157:H7 pathogenesis include molecular mechanisms of virulence, bacterial adherence, type three secretion effectors, intestinal microbiome, inflammation, and reservoir maintenance. SUMMARY Many aspects of E. coli O157:H7 disease remain unclear and include the role of the human and bovine intestinal microbiomes in infection. Therapeutic strategies involve controlling inflammatory responses and/or intestinal barrier function. Finally, elimination/reduction of E. coli O157:H7 in cattle using CRISPR-engineered conjugative bacterial plasmids and/or on-farm management likely hold solutions to reduce infections and increase food safety/security.
Collapse
|
19
|
Sadiq FA, Hansen MF, Burmølle M, Heyndrickx M, Flint S, Lu W, Chen W, Zhang H. Towards understanding mechanisms and functional consequences of bacterial interactions with members of various kingdoms in complex biofilms that abound in nature. FEMS Microbiol Rev 2022; 46:6595875. [PMID: 35640890 DOI: 10.1093/femsre/fuac024] [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: 12/08/2021] [Revised: 04/11/2022] [Accepted: 05/27/2022] [Indexed: 11/12/2022] Open
Abstract
The microbial world represents a phenomenal diversity of microorganisms from different kingdoms of life which occupy an impressive set of ecological niches. Most, if not all, microorganisms once colonise a surface develop architecturally complex surface-adhered communities which we refer to as biofilms. They are embedded in polymeric structural scaffolds serve as a dynamic milieu for intercellular communication through physical and chemical signalling. Deciphering microbial ecology of biofilms in various natural or engineered settings has revealed co-existence of microorganisms from all domains of life, including Bacteria, Archaea and Eukarya. The coexistence of these dynamic microbes is not arbitrary, as a highly coordinated architectural setup and physiological complexity show ecological interdependence and myriads of underlying interactions. In this review, we describe how species from different kingdoms interact in biofilms and discuss the functional consequences of such interactions. We highlight metabolic advances of collaboration among species from different kingdoms, and advocate that these interactions are of great importance and need to be addressed in future research. Since trans-kingdom biofilms impact diverse contexts, ranging from complicated infections to efficient growth of plants, future knowledge within this field will be beneficial for medical microbiology, biotechnology, and our general understanding of microbial life in nature.
Collapse
Affiliation(s)
- Faizan Ahmed Sadiq
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium
| | - Mads Frederik Hansen
- Section of Microbiology, Department of Biology, University of Copenhagen, Denmark
| | - Mette Burmølle
- Section of Microbiology, Department of Biology, University of Copenhagen, Denmark
| | - Marc Heyndrickx
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium.,Department of Pathology, Bacteriology and Poultry Diseases, Ghent University, Merelbeke, Belgium
| | - Steve Flint
- School of Food and Advanced Technology, Massey University, Private Bag, 11222, Palmerston North, New Zealand
| | - Wenwei Lu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
20
|
Petrova P, Arsov A, Tsvetanova F, Parvanova-Mancheva T, Vasileva E, Tsigoriyna L, Petrov K. The Complex Role of Lactic Acid Bacteria in Food Detoxification. Nutrients 2022; 14:2038. [PMID: 35631179 PMCID: PMC9147554 DOI: 10.3390/nu14102038] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
Toxic ingredients in food can lead to serious food-related diseases. Such compounds are bacterial toxins (Shiga-toxin, listeriolysin, Botulinum toxin), mycotoxins (aflatoxin, ochratoxin, zearalenone, fumonisin), pesticides of different classes (organochlorine, organophosphate, synthetic pyrethroids), heavy metals, and natural antinutrients such as phytates, oxalates, and cyanide-generating glycosides. The generally regarded safe (GRAS) status and long history of lactic acid bacteria (LAB) as essential ingredients of fermented foods and probiotics make them a major biological tool against a great variety of food-related toxins. This state-of-the-art review aims to summarize and discuss the data revealing the involvement of LAB in the detoxification of foods from hazardous agents of microbial and chemical nature. It is focused on the specific properties that allow LAB to counteract toxins and destroy them, as well as on the mechanisms of microbial antagonism toward toxigenic producers. Toxins of microbial origin are either adsorbed or degraded, toxic chemicals are hydrolyzed and then used as a carbon source, while heavy metals are bound and accumulated. Based on these comprehensive data, the prospects for developing new combinations of probiotic starters for food detoxification are considered.
Collapse
Affiliation(s)
- Penka Petrova
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Alexander Arsov
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (P.P.); (A.A.)
| | - Flora Tsvetanova
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Tsvetomila Parvanova-Mancheva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Evgenia Vasileva
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Lidia Tsigoriyna
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| | - Kaloyan Petrov
- Institute of Chemical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (F.T.); (T.P.-M.); (E.V.); (L.T.)
| |
Collapse
|
21
|
Du Y, Li H, Shao J, Wu T, Xu W, Hu X, Chen J. Adhesion and Colonization of the Probiotic Lactobacillus plantarum HC-2 in the Intestine of Litopenaeus Vannamei Are Associated With Bacterial Surface Proteins. Front Microbiol 2022; 13:878874. [PMID: 35535252 PMCID: PMC9076606 DOI: 10.3389/fmicb.2022.878874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Surface proteins are a type of proteins expressed on the surface of bacteria that play an important role in cell wall synthesis, maintenance of cell morphology, and signaling with the host. Our previous study showed that the probiotic Lactobacillus plantarum HC-2 improved the growth performance and immune response of Litopenaeus vannamei. To further investigate the probiotic mechanism, we determined the automatic aggregation ability of the bacteria and surface hydrophobicity of HC-2 after being treated with 5 M of lithium chloride (LiCl) and observed the morphology and adhesion of the bacteria to HCT116 cells. The results showed that with the removal of the HC-2 surface protein, the auto-aggregation ability and surface hydrophobicity of HC-2 decreased, and the crude mucus layer coated on the bacterial surface gradually dissociated. The adhesion rate of HC-2 to HCT116 cells decreased from 98.1 to 20.9%. Moreover, a total of 201 unique proteins were identified from the mixture of the surface proteins by mass spectrometry (MS). Several proteins are involved in transcription and translation, biosynthetic or metabolic process, cell cycle or division, cell wall synthesis, and emergency response. Meanwhile, a quantitative real-time PCR qPCR_ showed that HC-2 was mainly colonized in the midgut of shrimp, and the colonization numbers were 15 times higher than that in the foregut, while the colonization rate in the hindgut was lower. The adhesion activity measurement showed that the adhesion level of HC-2 to crude intestinal mucus of L. vannamei was higher than that of bovine serum albumin (BSA) and collagen, and the adhesion capacity of the bacterial cells decreased with the extension of LiCl-treatment time. Finally, we identified the elongation factor Tu, Type I glyceraldehyde-3-phosphate dehydrogenase, small heat shock protein, and 30S ribosomal protein from the surface proteins, which may be the adhesion proteins of HC-2 colonization in the shrimp intestine. The above results indicate that surface proteins play an important role in maintaining the cell structure stability and cell adhesion. Surface proteomics analysis contributes to describing potential protein-mediated probiotic-host interactions. The identification of some interacting proteins in this work may be beneficial to further understand the adhesion/colonization mechanism and probiotic properties of L. plantarum HC-2 in the shrimp intestine.
Collapse
Affiliation(s)
- Yang Du
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China.,Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hao Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China
| | - Jianchun Shao
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ting Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China
| | - WenLong Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China
| | - Xiaoman Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, China.,Key Laboratory of Aquacultural Biotechnology of Ministry of Education, Ningbo University, Ningbo, China
| |
Collapse
|
22
|
Xu H, Lao L, Ji C, Lu Q, Guo Y, Pan D, Wu Z. Anti-inflammation and adhesion enhancement properties of the multifunctional LPxTG-motif surface protein derived from the Lactobacillus reuteri DSM 8533. Mol Immunol 2022; 146:38-45. [PMID: 35421739 DOI: 10.1016/j.molimm.2022.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
LPxTG-motif protein (LMP) is one kind of a precursor protein that contains a conserved LPxTG-motif at the C-terminus, which can be recognized by sortase A (SrtA) and covalently bind to the bacterial peptidoglycan. In this study, LMP derived from Lactobacillus reuteri (L. reuteri) was heterologous expressed and the tolerance and intestinal colonization ability of the LMP on L. reuteri were analyzed in simulated gastrointestinal fluid. Meanwhile, the anti-inflammatory activity of LMP was also evaluated in the LPS-stimulated RAW 264.7 cell model. The results indicated that LMP can promote the intestinal survival rate and adhesion characteristics of L. reuteri and enhanced the autoinducer-2 (AI-2) signaling molecule of the Lactobacillus strains in quorum sensing. Furthermore, LMP can inhibit the expressions of inflammatory cytokine TNF-α and IL-1β via ERK-JNK related MAPK signaling cascades. These findings provide a better understanding of the multifunctional LPxTG-motif surface protein derived from L. reuteri in the gastrointestinal tract environment.
Collapse
Affiliation(s)
- Hai Xu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lifeng Lao
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chunyu Ji
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Qianqian Lu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Yuxing Guo
- School of Food Science & Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zhen Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
23
|
Reuben RC, Elghandour MMMY, Alqaisi O, Cone JW, Márquez O, Salem AZM. Influence of microbial probiotics on ruminant health and nutrition: sources, mode of action and implications. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:1319-1340. [PMID: 34775613 DOI: 10.1002/jsfa.11643] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 10/21/2021] [Accepted: 11/14/2021] [Indexed: 06/13/2023]
Abstract
Globally, ruminant production contributes immensely to the supply of the highest quality and quantity of proteins for human consumption, sustenance of livelihoods, and attainment of food security. Nevertheless, the phasing out of antibiotics in animal production has posed a myriad of challenges, including poor growth, performance and nutrient utilization, pathogen colonization, dysbiosis, and food safety issues in ruminants. Probiotics (direct-fed microbials), comprising live microbial strains that confer health and nutritional benefits to the host when administered in appropriate quantities, are emerging as a viable, safe, natural and sustainable alternative to antibiotics. Although the mechanisms of action exerted by probiotics on ruminants are not well elucidated, dietary probiotic dosage to ruminants enhances development and maturation, growth and performance, milk production and composition, nutrient digestibility, feed efficiency, pathogen reduction, and mitigation of gastrointestinal diseases. However, the beneficial response to probiotic supplementation in ruminants is not consistent, being dependent on the microbial strain selected, combination of strains, dose, time and frequency of supplementation, diet, animal breed, physiological stage, husbandry practice, and farm management. Nonetheless, several studies have recently reported beneficial effects of probiotics on ruminant performance, health and production. This review conclusively re-iterates the need for probiotics inclusion for the sustainability of ruminant production. Considering the role that ruminants play in food production and employment, global acceptance of sustainable ruminant production through supplementation with probiotics will undoubtedly ensure food security and food safety for the world. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Rine C Reuben
- German Centre for Integrative Biodiversity Research (iDiv), Halle-Jena-Leipzig, Leipzig University, Leipzig, Germany
| | - Mona M M Y Elghandour
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Othman Alqaisi
- Animal and Veterinary Sciences Department, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
| | - John W Cone
- Animal Nutrition Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Ofelia Márquez
- Centro Universitario UAEM Amecameca, Universidad Autónoma del Estado de México, Amecameca, Mexico
| | - Abdelfattah Z M Salem
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, Mexico
| |
Collapse
|
24
|
Zhang WX, Xiao CL, Li SY, Bai XC, Qi H, Tian H, Wang N, Yang B, Li XM, Sun Y. Streptococcus strain C17 T as a potential probiotic candidate to modulate oral health. Lett Appl Microbiol 2022; 74:901-908. [PMID: 35218013 DOI: 10.1111/lam.13680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
In the microbiome, probiotics modulate oral diseases. In this study, Streptococcus strain C17T was isolated from the oropharynx of a five-year-old healthy child, and its potential probiotic properties were analysed using human bronchial epithelial cells (16-HBE) used as an in vitro oropharyngeal mucosal model. The results demonstrated that the C17T strain showed tolerance to moderate pH ranges of 4-5 and 0.5-1% bile. However, it was more tolerant to 0.5% bile than 1% bile. It also demonstrated an ability to accommodate maladaptive oropharyngeal conditions (i.e., tolerating lysozyme at 200 μg mL-1) . It was also resistant to hydrogen peroxide at 0.8 mM . In addition, we found out that the strain possesses inhibitory activities against various common pathogenic bacteria. Furthermore, C17T was not cytotoxic to 16-HBE cells at different multiplicities of infection. Scanning electron microscopy disclosed that C17T adhesion to 16-HBE cells. Competition, exclusion, and displacement assays showed that it had good anti-adhesive effect against S. aureus. The present study revealed that Streptococcus strain C17T is a potentially efficacious oropharyngeal probiotic.
Collapse
Affiliation(s)
- Wen Xiao Zhang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Chun Ling Xiao
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Shu Yin Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Xiao Cui Bai
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - He Qi
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Han Tian
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Nan Wang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Biao Yang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Xin Ming Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Ye Sun
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| |
Collapse
|
25
|
Kober AKMH, Riaz Rajoka MS, Mehwish HM, Villena J, Kitazawa H. Immunomodulation Potential of Probiotics: A Novel Strategy for Improving Livestock Health, Immunity, and Productivity. Microorganisms 2022; 10:microorganisms10020388. [PMID: 35208843 PMCID: PMC8878146 DOI: 10.3390/microorganisms10020388] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the use of probiotics as feed supplements in animal production has increased considerably due to the ban on antibiotic growth promoters in livestock. This review provides an overview of the current situation, limitation, and prospects for probiotic formulations applied to livestock. Recently, the use of probiotics in livestock has been suggested to significantly improve their health, immunity, growth performance, nutritional digestibility, and intestinal microbial balance. Furthermore, it was reported that the use of probiotics in animals was helpful in equilibrating their beneficial microbial population and microbial turnover via stimulating the host immune response through specific secretions and competitive exclusion of potentially pathogenic bacteria in the digestive tract. Recently, there has been great interest in the understanding of probiotics targeted diet and its ability to compete with harmful microbes and acquire their niches. Therefore, the present review explores the most commonly used probiotic formulations in livestock feed and their effect on animal health. In summary, this article provides an in-depth knowledge about the formulation of probiotics as a step toward a better alternative to antibiotic healthy growth strategies.
Collapse
Affiliation(s)
- A. K. M. Humayun Kober
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Department of Dairy and Poultry Science, Chittagong Veterinary and Animal Sciences University, Khulshi, Chittagong 4225, Bangladesh
- Correspondence: or (A.K.M.H.K.); (H.K.); Tel.: +880-1712-164794 (A.K.M.H.K.); +81-22-757-4372 (H.K.)
| | - Muhammad Shahid Riaz Rajoka
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Hafiza Mahreen Mehwish
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman 4000, Argentina;
| | - Haruki Kitazawa
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan; (M.S.R.R.); (H.M.M.)
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: or (A.K.M.H.K.); (H.K.); Tel.: +880-1712-164794 (A.K.M.H.K.); +81-22-757-4372 (H.K.)
| |
Collapse
|
26
|
Nogacka AM, Arboleya S, Nikpoor N, Auger J, Salazar N, Cuesta I, Alvarez-Buylla JR, Mantecón L, Solís G, Gueimonde M, Tompkins TA, de los Reyes-Gavilán CG. In Vitro Probiotic Modulation of the Intestinal Microbiota and 2′Fucosyllactose Consumption in Fecal Cultures from Infants at Two Months of Age. Microorganisms 2022; 10:microorganisms10020318. [PMID: 35208773 PMCID: PMC8876326 DOI: 10.3390/microorganisms10020318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
2′-fucosyllactose (2′FL) is one of the most abundant oligosaccharides in human milk, with benefits on neonatal health. Previous results point to the inability of the fecal microbiota from some infants to ferment 2′FL. We evaluated a probiotic formulation, including the strains Lactobacillus helveticus Rosell®-52 (R0052), Bifidobacterium longum subsp. infantis Rosell®-33 (R0033), and Bifidobacterium bifidum Rosell®-71 (R0071), individually or in an 80:10:10 combination on the microbiota and 2′FL degradation. Independent batch fermentations were performed with feces from six full-term infant donors of two months of age (three breastfed and three formula-fed) with added probiotic formulation or the constituent strains in the presence of 2′FL. Microbiota composition was analyzed by 16S rRNA gene sequencing. Gas accumulation, pH decrease and 2′FL consumption, and levels of different metabolites were determined by chromatography. B. bifidum R0071 was the sole microorganism promoting a partial increase of 2′FL degradation during fermentation in fecal cultures of 2′FL slow-degrading donors. However, major changes in microbiota composition and metabolic activity occurred with L. helveticus R0052 or the probiotic formulation in cultures of slow degraders. Further studies are needed to decipher the role of the host intestinal microbiota in the efficacy of these strains.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Naghmeh Nikpoor
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Jeremie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Jorge R. Alvarez-Buylla
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Laura Mantecón
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Gonzalo Solís
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Thomas A. Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| |
Collapse
|
27
|
Bhawal S, Kumari A, Kapila S, Kapila R. Biofunctional Attributes of Surface Layer Protein and Cell-Bound Exopolysaccharide from Probiotic Limosilactobacillus fermentum (MTCC 5898). Probiotics Antimicrob Proteins 2022; 14:360-371. [PMID: 35066819 DOI: 10.1007/s12602-021-09891-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 12/01/2022]
Abstract
The probiotic extracellular matrix components (ECM) have been considered as an important factor in eliciting the beneficial roles of the bacteria. The study involved the growth phase-dependent extraction of the surface layer protein (SLP) and cell-bound exopolysaccharide (EPS-b) from novel Limosilactobacillus fermentum (MTCC 5898). Both SLP and EPS-b were optimally extracted at the late logarithmic phase of the bacteria upon 8 h of incubation. Furthermore, the adhesive, immunomodulatory, and anti-oxidative potential of the extracted components were evaluated using in vitro models. The major role of SLP was evidenced on bacterial adhesion to mucin and was related to its hydrophobic character. Under in vitro conditions, no effect of SLP and EPS-b was observed on the proliferation of murine splenocytes; however, both the components stimulated the phagocytosis of murine peritoneal macrophages at varying concentrations. Furthermore, all the components exhibited strong radical scavenging, chelating, and reducing potential with increasing concentration. Therefore, the ECM components of L. fermentum exhibited a variable biofunctional effect, providing crucial information to enable its further use as functional foods and overcome the challenges posed by probiotics.
Collapse
Affiliation(s)
- Shalaka Bhawal
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Ankita Kumari
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India.
| |
Collapse
|
28
|
Yu Y, Zong M, Lao L, Wen J, Pan D, Wu Z. Adhesion properties of the cell surface proteins in Lactobacillus strains under the GIT environment. Food Funct 2022; 13:3098-3109. [DOI: 10.1039/d1fo04328e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Lactic acid bacteria (LAB) play an essential role in the epithelial barrier and the gut immune system. It can antagonize pathogens by producing antimicrobial substances like bacteriocins, and compete with...
Collapse
|
29
|
Requena T, Pérez Martínez G. Probiotics, Prebiotics, Synbiotics, Postbiotics and Other Biotics. What's Next? COMPREHENSIVE GUT MICROBIOTA 2022:197-210. [DOI: 10.1016/b978-0-12-819265-8.00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Cheng YC, Duarte ME, Kim SW. Nutritional and functional values of lysed Corynebacterium glutamicum cell mass for intestinal health and growth of nursery pigs. J Anim Sci 2021; 99:skab331. [PMID: 34902029 PMCID: PMC8668180 DOI: 10.1093/jas/skab331] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022] Open
Abstract
The objective was to determine the nutritional and functional values of lysed Corynebacterium glutamicum cell mass (CGCM) as a protein supplement and a source of cell wall fragments supporting the growth and intestinal health of nursery pigs. Thirty-two pigs (21 d of age) were allotted to four treatments (n = 8) based on the randomized block design with sex and initial body weight (BW) as blocks. The main effect was the dietary supplementation of lysed CGCM (0, 0.7, 1.4, and 2.1%) replacing blood plasma and fed in two phases (10 and 11 d, respectively). Feed intake and BW were measured at the end of each phase. Pigs were euthanized on day 21 to collect jejunal tissue and mucosa to evaluate intestinal health. Ileal digesta were collected to measure the apparent ileal digestibility of nutrients in diets. Data were analyzed using Proc Mixed and Reg of SAS. Increasing daily intake of CGCM increased (linear; P < 0.05) ADG of pigs. Increasing CGCM supplementation affected (quadratic; P < 0.05) the relative abundance of Lactobacillaceae (minimum: 26.4% at 1.2% CGCM), Helicobacteraceae (maximum: 29.3% at 1.2% CGCM), and Campylobacteraceae (maximum: 9.0% at 1.0% CGCM). Increasing CGCM supplementation affected (quadratic; P < 0.05) the concentrations of immunoglobulin G (maximum: 4.94 µg/mg of protein at 1.0% CGCM) and protein carbonyl (PC; maximum: 6.12 nmol/mg of protein at 1.1% CGCM), whereas linearly decreased (P < 0.05) malondialdehyde (MDA) in the proximal jejunal mucosa. Increasing CGCM supplemention affected (quadratic; P < 0.05) intestinal enterocyte proliferation rate (maximum: 13.3% at 1.0% CGCM), whereas it did not affect intestinal morphology and the nutrient digestibility. In conclusion, supplementing 1.0% to 1.2%, reducing blood plasma supplementation by 0.7% to 0.9%, respectively, increased potential pathogenic microbiota associated in the jejunal mucosa resulting in increased immune response, enterocyte proliferation, and PC concentration. However, supplementing diets with 2.1% CGCM, replacing 1.5% blood plasma, improved growth performance, and reduced MDA without affecting nutrient digestibility, intestinal morphology, and microbiota in the jejunal mucosa. In this study, based on the polynomial contrast, supplementing 1.0% to 1.2% CGCM suppressed the benefits from blood plasma, whereas supplementing 2.1% CGCM showed functional benefits of CGCM with similar effects from blood plasma supplementation.
Collapse
Affiliation(s)
- Yi-Chi Cheng
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
31
|
Abstract
The introduction of next generation sequencing techniques has enabled the characterization of the urinary tract microbiome, which resulted in the rejection of the long-held notion of urinary bladder sterility. Since the discovery and confirmation of the human bladder microbiome, an increasing number of studies have defined this microbial community and understand better its relationship to urinary pathologies. The composition of microbial communities in the urinary tract is linked to a variety of urinary diseases. The purpose of this review is to provide an overview of current information about the urinary microbiome and diseases as well as the development of novel treatment methods.
Collapse
|
32
|
Jones-Freeman B, Chonwerawong M, Marcelino VR, Deshpande AV, Forster SC, Starkey MR. The microbiome and host mucosal interactions in urinary tract diseases. Mucosal Immunol 2021; 14:779-792. [PMID: 33542492 DOI: 10.1038/s41385-020-00372-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
The urinary tract consists of the bladder, ureters, and kidneys, and is an essential organ system for filtration and excretion of waste products and maintaining systemic homeostasis. In this capacity, the urinary tract is impacted by its interactions with other mucosal sites, including the genitourinary and gastrointestinal systems. Each of these sites harbors diverse ecosystems of microbes termed the microbiota, that regulates complex interactions with the local and systemic immune system. It remains unclear whether changes in the microbiota and associated metabolites may be a consequence or a driver of urinary tract diseases. Here, we review the current literature, investigating the impact of the microbiota on the urinary tract in homeostasis and disease including urinary stones, acute kidney injury, chronic kidney disease, and urinary tract infection. We propose new avenues for exploration of the urinary microbiome using emerging technology and discuss the potential of microbiome-based medicine for urinary tract conditions.
Collapse
Affiliation(s)
- Bernadette Jones-Freeman
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Vanessa R Marcelino
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Aniruddh V Deshpande
- Priority Research Centre GrowUpWell, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Department of Pediatric Urology and Surgery, John Hunter Children's Hospital, New Lambton Heights, NSW, Australia.,Urology Unit, Department of Pediatric Surgery, Children's Hospital at Westmead, Sydney Children's Hospital Network, Westmead, NSW, Australia
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Malcolm R Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia. .,Priority Research Centre GrowUpWell, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
33
|
Fan J, Qiu L, Qiao Y, Xue M, Dong X, Meng Z. Recent Advances in Sensing Applications of Molecularly Imprinted Photonic Crystals. Front Chem 2021; 9:665119. [PMID: 34195173 PMCID: PMC8236589 DOI: 10.3389/fchem.2021.665119] [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: 02/07/2021] [Accepted: 03/26/2021] [Indexed: 11/30/2022] Open
Abstract
Photonic crystals (PhCs) with a brightly colored structure are novel materials and are widely used in chemical and biological sensing. Combining PhCs with molecular imprinting technology (MIT), the molecularly imprinted PhC (MIPC) sensors are fabricated, which can specifically recognize the target molecules. Aside from high sensitivity and selectivity, the MIPC sensors could recognize the naked eye detection because of its optical properties. In this review, an overview of recent advances in sensing applications of MIPC sensors including the responsive mechanisms, application in environmental monitoring, and the application to human health were illustrated. The MIPC sensors all responded to the analytes specifically and also showed high sensitivity in real samples, which provided a method to realize the rapid, convenient, naked eye, and real-time detection. Furthermore, the current limitations and potential future directions of MIPC sensors were also discussed.
Collapse
Affiliation(s)
- Jing Fan
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Lili Qiu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Yu Qiao
- School of Design and Arts, Beijing Institute of Technology, Beijing, China
| | - Min Xue
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Xiao Dong
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| | - Zihui Meng
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
34
|
Lee KS, Jeong YJ, Lee MS. Escherichia coli Shiga Toxins and Gut Microbiota Interactions. Toxins (Basel) 2021; 13:toxins13060416. [PMID: 34208170 PMCID: PMC8230793 DOI: 10.3390/toxins13060416] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022] Open
Abstract
Escherichia coli (EHEC) and Shigella dysenteriae serotype 1 are enterohemorrhagic bacteria that induce hemorrhagic colitis. This, in turn, may result in potentially lethal complications, such as hemolytic uremic syndrome (HUS), which is characterized by thrombocytopenia, acute renal failure, and neurological abnormalities. Both species of bacteria produce Shiga toxins (Stxs), a phage-encoded exotoxin inhibiting protein synthesis in host cells that are primarily responsible for bacterial virulence. Although most studies have focused on the pathogenic roles of Stxs as harmful substances capable of inducing cell death and as proinflammatory factors that sensitize the host target organs to damage, less is known about the interface between the commensalism of bacterial communities and the pathogenicity of the toxins. The gut contains more species of bacteria than any other organ, providing pathogenic bacteria that colonize the gut with a greater number of opportunities to encounter other bacterial species. Notably, the presence in the intestines of pathogenic EHEC producing Stxs associated with severe illness may have compounding effects on the diversity of the indigenous bacteria and bacterial communities in the gut. The present review focuses on studies describing the roles of Stxs in the complex interactions between pathogenic Shiga toxin-producing E. coli, the resident microbiome, and host tissues. The determination of these interactions may provide insights into the unresolved issues regarding these pathogens.
Collapse
Affiliation(s)
- Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Correspondence: (Y.-J.J.); (M.-S.L.)
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Daejeon 34141, Korea;
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
- Correspondence: (Y.-J.J.); (M.-S.L.)
| |
Collapse
|
35
|
Nataraj BH, Ramesh C, Mallappa RH. Extractable surface proteins of indigenous probiotic strains confer anti-adhesion knack and protect against methicillin-resistant Staphylococcus aureus induced epithelial hyperpermeability in HT-29 cell line. Microb Pathog 2021; 158:104974. [PMID: 34015494 DOI: 10.1016/j.micpath.2021.104974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 11/28/2022]
Abstract
Probiotic intervention has been long believed to have beneficial effects on human health by curbing the intestinal colonization of pathogens. However, the application of live probiotics therapy may not be an ideal approach to circumvent the infections of superbug origin due to the risk of horizontal antibiotic resistance genes transfer. In this study, the anti-adhesion ability of extractable cell surface proteins from two indigenous potential probiotic strains (Lactiplantibacillus plantarum A5 and Limosilactobacillus fermentum Lf1) and two standard reference strains (Lactobacillus acidophilus NCFM and Lacticaseibacillus rhamnosus LGG) was evaluated against clinical isolates of Methicillin-Resistant Staphylococcus aureus (MRSA) on porcine gastric mucin and HT-29 cells. The surface proteins from the probiotic strains were extracted by treatment with 5 M lithium chloride. The surface protein quantification and SDS-PAGE profiling indicated that the yield and protein patterns were strain-specific. Surface proteins significantly hampered the mucoadhesion of MRSA isolates via protective, competitive, and displacement. Similarly, the treatment with surface proteins probiotic strains displayed anti-adhesion against MRSA isolates on HT-29 cells without affecting the viability of the cell line. Surface proteins treatment to the confluent monolayer of HT-29 cells maintained the epithelial integrity; however, MRSA isolates (109 cells/mL) showed considerable alteration in the epithelial integrity by exacerbating the FITC-dextran transflux. Contrarily, the co-treatment with surface proteins with MRSA isolates significantly lowered the FITC-dextran transflux across the differentiated HT-29 monolayer. Overall, the findings of this study suggest that probiotic-derived surface proteins could be the novel biotherapeutics to combat the MRSA colonization and their concomitant intestinal infections.
Collapse
Affiliation(s)
| | - Chette Ramesh
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| |
Collapse
|
36
|
Abbasi A, Rad AH, Ghasempour Z, Sabahi S, Kafil HS, Hasannezhad P, Rahbar Saadat Y, Shahbazi N. The biological activities of postbiotics in gastrointestinal disorders. Crit Rev Food Sci Nutr 2021; 62:5983-6004. [PMID: 33715539 DOI: 10.1080/10408398.2021.1895061] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
According to outcomes from clinical studies, an intricate relationship occurs between the beneficial microbiota, gut homeostasis, and the host's health status. Numerous studies have confirmed the health-promoting effects of probiotics, particularly in gastrointestinal diseases. On the other hand, the safety issues regarding the consumption of some probiotics are still a matter of debate, thus to overcome the problems related to the application of live probiotic cells in terms of clinical, technological, and economic aspects, microbial-derived biomolecules (postbiotics) were introducing as a potential alternative agent. Presently scientific literature confirms that the postbiotic components can be used as promising tools for both prevention and treatment strategies in gastrointestinal disorders with less undesirable side-effects, particularly in infants and children. Future head-to-head trials are required to distinguish appropriate strains of parent cells, optimal dosages of postbiotics, and assessment of the cost-effectiveness of postbiotics compared to alternative drugs. This review provides an overview of the concept and safety issues regarding postbiotics, with emphasis on their biological role in the treatment of some important gastrointestinal disorders.
Collapse
Affiliation(s)
- Amin Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Ghasempour
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Sabahi
- Department of Nutritional Sciences, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Paniz Hasannezhad
- Department of Medical Engineering Science, University College of Rouzbahan, Sari, Iran
| | - Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nayyer Shahbazi
- Faculty of Agriculture Engineering, Department of Food Science, Shahrood University of Technology, Shahrood, Iran
| |
Collapse
|
37
|
Dong H, Liu B, Li A, Iqbal M, Mehmood K, Jamil T, Chang YF, Zhang H, Wu Q. Microbiome Analysis Reveals the Attenuation Effect of Lactobacillus From Yaks on Diarrhea via Modulation of Gut Microbiota. Front Cell Infect Microbiol 2021; 10:610781. [PMID: 33665171 PMCID: PMC7920975 DOI: 10.3389/fcimb.2020.610781] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Domestic yaks (Bos grunniens) are indigenous to the Tibetan Plateau and display a high diarrhea rate due to poor habitat and husbandry conditions. Lactobacillus has been shown to exert beneficial effects as antimicrobial, growth promotion, and gut microbiota in humans and/or murine models, but the relevant data regarding Lactobacillus isolated from yaks was unavailable. Therefore, this study aimed to investigate the effects of Lactobacillus from yaks on the intestinal microbial community in a mouse model and determine whether Lactobacillus supplementation contributed in alleviating diarrhea by modulating gut microbiota. A total of 12 ileac samples from four groups were collected for 16S rRNA gene amplicon sequencing of V3-V4 region. Results revealed that although Lactobacillus supplementation did not change the diversity of gut microbiota in mice, the proportion of some intestinal microbiota significantly changed. Specifically, the proportion of Lactobacillus and Sphingomonas in the Lactobacillus treated-group (L-group) were increased as compared to control group (C-group), whereas Pantoea, Cutibacterium, Glutamicibacter, Turicibacter, Globicatella, Microbacterium, Facklamia, unidentified_Corynebacteriaceae, Brachybacterium, and Staphylococcus were significantly decreased in the L-group. In contrast, Escherichia coli (E. coli) infection significantly decreased the proportion of beneficial bacteria such as Globicatella, Acinetobacter, Aerococcus, and Comamonas, while loads of pathogenic bacteria significantly increased including Roseburia and Megasphaera. Interestingly, Lactobacillus administration could ameliorate the microbial community structure of E. coli-induced diarrheal mice by reducing the relative abundance of pathogenic bacteria such as Paenibacillus, Aerococcus, Comamonas, Acinetobacter, Corynebacterium, Facklamia, and Globicatella. Results in this study revealed that Lactobacillus supplementation not only improved the gut microbiota but also alleviated diarrhea in mice, which may be mediated by modulating the composition and function of gut microbiota. Moreover, this study is expected to provide a new theoretical basis for the establishment of a preventive and treatment system for diarrhea in yaks.
Collapse
Affiliation(s)
- Hailong Dong
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Bingxian Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mudassar Iqbal
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Khalid Mehmood
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Tariq Jamil
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qingxia Wu
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| |
Collapse
|
38
|
van Zyl WF, Deane SM, Dicks LM. Molecular insights into probiotic mechanisms of action employed against intestinal pathogenic bacteria. Gut Microbes 2020; 12:1831339. [PMID: 33112695 PMCID: PMC7595611 DOI: 10.1080/19490976.2020.1831339] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) diseases, and in particular those caused by bacterial infections, are a major cause of morbidity and mortality worldwide. Treatment is becoming increasingly difficult due to the increase in number of species that have developed resistance to antibiotics. Probiotic lactic acid bacteria (LAB) have considerable potential as alternatives to antibiotics, both in prophylactic and therapeutic applications. Several studies have documented a reduction, or prevention, of GI diseases by probiotic bacteria. Since the activities of probiotic bacteria are closely linked with conditions in the host's GI-tract (GIT) and changes in the population of enteric microorganisms, a deeper understanding of gut-microbial interactions is required in the selection of the most suitable probiotic. This necessitates a deeper understanding of the molecular capabilities of probiotic bacteria. In this review, we explore how probiotic microorganisms interact with enteric pathogens in the GIT. The significance of probiotic colonization and persistence in the GIT is also addressed.
Collapse
Affiliation(s)
- Winschau F. van Zyl
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Shelly M. Deane
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leon M.T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa,CONTACT Leon M.T. Dicks; Department of Microbiology; Stellenbosch University, Stellenbosch7602, South Africa
| |
Collapse
|
39
|
Jessberger N, Dietrich R, Granum PE, Märtlbauer E. The Bacillus cereus Food Infection as Multifactorial Process. Toxins (Basel) 2020; 12:E701. [PMID: 33167492 PMCID: PMC7694497 DOI: 10.3390/toxins12110701] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
The ubiquitous soil bacterium Bacillus cereus presents major challenges to food safety. It is responsible for two types of food poisoning, the emetic form due to food intoxication and the diarrheal form emerging from food infections with enteropathogenic strains, also known as toxico-infections, which are the subject of this review. The diarrheal type of food poisoning emerges after production of enterotoxins by viable bacteria in the human intestine. Basically, the manifestation of the disease is, however, the result of a multifactorial process, including B. cereus prevalence and survival in different foods, survival of the stomach passage, spore germination, motility, adhesion, and finally enterotoxin production in the intestine. Moreover, all of these processes are influenced by the consumed foodstuffs as well as the intestinal microbiota which have, therefore, to be considered for a reliable prediction of the hazardous potential of contaminated foods. Current knowledge regarding these single aspects is summarized in this review aiming for risk-oriented diagnostics for enteropathogenic B. cereus.
Collapse
Affiliation(s)
- Nadja Jessberger
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Schönleutnerstr. 8, 85764 Oberschleißheim, Germany; (R.D.); (E.M.)
| | - Richard Dietrich
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Schönleutnerstr. 8, 85764 Oberschleißheim, Germany; (R.D.); (E.M.)
| | - Per Einar Granum
- Department of Food Safety and Infection Biology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003 NMBU, 1432 Ås, Norway;
| | - Erwin Märtlbauer
- Department of Veterinary Sciences, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Schönleutnerstr. 8, 85764 Oberschleißheim, Germany; (R.D.); (E.M.)
| |
Collapse
|
40
|
Teame T, Wang A, Xie M, Zhang Z, Yang Y, Ding Q, Gao C, Olsen RE, Ran C, Zhou Z. Paraprobiotics and Postbiotics of Probiotic Lactobacilli, Their Positive Effects on the Host and Action Mechanisms: A Review. Front Nutr 2020; 7:570344. [PMID: 33195367 PMCID: PMC7642493 DOI: 10.3389/fnut.2020.570344] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/28/2020] [Indexed: 12/14/2022] Open
Abstract
Lactobacilli comprise an important group of probiotics for both human and animals. The emerging concern regarding safety problems associated with live microbial cells is enhancing the interest in using cell components and metabolites derived from probiotic strains. Here, we define cell structural components and metabolites of probiotic bacteria as paraprobiotics and postbiotics, respectively. Paraprobiotics and postbiotics produced from Lactobacilli consist of a wide range of molecules including peptidoglycans, surface proteins, cell wall polysaccharides, secreted proteins, bacteriocins, and organic acids, which mediate positive effect on the host, such as immunomodulatory, anti-tumor, antimicrobial, and barrier-preservation effects. In this review, we systematically summarize the paraprobiotics and postbiotics derived from Lactobacilli and their beneficial functions. We also discuss the mechanisms underlying their beneficial effects on the host, and their interaction with the host cells. This review may boost our understanding on the benefits and molecular mechanisms associated with paraprobiotics and probiotics from Lactobacilli, which may promote their applications in humans and animals.
Collapse
Affiliation(s)
- Tsegay Teame
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China.,Tigray Agricultural Research Institute, Mekelle, Ethiopia
| | - Anran Wang
- AgricultureIsLife/EnvironmentIsLife and Precision Livestock and Nutrition Unit, AgroBioChem/TERRA, Gembloux Agro-Bio Tech, University of Liege, Passage des Deportes, Gembloux, Belgium
| | - Mingxu Xie
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qianwen Ding
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chenchen Gao
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Rolf Erik Olsen
- Norway-China Fish Gastrointestinal Microbiota Joint Lab, Institute of Biology, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gastrointestinal Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
41
|
Malik I, Batra T, Das S, Kumar V. Light at night affects gut microbial community and negatively impacts host physiology in diurnal animals: Evidence from captive zebra finches. Microbiol Res 2020; 241:126597. [PMID: 32979783 DOI: 10.1016/j.micres.2020.126597] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/02/2020] [Accepted: 09/14/2020] [Indexed: 01/16/2023]
Abstract
The gastrointestinal tract (GIT) hosts a large number of diverse microorganisms, with mutualistic interactions with the host. Here, in two separate experiments, we investigated whether light at night (LAN) would affect GIT microbiota and, in turn, the host physiology in diurnal zebra finches (Taeniopygia guttata). Experiment I assessed the effects of no-night (LL) and dimly illuminated night (dim light at night, dLAN) on fecal microbiota diversity and host physiology of birds born and raised under 12 h photoperiod (LD; 12 h light: 12 h darkness). Under LL and dLAN, compared to LD, we found a significant increase in the body mass, subcutaneous fat deposition and hepatic accumulation of lipids. Although we found no difference in total 24 h food consumption, LL/ dLAN birds ate also at night, suggesting LAN-induced alteration in daily feeding times. Concurrently, there were marked differences in amplicon sequence and bacterial species richness between LD and LAN, with notable decline in Lactobacillus richness in birds under LL and dLAN. We attributed declined Lactobacillus population as causal (at least partially) to negative effects on the host metabolism. Therefore, in experiment II with similar protocol, birds under LL and dLAN were fed on diet with or without Lactobacillus rhamnosus GG (LGG) supplement. Clearly, LGG supplement ameliorated LL- and dLAN-induced negative effects in zebra finches. These results demonstrate adverse effects of unnatural lighting on GIT bacterial diversity and host physiology, and suggest the role of GIT microbiota in the maintenance of metabolic homeostasis in response to LAN environment in diurnal animals.
Collapse
Affiliation(s)
- Indu Malik
- Department of Zoology, University of Delhi, Delhi, 110 007, India
| | - Twinkle Batra
- Department of Zoology, University of Delhi, Delhi, 110 007, India
| | - Subhajit Das
- Department of Zoology, University of Delhi, Delhi, 110 007, India
| | - Vinod Kumar
- Department of Zoology, University of Delhi, Delhi, 110 007, India.
| |
Collapse
|
42
|
Abdelhamid AG, El-Dougdoug NK. Controlling foodborne pathogens with natural antimicrobials by biological control and antivirulence strategies. Heliyon 2020; 6:e05020. [PMID: 32995651 PMCID: PMC7511826 DOI: 10.1016/j.heliyon.2020.e05020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Foodborne diseases represent a global health threat besides the great economic losses encountered by the food industry. These hazards necessitate the implementation of food preservation methods to control foodborne pathogens, the causal agents of human illnesses. Until now, most control methods rely on inhibiting the microbial growth or eliminating the pathogens by applying lethal treatments. Natural antimicrobials, which inhibit microbial growth, include traditional chemicals, naturally occurring antimicrobials, or biological preservation (e.g. beneficial microbes, bacteriocins, or bacteriophages). Although having great antimicrobial effectiveness, challenges due to the adaptation of foodborne pathogens to such control methods are becoming apparent. Such adaptation enables the survival of the pathogens in foods or food-contact environments. This imperative concern inspires contemporary research and food industry sector to develop technologies which do not target microbial growth but disarming microbial virulence factors. These technologies, referred to as "antivirulence", render the microbe non-capable of causing the disease with very limited or no opportunities for the pathogenic microorganisms to develop resistance. For the sake of safer and fresh-like foods, with no effect on the sensory properties of foods, a combination of two or more natural antimicrobials or with other stressors, is now widespread, to preserve foods. This review introduces and critically describes the traditional versus the emerging uses of natural antimicrobials for controlling foodborne pathogens in foods. Development of biological control strategies using natural antimicrobials proved to be effective in inhibiting microbial growth in foods and allowing improved food safety. In the meanwhile, discovery of new antivirulence agents could be a transformative strategy in food preservation in the far future.
Collapse
Affiliation(s)
- Ahmed G. Abdelhamid
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, 13511, Egypt
| | - Noha K. El-Dougdoug
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, 13511, Egypt
| |
Collapse
|
43
|
Nataraj BH, Ali SA, Behare PV, Yadav H. Postbiotics-parabiotics: the new horizons in microbial biotherapy and functional foods. Microb Cell Fact 2020; 19:168. [PMID: 32819443 PMCID: PMC7441679 DOI: 10.1186/s12934-020-01426-w] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/13/2020] [Indexed: 12/20/2022] Open
Abstract
Probiotics have several health benefits by modulating gut microbiome; however, techno-functional limitations such as viability controls have hampered their full potential applications in the food and pharmaceutical sectors. Therefore, the focus is gradually shifting from viable probiotic bacteria towards non-viable paraprobiotics and/or probiotics derived biomolecules, so-called postbiotics. Paraprobiotics and postbiotics are the emerging concepts in the functional foods field because they impart an array of health-promoting properties. Although, these terms are not well defined, however, for time being these terms have been defined as here. The postbiotics are the complex mixture of metabolic products secreted by probiotics in cell-free supernatants such as enzymes, secreted proteins, short chain fatty acids, vitamins, secreted biosurfactants, amino acids, peptides, organic acids, etc. While, the paraprobiotics are the inactivated microbial cells of probiotics (intact or ruptured containing cell components such as peptidoglycans, teichoic acids, surface proteins, etc.) or crude cell extracts (i.e. with complex chemical composition)". However, in many instances postbiotics have been used for whole category of postbiotics and parabiotics. These elicit several advantages over probiotics like; (i) availability in their pure form, (ii) ease in production and storage, (iii) availability of production process for industrial-scale-up, (iv) specific mechanism of action, (v) better accessibility of Microbes Associated Molecular Pattern (MAMP) during recognition and interaction with Pattern Recognition Receptors (PRR) and (vi) more likely to trigger only the targeted responses by specific ligand-receptor interactions. The current review comprehensively summarizes and discussed various methodologies implied to extract, purify, and identification of paraprobiotic and postbiotic compounds and their potential health benefits.
Collapse
Affiliation(s)
- Basavaprabhu H Nataraj
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Syed Azmal Ali
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Pradip V Behare
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine and Microbiology and Immunology, Wake Forest School of Medicine, Biotech Place, Room 2E-034, 575 North Patterson Ave, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
44
|
Lactobacillus johnsonii L531 Ameliorates Escherichia coli-Induced Cell Damage via Inhibiting NLRP3 Inflammasome Activity and Promoting ATG5/ATG16L1-Mediated Autophagy in Porcine Mammary Epithelial Cells. Vet Sci 2020; 7:vetsci7030112. [PMID: 32823867 PMCID: PMC7558184 DOI: 10.3390/vetsci7030112] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 12/31/2022] Open
Abstract
Escherichia coli (E. coli), a main mastitis-causing pathogen in sows, leads to mammary tissue damage. Here, we explored the effects of Lactobacillus johnsonii L531 on attenuating E. coli-induced inflammatory damage in porcine mammary epithelial cells (PMECs). L. johnsonii L531 pretreatment reduced E. coli adhesion to PMECs by competitive exclusion and the production of inhibitory factors and decreased E. coli-induced destruction of cellular morphology and ultrastructure. E. coli induced activation of NLRP3 inflammasome associated with increased expression of NLRP3, ASC, and cleaved caspase-1, however, L. johnsonii L531 inhibited E. coli-induced activation of NLRP3 inflammasome. Up-regulation of interleukin (Il)-1β, Il-6, Il-8, Il-18, tumor necrosis factor alpha, and chemokine Cxcl2 expression after E. coli infection was attenuated by L. johnsonii L531. E. coli infection inhibited autophagy, whereas L. johnsonii L531 reversed the inhibitory effect of E. coli on autophagy by decreasing the expression of autophagic receptor SQSTM1/p62 and increasing the expression of autophagy-related proteins ATG5, ATG16L1, and light chain 3 protein by Western blotting analysis. Our findings suggest that L. johnsonii L531 pretreatment restricts NLRP3 inflammasome activity and induces autophagy through promoting ATG5/ATG16L1-mediated autophagy, thereby protecting against E. coli-induced inflammation and cell damage in PMECs.
Collapse
|
45
|
Qiao J, Sun Z, Liang D, Li H. Lactobacillus salivarius alleviates inflammation via NF-κB signaling in ETEC K88-induced IPEC-J2 cells. J Anim Sci Biotechnol 2020; 11:76. [PMID: 32774852 PMCID: PMC7398071 DOI: 10.1186/s40104-020-00488-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) K88 commonly colonize in the small intestine and keep releasing enterotoxins to impair the intestinal barrier function and trigger inflammatory reaction. Although Lactobacillus salivarius (L. salivarius) has been reported to enhance intestinal health, it remains to be seen whether there is a functional role of L. salivarius in intestinal inflammatory response in intestinal porcine epithelial cell line (IPEC-J2) when stimulated with ETEC K88. In the present study, IPEC-J2 cells were first treated with L. salivarius followed by the stimulation of ETEC K88 for distinct time period. ETEC K88 adherent status, pattern recognition receptors (PRRs) mRNA, mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) activation, the release of pro-inflammation cytokines and cell integrity were examined. Results Aside from an inhibited adhesion of ETEC K88 to IPEC-J2 cells, L. salivarius was capable of remarkably attenuating the expression levels of interleukin (IL)-1β, tumor necrosis factor-α (TNF-α), IL-8, Toll-like receptor (TLR) 4, nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain-containing protein (NLRP) 3 and NLRP6. This alternation was accompanied by a significantly decreased phosphorylation of p38 MAPK and p65 NF-κB during ETEC K88 infection with L. salivarius pretreatment. Western blot analysis revealed that L. salivarius increased the expression levels of zona occludens 1 (ZO-1) and occludin (P < 0.05) in ETEC K88-infected IPEC-J2 cells. Compared with ETEC K88-infected groups, the addition of L. salivarius as well as extra inhibitors for MAPKs and NF-κB to ETEC K88-infected IPEC-J2 cells had the capability to reduce pro-inflammatory cytokines. Conclusions Collectively, our results suggest that L. salivarius might reduce inflammation-related cytokines through attenuating phosphorylation of p38 MAPK and blocking the NF-κB signaling pathways. Besides, L. salivarius displayed a potency in the enhancement of IPEC-J2 cell integrity.
Collapse
Affiliation(s)
- Jiayun Qiao
- College of Life Sciences, Tianjin Key Laboratory of Animal and Plant Resistance, Tianjin Normal University, Tianjin, 300387 People's Republic of China
| | - Zeyang Sun
- College of Life Sciences, Tianjin Key Laboratory of Animal and Plant Resistance, Tianjin Normal University, Tianjin, 300387 People's Republic of China
| | - Dongmei Liang
- College of Life Sciences, Tianjin Key Laboratory of Animal and Plant Resistance, Tianjin Normal University, Tianjin, 300387 People's Republic of China
| | - Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, 22 Jinjing Road, Tianjin, 300384 People's Republic of China
| |
Collapse
|
46
|
Interaction between Bifidobacterium bifidum and Listeria monocytogenes enhances antioxidant activity through oxidoreductase system. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
47
|
Liu Q, Yu Z, Tian F, Zhao J, Zhang H, Zhai Q, Chen W. Surface components and metabolites of probiotics for regulation of intestinal epithelial barrier. Microb Cell Fact 2020; 19:23. [PMID: 32024520 PMCID: PMC7003451 DOI: 10.1186/s12934-020-1289-4] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
The gut microbiota can significantly affect the function of the intestinal barrier. Some intestinal probiotics (such as Lactobacillus, Bifidobacteria, a few Escherichia coli strains, and a new generation of probiotics including Bacteroides thetaiotaomicron and Akkermansia muciniphila) can maintain intestinal epithelial homeostasis and promote health. This review first summarizes probiotics' regulation of the intestinal epithelium via their surface compounds. Surface layer proteins, flagella, pili and capsular polysaccharides constitute microbial-associated molecular patterns and specifically bind to pattern recognition receptors, which can regulate signaling pathways to produce cytokines or inhibit apoptosis, thereby attenuating inflammation and enhancing the function of the gut epithelium. The review also explains the effects of metabolites (such as secreted proteins, organic acids, indole, extracellular vesicles and bacteriocins) of probiotics on host receptors and the mechanisms by which these metabolites regulate gut epithelial barrier function. Previous reviews summarized the role of the surface macromolecules or metabolites of gut microbes (including both probiotics and pathogens) in human health. However, these reviews were mostly focused on the interactions between these substances and the intestinal mucosal immune system. In the current review, we only focused on probiotics and discussed the molecular interaction between these bacteria and the gut epithelial barrier.
Collapse
Affiliation(s)
- Qing Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Zhiming Yu
- Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of 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, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China
- International Joint Research Laboratory for Probiotics at Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu, China
- Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, 100048, People's Republic of China
| |
Collapse
|
48
|
Ho SW, El-Nezami H, Shah NP. The protective effects of enriched citrulline fermented milk with Lactobacillus helveticus on the intestinal epithelium integrity against Escherichia coli infection. Sci Rep 2020; 10:499. [PMID: 31949265 PMCID: PMC6965087 DOI: 10.1038/s41598-020-57478-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 11/29/2019] [Indexed: 12/24/2022] Open
Abstract
This study examined the protective effects of citrulline enriched-fermented milk with live Lactobacillus helveticus ASCC 511 (LH511) on intestinal epithelial barrier function and inflammatory response in IPEC-J2 cells caused by pathogenic Escherichia coli. Five percent (v/v) of fermented milk with live LH511 and 4 mM citrulline (5%LHFM_Cit-4mM) significantly stimulated the population of IPEC-J2 cells by 36% as determined by MTT assay. Adhesion level of LH511 was significantly increased by 9.2% when incubated with 5%LHFM_Cit-4mM and 5%LHFM_Cit-4mM reduced the adhesion of enterohemorrhagic (EHEC) and entero-invasive (EIEC) E. coli in IPEC-J2 cells by 35.79% and 42.74%, respectively. Treatment with 5%LHFM_Cit-4mM ameliorated lipopolysaccharide (LPS) from E. coli O55:B5 induced activated inflammatory cytokines expression (TNF-α, IL-6 and IL-8) and concentration (IL-6 and IL-8) and early apoptosis. It restored the transepithelial electrical resistance (TEER) and regulated the expression and distribution of tight junction (TJ) proteins (zonula occluden-1 (ZO-1), occludin and claudin-1), toll-like receptors (TLRs) (TLR2 and TLR4) and negative regulators of TLRs signalling pathway (A20 and IRAK-M). In conclusion, our findings suggested that 5%LHFM_Cit-4mM might have the positive effects on improving and maintaining the intestinal epithelial cell integrity and inflammatory response under both normal and pathogenic LPS-stimulated conditions.
Collapse
Affiliation(s)
- Sze Wing Ho
- Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Hani El-Nezami
- Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Nagendra P Shah
- Food and Nutritional Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong.
| |
Collapse
|
49
|
Timing of Calorie Restriction in Mice Impacts Host Metabolic Phenotype with Correlative Changes in Gut Microbiota. mSystems 2019; 4:4/6/e00348-19. [PMID: 31796564 PMCID: PMC6890928 DOI: 10.1128/msystems.00348-19] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aberrant feeding patterns whereby people eat more frequently throughout the day and with a bias toward late-night eating are prevalent in society today. However, whether restriction of food to daytime in comparison to nighttime, coupled with restricted calorie intake, can influence gut microbiota, metabolism, and overall health requires further investigation. We surveyed the effects of the shift in feeding time on gut microbiota and metabolic phenotype in calorie-restricted mice and found that avoiding eating during the rest period may generate more beneficial effects in mice. This work strengthens the evidence for using “when to eat” as an intervention to improve health during calorie restriction. Calorie restriction (CR) is accompanied by self-imposed daily restriction of food intake and an extended fasting period between meals. The impact of restricting feeding to the dark or light phase on the effects of CR remains elusive. Here, light-fed CR mice showed physiological changes, such as muscle loss, concomitant with changes in the gut microbiota structure and composition. After switching to ad libitum access to food, light-fed mice had a period of food-craving behavior and short-lived physiological changes, while dark-fed mice displayed lasting changes in fat accumulation, glucose metabolism, intestinal barrier function, and systemic inflammatory markers. Moreover, the gut microbiota was modulated by when the food was consumed, and the most abundant Lactobacillus operational taxonomic unit (OTU) promoted by CR was enhanced in dark-fed mice. After switching to ad libitum feeding, the gut microbiota of dark-fed mice returned to the state resembling that of mice fed normal chow ad libitum, but that of light-fed mice was still significantly different from the other two groups. Together, these data indicate that for CR, restricting food consumption to the active phase brought better metabolic phenotype associated with potentially beneficial structural shifts in the gut microbiota. IMPORTANCE Aberrant feeding patterns whereby people eat more frequently throughout the day and with a bias toward late-night eating are prevalent in society today. However, whether restriction of food to daytime in comparison to nighttime, coupled with restricted calorie intake, can influence gut microbiota, metabolism, and overall health requires further investigation. We surveyed the effects of the shift in feeding time on gut microbiota and metabolic phenotype in calorie-restricted mice and found that avoiding eating during the rest period may generate more beneficial effects in mice. This work strengthens the evidence for using “when to eat” as an intervention to improve health during calorie restriction.
Collapse
|
50
|
Synergistic efficacy of pulsed magnetic fields and Litseacubeba essential oil treatment against Escherichia coli O157:H7 in vegetable juices. Food Control 2019. [DOI: 10.1016/j.foodcont.2019.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|