1
|
Davarzani S, Reza Sanjabi M, Mojgani N, Mirdamadi S, Soltani M. Investigating the Antibacterial, Antioxidant, and Cholesterol-lowering Properties of Yogurt Fortified with Postbiotic of Lactobacillus acidophilus and Lactiplantibacillus plantarum in the Wistar Rat Model. J Food Prot 2024:100408. [PMID: 39547582 DOI: 10.1016/j.jfp.2024.100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/21/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Postbiotics have gained attention in the food industry due to their functional properties and ease of use compared to their live parent cells. Postbiotics are the metabolic byproducts of probiotic microorganisms, offering advantages such as antimicrobial and anti-diabetic effects. The study aimed to explore the potential antibacterial, antioxidant, and cholesterol-lowering effects of postbiotics from Lactobacillus acidophilus (LA) and Lactiplantibacillus plantarum (LbP) through in vitro and in vivo studies. Freeze-dried postbiotics from L. acidophilus BLAC 258 and L. plantarum were used in yogurt to inhibit foodborne pathogens over a 21-day storage period at 4 °C. The cholesterol-lowering effects of the postbiotic yogurt were assessed in Wistar rats fed with Normal Basal Diet (NBD) and High Cholesterol Diet (HCD). All experiments were performed in triplicate, and the collected data were analyzed with a one-way ANOVA using SPSS v.20 (2021) software. The Tukey Honestly Significant Difference (HSD) test was used for means differences at the 95% confidence interval. The results showed that postbiotic-fortified yogurt exhibited significant antioxidant and antibacterial effects. The antioxidant capacity of the yogurt increasingly peaked at 48.81% on day 14. Also, Listeria monocytogenes counts in the postbiotic yogurt decreased by approximately 2 Log10 on day 3. High-cholesterol-fed rats receiving postbiotic yogurt experienced significant reductions in total cholesterol, triglycerides, and LDL levels. Overall results indicate that postbiotics functional yogurt might be a safe and effective strategy for managing cholesterol levels and inhibiting foodborne pathogens.
Collapse
Affiliation(s)
- Sareh Davarzani
- Department of Food Sciences and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Reza Sanjabi
- Department of Agriculture, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran.
| | - Naheed Mojgani
- Biotechnology Department, Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Saeed Mirdamadi
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran.
| | - Mostafa Soltani
- Department of Food Sciences and Technology, Faculty of Pharmacy Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Nutrition and Food Sciences Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Jalali S, Mojgani N, Sanjabi MR, Saremnezhad S, Haghighat S. Functional properties and safety traits of L. rhamnosus and L. reuteri postbiotic extracts. AMB Express 2024; 14:114. [PMID: 39384663 PMCID: PMC11465093 DOI: 10.1186/s13568-024-01768-3] [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: 07/01/2024] [Accepted: 09/12/2024] [Indexed: 10/11/2024] Open
Abstract
Postbiotics are the non-viable bacterial products or the low molecular weight metabolites produced by probiotics that have received considerable attention owing to their health promoting effects. The present study aimed to investigate the safety and antibacterial properties of postbiotic components of Lacticaseibacillus rhamnosus (Lra) and Limosilactobacillus reuteri (Lre) for their potential applications in food products. The freeze dried postbiotic metabolites (FD-P) from Lra and Lre were extensively analyzed for their physico-chemical properties and antibacterial actions against common food borne pathogens. Higher levels of total flavonoids (1971.79 ± 20 mg Qu/ g), total short-chain fatty acid (23 µg/g), sugar contents, CAT, and SOD anti-oxidative enzymes were detected in the Lra postbiotic, while GSH-px levels and riboflavin were higher in Lre postbiotics (P < 0.01). No significant differences were recorded in the total phenolic (2501 and 2518 mg GAE/ L) and crude protein contents (305. 58 and 296.23 µg/g) of the postbiotics (p ≥ 0.05), respectively. Both FD-P samples showed enhanced activities against Gram-Positive pathogens compared to Gram-Negative pathogens (p < 0.05), while combining the two postbiotics further potentiated the antibacterial actions. Both FD-P samples were non-hemolytic to human erythrocyte cells, and exhibited low cytotoxicity in MRC 5 and IPEC-J2 cell lines at the highest used concentrations (150 mg/ml). In summary, the postbiotics derived from Lra and Lre are safe bioactive ingredients with enhanced antibacterial and antioxidant capabilities, having potential applications as a natural preservatives in food system, potentially enhancing safety and extending the shelf life of food products.
Collapse
Affiliation(s)
- Safura Jalali
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Naheed Mojgani
- Biotechnology Department, Razi Vaccine and Serum Research Institute- Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Mohammad Reza Sanjabi
- Department of Agriculture, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Solmaz Saremnezhad
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Pagar R, Deshkar S, Mahore J, Patole V, Deshpande H, Gandham N, Mirza S, Junnarkar M, Nawani N. The microbial revolution: Unveiling the benefits of vaginal probiotics and prebiotics. Microbiol Res 2024; 286:127787. [PMID: 38851010 DOI: 10.1016/j.micres.2024.127787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Vaginal health is essential to a woman's overall well-being, as abnormalities in vaginal health can lead to a variety of gynaecological disorders, such as urinary tract infections, yeast infections, and bacterial vaginosis. The vaginal microbiome is essential for the prevention of these infections. Disruptions in this microbial ecosystem can significantly impact vaginal health. The concept of utilizing probiotics and prebiotics to stimulate the growth of protective vaginal microbiota has gathered substantial interest in recent years. Probiotics are live micro-organisms that strengthen and restore vaginal microbial balance by lowering pH levels, production of bacteriocins, biofilm disruption, modulation of immune response, and production of hydrogen peroxide (H2O2), consequently combating the development of pathogens. Prebiotics are oligosaccharides that encourage the development of probiotics such as lactobacilli species. Probiotics and prebiotics also have some broader implications for vaginal health, including their role in minimizing the incidence of premature birth, optimizing fertility, managing menopausal symptoms, and preventing vaginal infections. Synbiotics are a combination of probiotics and prebiotics that deliver additional benefits by encouraging the development and activity of beneficial microbes. Furthermore, postbiotics are bioactive compounds derived from probiotic bacteria during fermentation that have immunomodulatory actions and provide an additional layer of protection against vaginal infections. The present study highlights the most prevalent vaginal infections and limitations of existing therapies that influence the vaginal microbiota. The profound consequences of probiotics and prebiotics in women's health, including their role in minimizing the prevalence of vaginal infections and promoting overall vaginal health, as well as advanced therapeutic strategies such as synbiotics and postbiotics, are also discussed. The literature offers significant insights into the mechanism, efficacy, and safety of probiotics and prebiotics to healthcare providers and researchers.
Collapse
Affiliation(s)
- Roshani Pagar
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Sanjeevani Deshkar
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India.
| | - Jayashri Mahore
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Vinita Patole
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Hemant Deshpande
- Department of Obstetrics and Gynaecology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Nageswari Gandham
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Shahzad Mirza
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Manisha Junnarkar
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| | - Neelu Nawani
- Microbial Diversity Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Pune, India
| |
Collapse
|
4
|
Luongo D, De Sena V, Maurano F, Rossi M. Modulation of Mouse Dendritic Cells In Vitro by Lactobacillus gasseri Postbiotic Proteins. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10292-6. [PMID: 38836988 DOI: 10.1007/s12602-024-10292-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/06/2024]
Abstract
Different lactobacilli are probiotics for their beneficial effects that confer to the host. Recently, some of these effects were associated with released metabolic products/constituents (postbiotics). In the present study, the potential immunomodulatory capacity of the probiotic Lactobacillus gasseri OLL2809 cell-free supernatant (sup) was investigated in murine bone marrow-derived dendritic cells (DCs). Bacteria induced significantly higher expression of all examined cytokines than those induced by the stimulatory lipopolysaccharide (LPS) itself. On the contrary, sup only induced the anti-inflammatory IL-10 similarly to LPS, whereas IL-12 and IL-6 secretions were stimulated at a lower level. Moreover, sup reduced the surface expression of the analyzed co-stimulatory markers CD40, CD80, and CD86. Treatments of sup with different digestive enzymes indicated the proteinaceous nature of these immunomodulatory metabolites. Western blot and immunoadsorption analyzes revealed cross-reactivity of sup with the surface-layer proteins (SLPs) isolated from OLL2809. Therefore, we directly tested the ability of OLL2809 SLPs to stimulate specifically cytokine expression in iDCs. Interestingly, we found that all tested cytokines were induced by SLPs and in a dose-dependent manner. In conclusion, our results highlighted distinct immune properties between L. gasseri OLL2809 and its metabolites, supporting the concept that bacterial viability is not an essential prerequisite to exert immunomodulatory effects.
Collapse
Affiliation(s)
- Diomira Luongo
- Institute of Food Sciences, National Research Council, Avellino, Italy.
| | - Vincenzo De Sena
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Francesco Maurano
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Mauro Rossi
- Institute of Food Sciences, National Research Council, Avellino, Italy
| |
Collapse
|
5
|
Biniszewska O, Jacenik D, Tarasiuk A, Fichna J. Current and future pharmacotherapies for the management of constipation-predominant irritable bowel syndrome. Expert Opin Pharmacother 2024; 25:1039-1049. [PMID: 38856704 DOI: 10.1080/14656566.2024.2366993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common gastrointestinal disorder affecting 9-23% of the world's population, with a higher prevalence among women. IBS is a complex disorder influenced by psychosocial, physiological, and genetic factors, exacerbated by stress. AREAS COVERED Research confirms that the most common subtype of IBS is IBS-C. Therefore, new therapies are being developed to speed up bowel movement and reduce constipation, with drugs such as linaclotide, plecanatide, lubiprostone, or tegaserod available to reduce IBS-C symptoms. In addition, patients' condition is improved by foods rich in fiber and low in FODMAP and the use of biotics. EXPERT OPINION The topic is of great importance due to the growing number of patients suffering from IBS-C and its significant impact on quality of life. Current clinical trials of new therapeutic options are not too successful, and it seems that one of the plausible treatment options could be the multi-drug cocktail with some, or perhaps even all its ingredients emerging from drug re-purposing. Another important path that needs to be explored further in IBS-C patients is the adjustment of dietary habits and/or introduction of dietary or nutritional intervention.
Collapse
Affiliation(s)
- Olga Biniszewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
6
|
Odey TOJ, Tanimowo WO, Afolabi KO, Jahid IK, Reuben RC. Antimicrobial use and resistance in food animal production: food safety and associated concerns in Sub-Saharan Africa. Int Microbiol 2024; 27:1-23. [PMID: 38055165 PMCID: PMC10830768 DOI: 10.1007/s10123-023-00462-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
The use of antimicrobials in food animal (FA) production is a common practice all over the world, with even greater usage and dependence in the developing world, including Sub-Saharan Africa (SSA). However, this practice which serves obvious economic benefits to producers has raised public health concerns over the last decades, thus driving the selection and dissemination of antimicrobial resistance and adversely impacting food safety and environmental health. This review presents the current and comprehensive antimicrobial usage practices in food animal production across SSA. We further highlighted the overall regional drivers as well as the public health, environmental, and economic impact of antimicrobial use in the production of food animals. Antimicrobial use is likely to increase with even exacerbated outcomes unless cost-effective, safe, and sustainable alternatives to antibiotics, especially probiotics, prebiotics, bacteriocins, antimicrobial peptides, bacteriophages, vaccines, etc. are urgently advocated for and used in food animal production in SSA. These, in addition to the implementation of strong legislation on antimicrobial use, and improved hygiene will help mitigate the public health concerns associated with antimicrobial use in food animals and improve the well-being and safety of food animals and their products.
Collapse
Affiliation(s)
- Timothy Obiebe Jason Odey
- Department of Biological Sciences, Faculty of Natural, Applied, and Health Sciences, Anchor University, Lagos, Nigeria
| | - Williams Omotola Tanimowo
- Department of Biological Sciences, Faculty of Natural, Applied, and Health Sciences, Anchor University, Lagos, Nigeria
| | - Kayode Olayinka Afolabi
- Department of Biological Sciences, Faculty of Natural, Applied, and Health Sciences, Anchor University, Lagos, Nigeria
- Pathogenic Yeasts Research Group, Department of Microbiology and Biochemistry, University of The Free State, Bloemfontein, South Africa
| | - Iqbal Kabir Jahid
- Department of Microbiology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Rine Christopher Reuben
- Department of Biological Sciences, Faculty of Natural, Applied, and Health Sciences, Anchor University, Lagos, Nigeria.
- Area of Biochemistry and Molecular Biology, OneHealth-UR Research Group, University of La Rioja, 26006, Logroño, Spain.
| |
Collapse
|
7
|
Zaib S, Hayat A, Khan I. Probiotics and their Beneficial Health Effects. Mini Rev Med Chem 2024; 24:110-125. [PMID: 37291788 DOI: 10.2174/1389557523666230608163823] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Probiotics are living microorganisms that are present in cultured milk and fermented food. Fermented foods are a rich source for the isolation of probiotics. They are known as good bacteria. They have various beneficial effects on human health including antihypertensive effects, antihypercholesterolemic effects, prevention of bowel disease, and improving the immune system. Microorganisms including bacteria, yeast, and mold are used as probiotics but the major microorganisms that are used as probiotics are bacteria from the genus Lactobacillus, Lactococcus, Streptococcus, and Bifidobacterium. Probiotics are beneficial in the prevention of harmful effects. Recently, the use of probiotics for the treatment of various oral and skin diseases has also gained significant attention. Clinical studies indicate that the usage of probiotics can alter gut microbiota composition and provoke immune modulation in a host. Due to their various health benefits, probiotics are attaining more interest as a substitute for antibiotics or anti-inflammatory drugs leading to the growth of the probiotic market.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Aqsa Hayat
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Department of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
8
|
Wen Y, Wang X, Si K, Xu L, Huang S, Zhan Y. Exploring the Mechanisms of Self-made Kuiyu Pingchang Recipe for the Treatment of Ulcerative Colitis and Irritable Bowel Syndrome using a Network Pharmacology-based Approach and Molecular Docking. Curr Comput Aided Drug Des 2024; 20:534-550. [PMID: 37190808 DOI: 10.2174/1573409919666230515103224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 03/24/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) and irritable bowel syndrome (IBS) are common intestinal diseases. According to the clinical experience and curative effect, the authors formulated Kuiyu Pingchang Decoction (KYPCD) comprised of Paeoniae radix alba, Aurantii Fructus, Herba euphorbiae humifusae, Lasiosphaera seu Calvatia, Angelicae sinensis radix, Panax ginseng C.A. Mey., Platycodon grandiforus and Allium azureum Ledeb. OBJECTIVE The aim of the present study was to explore the mechanisms of KYPCD in the treatment of UC and IBS following the Traditional Chinese Medicine (TCM) theory of "Treating different diseases with the same treatment". METHODS The chemical ingredients and targets of KYPCD were obtained using the Traditional Chinese Medicine Systems Pharmacology database and analysis platform (TCMSP). The targets of UC and IBS were extracted using the DisGeNET, GeneCards, DrugBANK, OMIM and TTD databases. The "TCM-component-target" network and the "TCM-shared target-disease" network were imaged using Cytoscape software. The protein-protein interaction (PPI) network was built using the STRING database. The DAVID platform was used to analyze the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Using Autodock Tools software, the main active components of KYPCD were molecularly docked with their targets and visualized using PyMOL. RESULTS A total of 46 active ingredients of KYPCD corresponding to 243 potential targets, 1,565 targets of UC and 1,062 targets of IBS, and 70 targets among active ingredients and two diseases were screened. Core targets in the PPI network included IL6, TNF, AKT1, IL1B, TP53, EGFR and VEGFA. GO and KEGG enrichment analysis demonstrated 563 biological processes, 48 cellular components, 82 molecular functions and 144 signaling pathways. KEGG enrichment results revealed that the regulated pathways were mainly related to the PI3K-AKT, MAPK, HIF-1 and IL-17 pathways. The results of molecular docking analysis indicated that the core active ingredients of KYPCD had optimal binding activity to their corresponding targets. CONCLUSION KYPCD may use IL6, TNF, AKT1, IL1B, TP53, EGFR and VEGFA as the key targets to achieve the treatment of UC and IBS through the PI3K-AKT, MAPK, HIF-1 and IL-17 pathways.
Collapse
Affiliation(s)
- Yong Wen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Anorectal Integration of Traditional Chinese and Western Medicine, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Xiaoxiang Wang
- Gastroenterology Department, Chengdu First People's Hospital, Chengdu, 610000, China
| | - Ke Si
- Gastroenterology Department, Chengdu First People's Hospital, Chengdu, 610000, China
| | - Ling Xu
- Anorectal Department, Luzhou Hospital of Traditional Chinese Medicine, Luzhou, 646000, China
| | - Shuoyang Huang
- Gastrointestinal Surgery Department, Chengdu Second People's Hospital, Chengdu, 610017, China
| | - Yu Zhan
- Gastroenterology Department, Chengdu First People's Hospital, Chengdu, 610000, China
- Anorectal Department, Chengdu First People's Hospital, Chengdu, 610000, China
- Anorectal Department, Affiliated Hospital of Integrative Chinese Medicine and Western Medicine of Chengdu University of TCM, Chengdu 610041, China
| |
Collapse
|
9
|
Gurunathan S, Thangaraj P, Kim JH. Postbiotics: Functional Food Materials and Therapeutic Agents for Cancer, Diabetes, and Inflammatory Diseases. Foods 2023; 13:89. [PMID: 38201117 PMCID: PMC10778838 DOI: 10.3390/foods13010089] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Postbiotics are (i) "soluble factors secreted by live bacteria, or released after bacterial lysis, such as enzymes, peptides, teichoic acids, peptidoglycan-derived muropeptides, polysaccharides, cell-surface proteins and organic acids"; (ii) "non-viable metabolites produced by microorganisms that exert biological effects on the hosts"; and (iii) "compounds produced by microorganisms, released from food components or microbial constituents, including non-viable cells that, when administered in adequate amounts, promote health and wellbeing". A probiotic- and prebiotic-rich diet ensures an adequate supply of these vital nutrients. During the anaerobic fermentation of organic nutrients, such as prebiotics, postbiotics act as a benevolent bioactive molecule matrix. Postbiotics can be used as functional components in the food industry by offering a number of advantages, such as being added to foods that are harmful to probiotic survival. Postbiotic supplements have grown in popularity in the food, cosmetic, and healthcare industries because of their numerous health advantages. Their classification depends on various factors, including the type of microorganism, structural composition, and physiological functions. This review offers a succinct introduction to postbiotics while discussing their salient features and classification, production, purification, characterization, biological functions, and applications in the food industry. Furthermore, their therapeutic mechanisms as antibacterial, antiviral, antioxidant, anticancer, anti-diabetic, and anti-inflammatory agents are elucidated.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
10
|
Liang B, Xing D. The Current and Future Perspectives of Postbiotics. Probiotics Antimicrob Proteins 2023; 15:1626-1643. [PMID: 36763279 PMCID: PMC9913028 DOI: 10.1007/s12602-023-10045-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/11/2023]
Abstract
With the emphasis on intestinal health, probiotics have exploded into a vast market potential. However, new scientific evidence points out that the beneficial health benefits of probiotics are not necessarily directly related to viable bacteria. However, the metabolites or bacterial components of the live bacteria are the driving force behind health promotion. Therefore, scientists gradually noticed that the beneficial effects of probiotics are based on bacteria itself, metabolites, or cell lysates, and these factors are officially named "postbiotics" by the ISAPP. Postbiotic components are diverse and outperform live probiotics in terms of technology, safety, and cost due to their good absorption, metabolism, and organismal distribution. Postbiotics have been shown to have bioactivities such as antimicrobial, antioxidant, anti-inflammatory, anti-proliferative, and immunomodulation. Moreover, numerous studies have revealed the significant potential of postbiotics for disease treatment. This paper first presents the production and classification of postbiotics with examples from lactic acid bacteria (LAB), followed by the mechanisms of action with the most recent pre-clinical and clinical studies and the wide range of non-clinical and clinical applications of postbiotics. Furthermore, the current and future prospects of the postbiotic market with commercial available products are discussed. Finally, we comment on the knowledge gaps and future clinical applications with several examples.
Collapse
Affiliation(s)
- Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.
- Cancer Institute, Qingdao University, Qingdao, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- Cancer Institute, Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
11
|
Napolitano M, Fasulo E, Ungaro F, Massimino L, Sinagra E, Danese S, Mandarino FV. Gut Dysbiosis in Irritable Bowel Syndrome: A Narrative Review on Correlation with Disease Subtypes and Novel Therapeutic Implications. Microorganisms 2023; 11:2369. [PMID: 37894027 PMCID: PMC10609453 DOI: 10.3390/microorganisms11102369] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder characterized by chronic abdominal pain and altered bowel habits. It can be subclassified in different subtypes according to the main clinical manifestation: constipation, diarrhea, mixed, and unclassified. Over the past decade, the role of gut microbiota in IBS has garnered significant attention in the scientific community. Emerging research spotlights the intricate involvement of microbiota dysbiosis in IBS pathogenesis. Studies have demonstrated reduced microbial diversity and stability and specific microbial alterations for each disease subgroup. Microbiota-targeted treatments, such as antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and even diet, offer exciting prospects for managing IBS. However, definitive conclusions are hindered by the heterogeneity of these studies. Further research should focus on elucidating the mechanisms, developing microbiome-based diagnostics, and enabling personalized therapies tailored to an individual's microbiome profile. This review takes a deep dive into the microscopic world inhabiting our guts, and its implications for IBS. Our aim is to elucidate the complex interplay between gut microbiota and each IBS subtype, exploring novel microbiota-targeted treatments and providing a comprehensive overview of the current state of knowledge.
Collapse
Affiliation(s)
- Maria Napolitano
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Ernesto Fasulo
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Federica Ungaro
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Luca Massimino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy;
| | - Silvio Danese
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Francesco Vito Mandarino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| |
Collapse
|
12
|
Wang X, Zhang Y, Wang D, Su N, Yang L, Fu H, Zhang J, Li M, Wang C. Protective effects of Aureobasidium pullulans lysate on UV-damaged human skin fibroblasts and HaCaT cells. BIORESOUR BIOPROCESS 2023; 10:55. [PMID: 38647892 PMCID: PMC10992526 DOI: 10.1186/s40643-023-00678-9] [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: 05/29/2023] [Accepted: 08/16/2023] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Aureobasidium pullulans (A. pullulans) has a wide range of applications. Ultraviolet (UV) rays from the sun can cause skin photoaging. In order to explore the protective effect and application potential of A. pullulans lysate on UV-damaged human skin fibroblasts (HSF) and HaCaT Cells, this study investigates the anti-aging and anti-inflammatory effects of A. pullulans lysate as well as the mechanism of anti-oxidative stress at the cellular and molecular levels through cytotoxicity experiments, enzyme-linked immunosorbent assays (ELISA), and real-time quantitative PCR (RT-qPCR). RESULTS The experimental results have shown that the A. pullulans lysate can effectively reduce the loss of extracellular matrix components (EMC), such as collagen and hyaluronic acid (HA). It is also capable of scavenging excess reactive oxygen species (ROS) from the body, thereby increasing the activity of catalase, decreasing the overexpression of intracellular matrix metalloproteinases (MMPs), enhancing the gene expression of metalloproteinase inhibitors (TIMPs), and decreasing the level of inflammatory factors, reducing UV-induced apoptosis of HaCaT cells. Meanwhile, oxidative stress homeostasis is also regulated through the Nrf2/Keap1 and MAPK signaling pathways. CONCLUSIONS This study shows that the A. pullulans lysate has the potential to resist photoaging.
Collapse
Affiliation(s)
- Xin Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| | - Yongtao Zhang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| | - Dongdong Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| | - Ning Su
- Chinese Academy of Inspection and Quarantine, Beijing, People's Republic of China
| | - Li Yang
- Beijing Sino-German Union Cosmetic Institute Co., Ltd, Beijing, People's Republic of China
| | - Hao Fu
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| | - Jiachan Zhang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| | - Meng Li
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China.
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China.
| | - Changtao Wang
- Beijing Key Laboratory of Plant Resource Research and Development, College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, People's Republic of China
- Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, People's Republic of China
| |
Collapse
|
13
|
Sun J, Zhang M, Liu W, Liu Y, Zhang D, Fan X, Zhang J, Li T, Lu M. Evaluation of the effectiveness and mechanism of action of the Chang-Kang-Fang formula combined with bifid triple viable capsules on diarrhea-predominant irritable bowel syndrome. Front Microbiol 2023; 14:1160783. [PMID: 37440881 PMCID: PMC10333534 DOI: 10.3389/fmicb.2023.1160783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction The Chang-Kang-Fang (CKF) formula, a traditional Chinese herbal formula, can decrease serotonin (5-HT) levels and treat irritable bowel syndrome (IBS). Probiotics have a better synergistic effect on diarrhea-predominant IBS (IBS-D) when combined with 5-HT3 receptor antagonists. The present study aimed to elucidate the efficacy and the mechanisms of action of the CKF formula combined with bifid triple viable capsules (PFK) against IBS-D. Methods The rat models of IBS-D were induced by gavage with senna decoction plus restraint stress. The CKF formula, PFK and their combination were administered to the rats. Their effects were evaluated based on general condition of the rats and the AWR score. The levels of 5-HT and fos protein in the colon and hippocampus were measured by immunohistochemistry. The levels of SP and VIP, as well as ZO-1 and occludin in the colon, were determined by enzyme-linked immunosorbent assay and immunohistochemistry. The intestinal microbiota in faeces was analyzed by 16S rRNA high-throughput sequencing. Results The results showed that the oral CKF formula combined with PFK (CKF + PFK) could significantly relieve the symptoms of IBS-D, including elevating the weight rate and decreasing the AWR score. Compared with the MC group, administration of CKF + PFK significantly reduced the expression of fos in the colon and hippocampus and that of 5-HT, SP and VIP in the colon and increased the levels of 5-HT in the hippocampus and ZO-1 and occludin in the colon. The above indexes exhibited statistical significance in the CKF + PFK group relative to those in the other groups. Moreover, treatment with CKF + PFK improved the diversity of intestinal microbiota and the abundance of Firmicutes, Lachnospiraceae and Ruminococcaceae but decreased those of Bacteroidetes and Prevotellaceae. Conclusions The CKF formula combined with PFK may have a synergistic effect on IBS-D by slowing gastrointestinal motility, lowering visceral hypersensitivity, enhancing the intestinal barrier function and modulating the composition of intestinal microbiota.
Collapse
Affiliation(s)
- Jing Sun
- Department of Central laboratory, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengqiu Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Suqian Hospital of Traditional Chinese Medicine, Suqian, China
| | - Wei Liu
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Youqian Liu
- Department of Gastroenterology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Dongjian Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyu Fan
- Department of Gastroenterology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian Zhang
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tian Li
- Department of Central laboratory, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Min Lu
- School of Third Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
14
|
Lupu VV, Ghiciuc CM, Stefanescu G, Mihai CM, Popp A, Sasaran MO, Bozomitu L, Starcea IM, Adam Raileanu A, Lupu A. Emerging role of the gut microbiome in post-infectious irritable bowel syndrome: A literature review. World J Gastroenterol 2023; 29:3241-3256. [PMID: 37377581 PMCID: PMC10292139 DOI: 10.3748/wjg.v29.i21.3241] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/04/2023] [Accepted: 05/08/2023] [Indexed: 06/01/2023] Open
Abstract
Post-infectious irritable bowel syndrome (PI-IBS) is a particular type of IBS, with symptom onset after an acute episode of infectious gastroenteritis. Despite infectious disease resolution and clearance of the inciting pathogen agent, 10% of patients will develop PI-IBS. In susceptible individuals, the exposure to pathogenic organisms leads to a marked shift in the gut microbiota with prolonged changes in host-microbiota interactions. These changes can affect the gut-brain axis and the visceral sensitivity, disrupting the intestinal barrier, altering neuromuscular function, triggering persistent low inflammation, and sustaining the onset of IBS symptoms. There is no specific treatment strategy for PI-IBS. Different drug classes can be used to treat PI-IBS similar to patients with IBS in general, guided by their clinical symptoms. This review summarizes the current evidence for microbial dysbiosis in PI-IBS and analyzes the available data regarding the role of the microbiome in mediating the central and peripheral dysfunctions that lead to IBS symptoms. It also discusses the current state of evidence on therapies targeting the microbiome in the management of PI-IBS. The results of microbial modulation strategies used in relieving IBS symptomatology are encouraging. Several studies on PI-IBS animal models reported promising results. However, published data that describe the efficacy and safety of microbial targeted therapy in PI-IBS patients are scarce. Future research is required.
Collapse
Affiliation(s)
- Vasile Valeriu Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Cristina Mihaela Ghiciuc
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Gabriela Stefanescu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | | | - Alina Popp
- Faculty of General Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest 020021, Romania
| | - Maria Oana Sasaran
- Faculty of General Medicine, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, Targu Mures 540142, Romania
| | - Laura Bozomitu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Iuliana Magdalena Starcea
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Anca Adam Raileanu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| | - Ancuta Lupu
- Faculty of General Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, Iasi 700115, Romania
| |
Collapse
|
15
|
Gut microbiota affects sensitivity to immune-mediated isoniazid-induced liver injury. Biomed Pharmacother 2023; 160:114400. [PMID: 36805186 DOI: 10.1016/j.biopha.2023.114400] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Isoniazid (INH) is a highly effective single and/or combined first-line anti-tuberculosis (anti-TB) therapy drug, and the hepatotoxicity greatly limits its clinical application. INH-induced liver injury (INH-DILI) is a typical immune-mediated idiosyncratic drug-induced liver injury. Existing mechanisms including genetic variations in drug metabolism and immune responses cannot fully explain the differences in susceptibility and sensitivity to INH-DILI, suggesting that other factors may be involved. Accumulating evidence indicates that the development and severity of immune-mediated liver injury is related to gut microbiota. In this study, INH exposure caused liver damage, immune disregulation and microbiota profile alteration. Depletion of gut microbiota ameliorated INH-DILI, and improved INH-DILI-associated immune disorder and inflammatory response. Moreover, hepatotoxicity of INH was ameliorated by fecal microbiota transplantation (FMT) from INH-treated mice. Notably, Bifidobacterium abundance was significantly associated with transaminase levels. In conclusion, our results suggested that the effect of gut microbiota on INH-DILI was related to immunity, and the difference in INH-DILI sensitivity was related to the structure of gut microbiota. Changes in the structure of gut microbiota by continuous exposure of INH resulted in the tolerance to liver injury, and probiotics such as Bifidobacterium might play an important role in INH-DILI and its "adaptation" phenomenon. This work provides novel evidence for elucidating the underlying mechanism of difference in individual's response to INH-DILI and potential approach for intervening anti-TB drug liver injury by modulating gut microbiota.
Collapse
|
16
|
Altered Gut Microbic Flora and Haemorrhoids: Could They Have a Possible Relationship? J Clin Med 2023; 12:jcm12062198. [PMID: 36983199 PMCID: PMC10054427 DOI: 10.3390/jcm12062198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/14/2023] Open
Abstract
To date, the exact pathophysiology of haemorrhoids is poorly understood. The different philosophies on haemorrhoids aetiology may lead to different approaches of treatment. A pathogenic theory involving a correlation between altered anal canal microflora, local inflammation, and muscular dyssynergia is proposed through an extensive review of the literature. Since the middle of the twentieth century, three main theories exist: (1) the varicose vein theory, (2) the vascular hyperplasia theory, and (3) the concept of a sliding anal lining. These phenomena determine changes in the connective tissue (linked to inflammation), including loss of organization, muscular hypertrophy, fragmentation of the anal subepithelial muscle and the elastin component, and vascular changes, including abnormal venous dilatation and vascular thrombosis. Recent studies have reported a possible involvement of gut microbiota in gut motility alteration. Furthermore, dysbiosis seems to represent the leading cause of bowel mucosa inflammation in any intestinal district. The alteration of the gut microbioma in the anorectal district could be responsible for haemorrhoids and other anorectal disorders. A deeper knowledge of the gut microbiota in anorectal disorders lays the basis for unveiling the roles of these various gut microbiota components in anorectal disorder pathogenesis and being conductive to instructing future therapeutics. The therapeutic strategy of antibiotics, prebiotics, probiotics, and fecal microbiota transplantation will benefit the effective application of precision microbiome manipulation in anorectal disorders.
Collapse
|
17
|
Giordani B, Abruzzo A, Parolin C, Foschi C, Laghi L, Marangoni A, Luppi B, Vitali B. Prebiotic Activity of Vaginal Lactobacilli on Bifidobacteria: from Concept to Formulation. Microbiol Spectr 2023; 11:e0200922. [PMID: 36602371 PMCID: PMC9927276 DOI: 10.1128/spectrum.02009-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The gut of babies born vaginally is rapidly colonized by Bifidobacterium spp. after birth, while in infants born by cesarean section (C-section), the presence of bifidobacteria drops dramatically, increasing the risk of developing gastrointestinal disorders. Considering that newborns naturally come into contact with maternal lactobacilli as they pass through the birth canal, the aim of this work is to exploit for the first time the bifidogenic activity exerted by the cell-free supernatants (CFSs) from lactobacilli of vaginal origin, belonging to the species Lactobacillus crispatus, Lactobacillus gasseri, Limosilactobacillus vaginalis, and Lactiplantibacillus plantarum. CFSs were recovered after 7 h, 13 h, and 24 h of fermentation and assessed for the ability to stimulate the planktonic growth and biofilms of Bifidobacterium strains belonging to species widely represented in the gut tract. A bifidogenic effect was observed for all CFSs; such activity was maximal for CFSs recovered in exponential phase and was strongly dependent on the species of lactobacilli. Importantly, no stimulating effects on an intestinal Escherichia coli strain were observed. CFSs from L. vaginalis BC17 showed the best bifidogenic profile since they increased bifidobacterial planktonic growth by up to 432% and biofilm formation by up to 289%. The CFS at 7 h from BC17 was successfully formulated with a hyaluronic acid-based hydrogel aimed at preventing and treating breast sores in lactating women and exerting bifidogenic activity in infants born mainly by C-section. IMPORTANCE Bifidobacteria in the gut tract of infants play crucial roles in the prevention of gastrointestinal diseases and the maturation of the immune system. Consequently, strategies to trigger a bifidogenic shift in the infant gut are highly desirable. Evidences suggest that the presence of a maternal vaginal microbiota dominated by health-promoting lactobacilli and the development of a bifidobacterium-enriched gut microbiota in newborns are interconnected. In this context, we found out that the cell-free supernatants from lactobacilli of vaginal origin were able to effectively stimulate the proliferation of Bifidobacterium spp. grown in free-floating and biofilm forms. The cell-free supernatant from Limosilactobacillus vaginalis BC17 showed excellent bifidogenic behavior, which was preserved even after its incorporation into a nipple formulation for lactating women. Lactobacilli derivatives, such as cell-free supernatants, have gained increasing interest by virtue of their safer profile than that of living cells and can be proposed as an ecosustainable approach to favor gut colonization of infants by bifidobacteria.
Collapse
Affiliation(s)
- Barbara Giordani
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Angela Abruzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Claudio Foschi
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Antonella Marangoni
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Barbara Luppi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
18
|
Saravanan P, R P, Balachander N, K KRS, S S, S R. Anti-inflammatory and wound healing properties of lactic acid bacteria and its peptides. Folia Microbiol (Praha) 2023; 68:337-353. [PMID: 36780113 PMCID: PMC9924211 DOI: 10.1007/s12223-022-01030-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 12/29/2022] [Indexed: 02/14/2023]
Abstract
Recent studies manifest an increase of inflammatory diseases at an alarming rate due to gut microbiota dysbiosis, genetic and other environmental factors. Lactic acid bacteria (LAB) are known for their antimicrobial properties and their extensive applications in food and pharmaceutical industries. Cyclic peptides are receiving increased attention due to their remarkable stability to withstand variations in temperature and pH. LAB produces anti-inflammatory that can inhibit lipopolysaccharide-induced production of proinflammatory cytokines in macrophages. The structural backbones of cyclic peptides offer a promising approach for the treatment of chronic inflammatory conditions. The current review aims to present the overview of anti-inflammatory and wound healing properties of LAB-derived cyclic peptides.
Collapse
Affiliation(s)
- Parikhshith Saravanan
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India
| | - Pooja R
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India
| | - Nanditaa Balachander
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India
| | - Kesav Ram Singh K
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India
| | - Silpa S
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India
| | - Rupachandra S
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering & Technology, SRM Institute of Science and Technology, Kattankulathur, Chennai, 603 203, India.
| |
Collapse
|
19
|
Casula E, Pisano MB, Serreli G, Zodio S, Melis MP, Corona G, Costabile A, Cosentino S, Deiana M. Probiotic lactobacilli attenuate oxysterols-induced alteration of intestinal epithelial cell monolayer permeability: Focus on tight junction modulation. Food Chem Toxicol 2023; 172:113558. [PMID: 36528245 DOI: 10.1016/j.fct.2022.113558] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022]
Abstract
Oxidative stress and inflammation lead by dietary oxidised lipids, as oxysterols, have been linked to the loss of intestinal barrier integrity, a crucial event in the initiation and progression of intestinal disorders. In the last decade, probiotic lactobacilli have emerged as an interesting tool to improve intestinal health, thanks to their antioxidant and anti-inflammatory properties. The aim of the present study was to evaluate the ability of two commercial probiotic strains of lactobacilli (Lactiplantibacillus plantarum 299v® (DMS 9843) and Lacticaseibacillus casei DG® (CNCMI-1572)), both as live bacteria and intracellular content, to attenuate the oxysterols-induced alteration of intestinal epithelial Caco-2 cell monolayer permeability. Our investigation was focused on the modulation of tight junctions (TJs) proteins, occludin, ZO-1 and JAM-A, in relation to redox-sensitive MAPK p38 activation. Obtained results provided evidence on the ability of the two probiotics to counteract the alteration of monolayer permeability and loss of TJs proteins, at least in part, through the modulation of p38 pathway. The protective action was exerted by live bacteria, whose adhesion to Caco-2 cells was not altered by oxysterols, and bacterial intracellular components equally able to interact with the signaling pathway.
Collapse
Affiliation(s)
- Emanuela Casula
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Maria Barbara Pisano
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Gabriele Serreli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy.
| | - Sonia Zodio
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Maria Paola Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Giulia Corona
- School of Life and Health Sciences, University of Roehampton, London, SW15 4JD, UK
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, SW15 4JD, UK
| | - Sofia Cosentino
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042, Cagliari, Italy.
| | - Monica Deiana
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| |
Collapse
|
20
|
Campbell C, Kandalgaonkar MR, Golonka RM, Yeoh BS, Vijay-Kumar M, Saha P. Crosstalk between Gut Microbiota and Host Immunity: Impact on Inflammation and Immunotherapy. Biomedicines 2023; 11:294. [PMID: 36830830 PMCID: PMC9953403 DOI: 10.3390/biomedicines11020294] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Gut microbes and their metabolites are actively involved in the development and regulation of host immunity, which can influence disease susceptibility. Herein, we review the most recent research advancements in the gut microbiota-immune axis. We discuss in detail how the gut microbiota is a tipping point for neonatal immune development as indicated by newly uncovered phenomenon, such as maternal imprinting, in utero intestinal metabolome, and weaning reaction. We describe how the gut microbiota shapes both innate and adaptive immunity with emphasis on the metabolites short-chain fatty acids and secondary bile acids. We also comprehensively delineate how disruption in the microbiota-immune axis results in immune-mediated diseases, such as gastrointestinal infections, inflammatory bowel diseases, cardiometabolic disorders (e.g., cardiovascular diseases, diabetes, and hypertension), autoimmunity (e.g., rheumatoid arthritis), hypersensitivity (e.g., asthma and allergies), psychological disorders (e.g., anxiety), and cancer (e.g., colorectal and hepatic). We further encompass the role of fecal microbiota transplantation, probiotics, prebiotics, and dietary polyphenols in reshaping the gut microbiota and their therapeutic potential. Continuing, we examine how the gut microbiota modulates immune therapies, including immune checkpoint inhibitors, JAK inhibitors, and anti-TNF therapies. We lastly mention the current challenges in metagenomics, germ-free models, and microbiota recapitulation to a achieve fundamental understanding for how gut microbiota regulates immunity. Altogether, this review proposes improving immunotherapy efficacy from the perspective of microbiome-targeted interventions.
Collapse
Affiliation(s)
- Connor Campbell
- Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA
| | - Mrunmayee R. Kandalgaonkar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| |
Collapse
|
21
|
Bednarska O, Biskou O, Israelsen H, Winberg ME, Walter S, Keita ÅV. A postbiotic fermented oat gruel may have a beneficial effect on the colonic mucosal barrier in patients with irritable bowel syndrome. Front Nutr 2022; 9:1004084. [PMID: 36570171 PMCID: PMC9773395 DOI: 10.3389/fnut.2022.1004084] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/17/2022] [Indexed: 12/13/2022] Open
Abstract
Background Impaired intestinal permeability and microbial dysbiosis are important pathophysiological mechanisms underlying irritable bowel syndrome (IBS). ReFerm®, also called Profermin®, is a postbiotic product of oat gruel fermented with Lactobacillus plantarum 299v. In this study, we investigated whether ReFerm® has a beneficial effect on the intestinal epithelial barrier function in patients with IBS. Materials and methods Thirty patients with moderate to severe IBS-diarrhoea (IBS-D) or IBS-mixed (IBS-M) were treated with enema containing ReFerm® or placebo. The patients underwent sigmoidoscopy with biopsies obtained from the distal colon at baseline and after 14 days of treatment with ReFerm® or placebo twice daily. The biopsies were mounted in Ussing chambers, and paracellular and transcellular permeabilities were measured for 120 min. In addition, the effects of ReFerm® or placebo on the epithelial barrier were investigated in vitro using Caco-2 cells. Results ReFerm® reduced paracellular permeability (p < 0.05) and increased transepithelial resistance (TER) over time (p < 0.01), whereas the placebo had no significant effect in patients. In ReFerm®-treated Caco-2 cells, paracellular and transcellular permeabilities were decreased compared to the control (p < 0.05) and placebo (p < 0.01). TER was increased in Caco-2 ReFerm®-treated cells, and normalised TER was increased in ReFerm®-treated Caco-2 cells compared to control (p < 0.05) and placebo-treated (p < 0.05) cells. Conclusion ReFerm® significantly reduced paracellular permeability and improved TER in colonic biopsies collected from patients with IBS and in a Caco-2 cell model. Our results offer new insights into the potential benefits of ReFerm® in IBS management. Further studies are needed to identify the molecular mechanisms underlying the barrier-protective properties of ReFerm®. Clinical trial registration [https://clinicaltrials.gov/], identifier [NCT05475314].
Collapse
Affiliation(s)
- Olga Bednarska
- Department of Gastroenterology, Linköping University Hospital, Linköping, Sweden
| | - Olga Biskou
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Martin E. Winberg
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Susanna Walter
- Department of Gastroenterology, Linköping University Hospital, Linköping, Sweden,Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Åsa V. Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden,*Correspondence: Åsa V. Keita,
| |
Collapse
|
22
|
Viswanathan K, Muthusamy S. Review on the current trends and future perspectives of postbiotics for developing healtheir foods. EFOOD 2022. [DOI: 10.1002/efd2.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
23
|
Ramos-Lopez O, Martinez-Urbistondo D, Vargas-Nuñez JA, Martinez JA. The Role of Nutrition on Meta-inflammation: Insights and Potential Targets in Communicable and Chronic Disease Management. Curr Obes Rep 2022; 11:305-335. [PMID: 36258149 PMCID: PMC9579631 DOI: 10.1007/s13679-022-00490-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/27/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Chronic low-grade inflammation may contribute to the onset and progression of communicable and chronic diseases. This review examined the effects and eventual mediation roles of different nutritional factors on inflammation. RECENT FINDINGS Potential nutritional compounds influencing inflammation processes include macro and micronutrients, bioactive molecules (polyphenols), specific food components, and culinary ingredients as well as standardized dietary patterns, eating habits, and chrononutrition features. Therefore, research in this field is still required, taking into account critical aspects of heterogeneity including type of population, minimum and maximum intakes and adverse effects, cooking methods, physiopathological status, and times of intervention. Moreover, the integrative analysis of traditional variables (age, sex, metabolic profile, clinical history, body phenotype, habitual dietary intake, physical activity levels, and lifestyle) together with individualized issues (genetic background, epigenetic signatures, microbiota composition, gene expression profiles, and metabolomic fingerprints) may contribute to the knowledge and prescription of more personalized treatments aimed to improving the precision medical management of inflammation as well as the design of anti-inflammatory diets in chronic and communicable diseases.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Universidad 14418, UABC, Parque Internacional Industrial Tijuana, 22390, Tijuana, Baja California, Mexico.
| | | | - Juan A Vargas-Nuñez
- Servicio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, 28222, Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, 28029, Madrid, Spain
| | - J Alfredo Martinez
- Precision Nutrition and Cardiometabolic Health, IMDEA Food Institute, CEI UAM+CSIC, 28049, Madrid, Spain
- Department of Nutrition, Food Science, Physiology and Toxicology, Centre for Nutrition Research, University of Navarra, 31009, Pamplona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBERobn), 28029, Madrid, Spain
| |
Collapse
|
24
|
Șchiopu CG, Ștefănescu C, Boloș A, Diaconescu S, Gilca-Blanariu GE, Ștefănescu G. Functional Gastrointestinal Disorders with Psychiatric Symptoms: Involvement of the Microbiome-Gut-Brain Axis in the Pathophysiology and Case Management. Microorganisms 2022; 10:2199. [PMID: 36363791 PMCID: PMC9694215 DOI: 10.3390/microorganisms10112199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022] Open
Abstract
Functional Gastrointestinal Disorders have been an important cause of poor life quality in affected populations. The unclear etiology and pathophysiological mechanism alter the clinical evolution of the patient. Although a strong connection with psychological stress has been observed, it was not until recently that the gut-brain axis involvement has been revealed. Furthermore, the current literature not only promotes the gut-brain axis modulation as a therapeutical target for functional digestive disorders but also states that the gut microbiome has a main role in this bi-directional mechanism. Psychiatric symptoms are currently recognized as an equally important aspect of the clinical manifestation and modulation of both the digestive and central nervous systems and could be the best approach in restoring the balance. As such, this article proposes a detailed description of the physiology of the microbiome-gut-brain axis, the pathophysiology of the functional gastrointestinal disorders with psychiatric symptoms and current perspectives for therapeutical management, as revealed by the latest studies in the scientific literature.
Collapse
Affiliation(s)
- Cristina Gabriela Șchiopu
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Cristinel Ștefănescu
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Alexandra Boloș
- Department of Psychiatry, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| | - Smaranda Diaconescu
- Medical-Surgical Department, Faculty of Medicine, University “Titu Maiorescu”, 040441 Bucuresti, Romania
| | | | - Gabriela Ștefănescu
- Department of Gastroentereology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iași, Romania
| |
Collapse
|
25
|
Rastogi S, Singh A. Gut microbiome and human health: Exploring how the probiotic genus Lactobacillus modulate immune responses. Front Pharmacol 2022; 13:1042189. [PMID: 36353491 PMCID: PMC9638459 DOI: 10.3389/fphar.2022.1042189] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/12/2022] [Indexed: 12/15/2022] Open
Abstract
The highest density of microbes resides in human gastrointestinal tract, known as “Gut microbiome”. Of note, the members of the genus Lactobacillus that belong to phyla Firmicutes are the most important probiotic bacteria of the gut microbiome. These gut-residing Lactobacillus species not only communicate with each other but also with the gut epithelial lining to balance the gut barrier integrity, mucosal barrier defence and ameliorate the host immune responses. The human body suffers from several inflammatory diseases affecting the gut, lungs, heart, bone or neural tissues. Mounting evidence supports the significant role of Lactobacillus spp. and their components (such as metabolites, peptidoglycans, and/or surface proteins) in modulatingimmune responses, primarily through exchange of immunological signals between gastrointestinal tract and distant organs. This bidirectional crosstalk which is mediated by Lactobacillus spp. promotes anti-inflammatory response, thereby supporting the improvement of symptoms pertaining to asthma, chronic obstructive pulmonary disease (COPD), neuroinflammatory diseases (such as multiple sclerosis, alzheimer’s disease, parkinson’s disease), cardiovascular diseases, inflammatory bowel disease (IBD) and chronic infections in patients. The metabolic disorders, obesity and diabetes are characterized by a low-grade inflammation. Genus Lactobacillus alleviates metabolic disorders by regulating the oxidative stress response and inflammatory pathways. Osteoporosis is also associated with bone inflammation and resorption. The Lactobacillus spp. and their metabolites act as powerful immune cell controllers and exhibit a regulatory role in bone resorption and formation, supporting bone health. Thus, this review demonstrated the mechanisms and summarized the evidence of the benefit of Lactobacillus spp. in alleviating inflammatory diseases pertaining to different organs from animal and clinical trials. The present narrative review explores in detail the complex interactions between the gut-dwelling Lactobacillus spp. and the immune components in distant organs to promote host’s health.
Collapse
|
26
|
Park M, Joung M, Park JH, Ha SK, Park HY. Role of Postbiotics in Diet-Induced Metabolic Disorders. Nutrients 2022; 14:nu14183701. [PMID: 36145077 PMCID: PMC9503758 DOI: 10.3390/nu14183701] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022] Open
Abstract
Although the prevalence of metabolic disorders has progressively increased over the past few decades, metabolic disorders can only be effectively treated with calorie restriction and improved physical activity. Recent research has focused on altering the gut microbiome using prebiotics, probiotics, and postbiotics because various metabolic syndromes are caused by gut microbial dysbiosis. Postbiotics, substances produced or released by microorganism metabolic activities, play an important role in maintaining and restoring host health. Because postbiotics have a small amount of literature on their consumption, there is a need for more experiments on short- and long-term intake. This review discusses current postbiotic research, categories of postbiotics, positive roles in metabolic syndromes, and potential therapeutic applications. It covers postbiotic pleiotropic benefits, such as anti-obesity, anti-diabetic, and anti-hypertensive qualities, that could aid in the management of metabolic disorders. Postbiotics are promising tools for developing health benefits and therapeutic goals owing to their clinical, technical, and economic properties. Postbiotic use is attractive for altering the microbiota; however, further studies are needed to determine efficacy and safety.
Collapse
|
27
|
Wu Y, Wang Y, Hu A, Shu X, Huang W, Liu J, Wang B, Zhang R, Yue M, Yang C. Lactobacillus plantarum-derived postbiotics prevent Salmonella-induced neurological dysfunctions by modulating gut-brain axis in mice. Front Nutr 2022; 9:946096. [PMID: 35967771 PMCID: PMC9365972 DOI: 10.3389/fnut.2022.946096] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/20/2022] [Indexed: 01/04/2023] Open
Abstract
Postbiotics are the inactive bacteria and/or metabolites of beneficial microbes which have been recently found to be as effective as their live probiotic. This study aimed to evaluate the benefits of Lactobacillus plantarum (LP)-derived postbiotics on ameliorating Salmonella-induced neurological dysfunctions. Mice were pretreated with LP postbiotics (heat-killed bacteria or the metabolites) or active bacteria, and then challenged with Salmonella enterica Typhimurium (ST). Results showed that LP postbiotics, particularly the metabolites, effectively prevented ST infection in mice, as evidenced by the inhibited weight loss, bacterial translocation, and tissue damages. The LP postbiotics markedly suppressed brain injuries and neuroinflammation (the decreased interleukin (IL)-1β and IL-6, and the increased IL-4 and IL-10). Behavior tests indicated that LP postbiotics, especially the metabolites, protected mice from ST-induced anxiety and depressive-like behaviors and cognitive impairment. A significant modulation of neuroactive molecules (5-hydroxytryptamine, gamma-aminobutyric acid, brain-derived neurotrophic factor, dopamine, acetylcholine, and neuropeptide Y) was also found by LP postbiotic pretreatment. Microbiome analysis revealed that LP postbiotics optimized the cecal microbial composition by increasing Helicobacter, Lactobacillus and Dubosiella, and decreasing Mucispirillum, norank_f_Oscillospiraceae, and Eubacterium_siraeum_group. Moreover, LP postbiotics inhibited the reduction of short-chain fatty acids caused by ST infection. Pearson's correlation assays further confirmed the strong relationship of LP postbiotics-mediated benefits and gut microbiota. This study highlights the effectiveness of postbiotics and provide a promising strategy for preventing infection-induced brain disorders by targeting gut–brain axis.
Collapse
Affiliation(s)
- Yanping Wu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Yan Wang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Aixin Hu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Xin Shu
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Wenxia Huang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Jinsong Liu
- Zhejiang Vegamax Biotechnology Co., Ltd., Huzhou, China
| | - Baikui Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Ruiqiang Zhang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| | - Min Yue
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Caimei Yang
- College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou, China
| |
Collapse
|
28
|
Yang YH, Qian W, Hou XH, Dai CB. Bifidobacterium bifidum and Bacteroides fragilis Induced Differential Immune Regulation of Enteric Glial Cells Subjected to Exogenous Inflammatory Stimulation. Inflammation 2022; 45:2388-2405. [PMID: 35776290 DOI: 10.1007/s10753-022-01700-6] [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: 12/24/2021] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022]
Abstract
Enteric glial cells (EGCs) are involved in intestinal inflammation. In this study, we will investigate how Bifidobacterium bifidum (B.b.) and Bacteroides fragilis (B.f.) influence EGC regulation. After pretreatment with lipopolysaccharide (LPS) and interferon-γ (IFN-γ), the expressions of major histocompatibility complex class II (MHC-II), CD80, CD86, glial cell line-derived neurotrophic factor (GDNF), toll-like receptor 2 (TLR-2), and tumor necrosis factor-α (TNF-α) in EGCs were detected using polymerase chain reaction and western blot after co-culture with the supernatants of B.b. or B.f. (multiplicity of infection, 40:1 or 80:1). Finally, EGCs were co-cultured with naive CD4+ T cells, and the expressions of interleukin (IL)-2, IL-4, IL-10, and IL-17 in supernatant were measured using enzyme-linked immunosorbent assay (ELISA). The mRNA expressions of MHC-II and CD86 in EGCs were increased after combined stimulation with LPS and IFN-γ. The expressions of MHC-II, GDNF, TLR-2, and TNF-α were all significantly upregulated in stimulated EGCs. The B.b. supernatant downregulated the expressions of MHC-II, GDNF, TLR-2, and TNF-α in stimulated EGCs, whereas the B.f. supernatant upregulated TLR-2 expression and downregulated MHC-II expression. The expressions of IL-4, IL-2, and IL-17 after co-culture of naive CD4+ T cells and stimulated EGCs were significantly increased. The supernatant of B.b. or B.f. downregulated the expressions of these cytokines. The low-concentration B.b. supernatant upregulated IL-10 expression. Conclusions B.b. and B.f. may influence intestinal inflammation by regulating MHC-II, GDNF, TLR-2, and TNF-α expression in EGCs and IL-4, IL-2, IL-17, and IL-10 secretion.
Collapse
Affiliation(s)
- Yan-Hua Yang
- Division of Gastroenterology, Affiliated RenHe Hospital of Three Gorges University, Yichang, 443001, China
- Division of Gastroenterology, Central Hospital of Enshi Autonomous Prefecture, Hubei Province, Enshi, 445000, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao-Hua Hou
- Division of Gastroenterology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chi-Bing Dai
- Division of Gastroenterology, Affiliated RenHe Hospital of Three Gorges University, Yichang, 443001, China.
| |
Collapse
|
29
|
Boahen A, Than LTL, Loke YL, Chew SY. The Antibiofilm Role of Biotics Family in Vaginal Fungal Infections. Front Microbiol 2022; 13:787119. [PMID: 35694318 PMCID: PMC9179178 DOI: 10.3389/fmicb.2022.787119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/25/2022] [Indexed: 11/15/2022] Open
Abstract
“Unity in strength” is a notion that can be exploited to characterize biofilms as they bestow microbes with protection to live freely, escalate their virulence, confer high resistance to therapeutic agents, and provide active grounds for the production of biofilms after dispersal. Naturally, fungal biofilms are inherently resistant to many conventional antifungals, possibly owing to virulence factors as their ammunitions that persistently express amid planktonic transition to matured biofilm state. These ammunitions include the ability to form polymicrobial biofilms, emergence of persister cells post-antifungal treatment and acquisition of resistance genes. One of the major disorders affecting vaginal health is vulvovaginal candidiasis (VVC) and its reoccurrence is termed recurrent VVC (RVVC). It is caused by the Candida species which include Candida albicans and Candida glabrata. The aforementioned Candida species, notably C. albicans is a biofilm producing pathogen and habitually forms part of the vaginal microbiota of healthy women. Latest research has implicated the role of fungal biofilms in VVC, particularly in the setting of treatment failure and RVVC. Consequently, a plethora of studies have advocated the utilization of probiotics in addressing these infections. Specifically, the excreted or released compounds of probiotics which are also known as postbiotics are being actively researched with vast potential to be used as therapeutic options for the treatment and prevention of VVC and RVVC. These potential sources of postbiotics are harnessed due to their proven antifungal and antibiofilm. Hence, this review discusses the role of Candida biofilm formation in VVC and RVVC. In addition, we discuss the application of pro-, pre-, post-, and synbiotics either individually or in combined regimen to counteract the abovementioned problems. A clear understanding of the role of biofilms in VVC and RVVC will provide proper footing for further research in devising novel remedies for prevention and treatment of vaginal fungal infections.
Collapse
|
30
|
Fathima S, Shanmugasundaram R, Adams D, Selvaraj RK. Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens. Foods 2022; 11:1401. [PMID: 35626971 PMCID: PMC9140538 DOI: 10.3390/foods11101401] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 12/17/2022] Open
Abstract
The gut of warm-blooded animals is colonized by microbes possibly constituting at least 100 times more genetic material of microbial cells than that of the somatic cells of the host. These microbes have a profound effect on several physiological functions ranging from energy metabolism to the immune response of the host, particularly those associated with the gut immune system. The gut of a newly hatched chick is typically sterile but is rapidly colonized by microbes in the environment, undergoing cycles of development. Several factors such as diet, region of the gastrointestinal tract, housing, environment, and genetics can influence the microbial composition of an individual bird and can confer a distinctive microbiome signature to the individual bird. The microbial composition can be modified by the supplementation of probiotics, prebiotics, or synbiotics. Supplementing these additives can prevent dysbiosis caused by stress factors such as infection, heat stress, and toxins that cause dysbiosis. The mechanism of action and beneficial effects of probiotics vary depending on the strains used. However, it is difficult to establish a relationship between the gut microbiome and host health and productivity due to high variability between flocks due to environmental, nutritional, and host factors. This review compiles information on the gut microbiota, dysbiosis, and additives such as probiotics, postbiotics, prebiotics, and synbiotics, which are capable of modifying gut microbiota and elaborates on the interaction of these additives with chicken gut commensals, immune system, and their consequent effects on health and productivity. Factors to be considered and the unexplored potential of genetic engineering of poultry probiotics in addressing public health concerns and zoonosis associated with the poultry industry are discussed.
Collapse
Affiliation(s)
- Shahna Fathima
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Revathi Shanmugasundaram
- Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA
| | - Daniel Adams
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Ramesh K. Selvaraj
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| |
Collapse
|
31
|
Zhu LY, Huang BW, Zhang XL, Zhu N. Progress in application of probiotics, prebiotics, and synbiotics in irritable bowel syndrome. Shijie Huaren Xiaohua Zazhi 2022; 30:375-380. [DOI: 10.11569/wcjd.v30.i8.375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common functional bowel disorder characterized by recurrent abdominal pain accompanied by changes in defecation frequency and/or stool characteristics. The global incidence of IBS is increasing year by year. Intestinal symptoms caused by IBS (such as constipation, diarrhea, abdominal pain, and abdominal distension) and accompanying changes in general nervous system function can significantly reduce patients' quality of life and work efficiency, and lead to high medical costs. Therefore, finding safe, effective, and economical treatments has become a hot research topic in recent years. Studies have shown that the intestinal flora of patients with IBS is different from that of healthy subjects, and regulating the intestinal flora can treat IBS. The purpose of this review is to summarize the application and recent progress of probiotics, prebiotics, and synbiotics in the treatment of IBS by regulating the intestinal flora.
Collapse
Affiliation(s)
- Liu-Yan Zhu
- Department of General Practice, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou 325000, Zhejiang Province, China
| | - Bing-Wu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Xue-Liang Zhang
- Department of General Practice, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou 325000, Zhejiang Province, China
| | - Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
32
|
Lactobacillus paracasei CNCM I 1572: A Promising Candidate for Management of Colonic Diverticular Disease. J Clin Med 2022; 11:jcm11071916. [PMID: 35407527 PMCID: PMC8999804 DOI: 10.3390/jcm11071916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Diverticular disease (DD) is a common gastrointestinal condition. Patients with DD experience a huge variety of chronic nonspecific symptoms, including abdominal pain, bloating, and altered bowel habits. They are also at risk of complications such as acute diverticulitis, abscess formation, hemorrhage, and perforation. Intestinal dysbiosis and chronic inflammation have recently been recognized as potential key factors contributing to disease progression. Probiotics, due to their ability to modify colonic microbiota balance and to their immunomodulatory effects, could present a promising treatment option for patients with DD. Lactobacillus paracasei CNCM I 1572 (LCDG) is a probiotic strain with the capacity to rebalance gut microbiota and to decrease intestinal inflammation. This review summarizes the available clinical data on the use of LCDG in subjects with colonic DD.
Collapse
|
33
|
Wu H, Lei Y, Mao J. Non-alcoholic fatty liver disease and intestinal immune status: a narrative review. Scand J Gastroenterol 2022:1-8. [PMID: 35188038 DOI: 10.1080/00365521.2022.2032320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023]
Abstract
Background and objectives: Non-alcoholic fatty liver disease (NAFLD) interacts with the gut immunity. However, the mechanisms underlying alternations of intestinal immune system in NAFLD remains unclear. To date, no effective medical interventions exist that completely reverse the disease. In this review, we mainly elaborates on the impact of NAFLD on intestinal immune cells and briefly summarize the new treatment methods for NAFLD targeting at intestinal immune cells.Methods: We searched MEDLINE, EMBASE and Web of Science for English-language sources. The preferred citations were meta-analyses and systematic or narrative reviews. Citation tracking was completed for all identified studies included in the refined library, using Google Scholar. No restriction was placed on the year of publication for the included reports.Results: The intestinal immune imbalance promotes liver inflammation and fibrosis in the process of NAFLD, and meanwhile, NAFLD influences disorders of immune cells in the liver and intestinal tract. Biological agents targeting at intestinal immunity has been shown in preclinical studies to be an effective method for systemic immune modulation and alleviates immune-mediated injury.Conclusions: Intestinal immune disorder plays an important role in triggering and amplifying hepatic inflammation in NAFLD. Advances in knowledge of the gut-liver axis are driving the development of diagnostic, prognostic and therapeutic tools based on intestine immunity for the management of NAFLD.
Collapse
Affiliation(s)
- Hao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yalan Lei
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Gastroenterology, The First People's Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Jingwei Mao
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
34
|
Liu Y, Wang J, Wu C. Modulation of Gut Microbiota and Immune System by Probiotics, Pre-biotics, and Post-biotics. Front Nutr 2022; 8:634897. [PMID: 35047537 PMCID: PMC8761849 DOI: 10.3389/fnut.2021.634897] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
The human gastrointestinal tract harbours a complex microbial community, which interacts with the mucosal immune system closely. Gut microbiota plays a significant role in maintaining host health, which could supply various nutrients, regulate energy balance, modulate the immune response, and defence against pathogens. Therefore, maintaining a favourable equilibrium of gut microbiota through modulating bacteria composition, diversity, and their activity is beneficial to host health. Several studies have shown that probiotics and pre-biotics could directly and indirectly regulate microbiota and immune response. In addition, post-biotics, such as the bioactive metabolites, produced by gut microbiota, and/or cell-wall components released by probiotics, also have been shown to inhibit pathogen growth, maintain microbiota balance, and regulate an immune response. This review summarises the studies concerning the impact of probiotics, pre-biotics, and post-biotics on gut microbiota and immune systems and also describes the underlying mechanisms of beneficial effects of these substances. Finally, the future and challenges of probiotics, pre-biotics, and post-biotics are proposed.
Collapse
Affiliation(s)
- Yue Liu
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China.,The Provincial Key Laboratories for Prevention and Treatment of Major Infectious Diseases Shanxi, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Jiaqi Wang
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China.,The Provincial Key Laboratories for Prevention and Treatment of Major Infectious Diseases Shanxi, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Changxin Wu
- Key Lab of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China.,The Provincial Key Laboratories for Prevention and Treatment of Major Infectious Diseases Shanxi, Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| |
Collapse
|
35
|
Bi C, Xiao G, Liu C, Yan J, Chen J, Si W, Zhang J, Liu Z. Molecular Immune Mechanism of Intestinal Microbiota and Their Metabolites in the Occurrence and Development of Liver Cancer. Front Cell Dev Biol 2021; 9:702414. [PMID: 34957088 PMCID: PMC8693382 DOI: 10.3389/fcell.2021.702414] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Intestinal microorganisms are closely associated with immunity, metabolism, and inflammation, and play an important role in health and diseases such as inflammatory bowel disease, diabetes, cardiovascular disease, Parkinson’s disease, and cancer. Liver cancer is one of the most fatal cancers in humans. Most of liver cancers are slowly transformed from viral hepatitis, alcoholic liver disease, and non-alcoholic fatty liver disease. However, the relationship between intestinal microbiota and their metabolites, including short-chain fatty acids, bile acids, indoles, and ethanol, and liver cancer remains unclear. Here, we summarize the molecular immune mechanism of intestinal microbiota and their metabolites in the occurrence and development of liver cancer and reveal the important role of the microbiota-gut-liver axis in liver cancer. In addition, we describe how the intestinal flora can be balanced by antibiotics, probiotics, postbiotics, and fecal bacteria transplantation to improve the treatment of liver cancer. This review describes the immunomolecular mechanism of intestinal microbiota and their metabolites in the occurrence and development of hepatic cancer and provides theoretical evidence support for future clinical practice.
Collapse
Affiliation(s)
- Chenchen Bi
- Department of Pharmacology, Medical College of Shaoxing University, Shaoxing, China
| | - Geqiong Xiao
- Department of Oncology, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Chunyan Liu
- Department of Clinical Medicine, Shaoxing People's Hospital, Shaoxing, China
| | - Junwei Yan
- Department of Pharmacology, Medical College of Shaoxing University, Shaoxing, China
| | - Jiaqi Chen
- Department of Pharmacology, Medical College of Shaoxing University, Shaoxing, China
| | - Wenzhang Si
- Department of General Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Jian Zhang
- Department of Pharmacology, Medical College of Shaoxing University, Shaoxing, China
| | - Zheng Liu
- Department of Pharmacology, Medical College of Shaoxing University, Shaoxing, China
| |
Collapse
|
36
|
Reuben RC, Sarkar SL, Roy PC, Anwar A, Hossain MA, Jahid IK. Prebiotics, probiotics and postbiotics for sustainable poultry production. WORLD POULTRY SCI J 2021. [DOI: 10.1080/00439339.2021.1960234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rine Christopher Reuben
- Department of Microbiology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
- German Centre for Integrative Biodiversity Research (Idiv), Halle-Jena-Leipzig, Germany
| | - Shovon Lal Sarkar
- Department of Microbiology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Pravas Chandra Roy
- Department of Microbiology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | | | - M. Anwar Hossain
- Department of Microbiology, University of Dhaka and Vice Chancellor, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Iqbal Kabir Jahid
- Department of Microbiology, Faculty of Biological Sciences and Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| |
Collapse
|
37
|
Castagliuolo I, Scarpa M, Brun P, Bernabe G, Sagheddu V, Elli M, Fiore W, De Vitis V, Guglielmetti S. Co-administration of vitamin D3 and Lacticaseibacillus paracasei DG increase 25-hydroxyvitamin D serum levels in mice. ANN MICROBIOL 2021; 71:42. [PMID: 34690623 PMCID: PMC8522538 DOI: 10.1186/s13213-021-01655-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
Purpose Subclinical vitamin D (vitD) deficiency enhances the predisposition to a myriad of acute and chronic pathologies in many people worldwide. Due to the scarcity of vitD-rich foods, the consumption of supplements or fortified foods can be required to maintain healthy serum levels of 25-hydroxyvitamin D [25(OH)D], and the major circulating form of vitD that is commonly measured in serum to determine the vitD status. Since the vitD absorption seems to resemble that of lipids, improved emulsification in the gut could favor vitD permeation through the enterocyte membrane. Contextually, we hypothesized that a microorganism with cholecalciferol (vitD3)-solubilization properties may potentially result in enhanced serum vitD levels. Methods and results Six probiotic strains were screened for their ability to create a stable suspension of vitD3 in water: Lacticaseibacillus paracasei DG, L. paracasei LPC-S01, L. paracasei Shirota, L. rhamnosus GG, Limosilactobacillus reuteri DSM 17938, and Lactobacillus acidophilus LA5. The DG strain displayed the strongest vitD3 solubilization ability and, consequently, were used in an in vivo trial where a commercial preparation of vitD3 in refined olive oil was administered by gavage to CD-1 mice with or without the concurrent administration of L. paracasei DG. ELISA measurements showed that the DG strain significantly increased the serum levels of 25(OH) D when administered once a day for 1 week in association with the vitD3 supplement. Conclusion This preliminary pre-clinical study suggests that the combined administration of L. paracasei DG with an oil-based cholecalciferol supplement could contribute to the maintenance of the adequate 25(OH) D serum levels in people at risk of vitD deficiency.
Collapse
Affiliation(s)
| | - Melania Scarpa
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Paola Brun
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Giulia Bernabe
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Valeria Sagheddu
- AAT-Advanced Analytical Technologies S.r.l., Fiorenzuola d'Arda, Piacenza, Italy
| | - Marina Elli
- AAT-Advanced Analytical Technologies S.r.l., Fiorenzuola d'Arda, Piacenza, Italy
| | | | | | - Simone Guglielmetti
- Division of Food Microbiology and Bioprocesses, Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, 20133 Milan, Italy
| |
Collapse
|
38
|
The Combined Beneficial Effects of Postbiotic Butyrate on Active Vitamin D3-Orchestrated Innate Immunity to Salmonella Colitis. Biomedicines 2021; 9:biomedicines9101296. [PMID: 34680413 PMCID: PMC8533643 DOI: 10.3390/biomedicines9101296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
Salmonella spp. Remains a major public health problem globally. Biomedicine is the cornerstone of modern health care and could be a solution for antibiotic-resistant Salmonellosis. Although postbiotics seem to be an effective treatment in various clinical conditions, their clinical effects on Salmonella colitis have not been reported. Our previous report revealed that active vitamin D attenuates the severity of Salmonella colitis and invasiveness by reducing inflammation and enhancing the production of antimicrobial peptides. Therefore, we investigated the synergistic effects of butyrate, the most studied postbiotic, and active vitamin D on the severity of Salmonella colitis, invasiveness of Salmonella, and host immune responses, as well as its novel mechanisms, using in vitro and in vivo studies. We demonstrated that a combination of butyrate and active vitamin D (1 alpha, 25-dihydroxyvitamin D3) synergically reduced the severity of Salmonella colitis in C57BL/6 mice and reduced cecal inflammatory mIL-6, mIL-8, mTNF-α, and mIL-1β mRNA expression, but enhanced the antimicrobial peptide mhBD-3 mRNA, compared to a single treatment. Additionally, upregulated vitamin D receptor (VDR) plays a critical role in the synergistic effects. This suggests combined benefits of butyrate and active vitamin D on Salmonella colitis through VDR-mediated antibacterial and anti-inflammatory responses. The combined use of both supplements could be a potential biomedicine for infectious and autoimmune colitis.
Collapse
|
39
|
Taverniti V, Cesari V, Gargari G, Rossi U, Biddau C, Lecchi C, Fiore W, Arioli S, Toschi I, Guglielmetti S. Probiotics Modulate Mouse Gut Microbiota and Influence Intestinal Immune and Serotonergic Gene Expression in a Site-Specific Fashion. Front Microbiol 2021; 12:706135. [PMID: 34539604 PMCID: PMC8441017 DOI: 10.3389/fmicb.2021.706135] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Probiotic microorganisms may benefit the host by influencing diverse physiological processes, whose nature and underlying mechanisms are still largely unexplored. Animal models are a unique tool to understand the complexity of the interactions between probiotic microorganisms, the intestinal microbiota, and the host. In this regard, in this pilot study, we compared the effects of 5-day administration of three different probiotic bacterial strains (Bifidobacterium bifidum MIMBb23sg, Lactobacillus helveticus MIMLh5, and Lacticaseibacillus paracasei DG) on three distinct murine intestinal sites (ileum, cecum, and colon). All probiotics preferentially colonized the cecum and colon. In addition, probiotics reduced in the ileum and increased in the cecum and colon the relative abundance of numerous bacterial taxonomic units. MIMBb23sg and DG increased the inducible nitric oxide synthase (iNOS) in the ileum, which is involved in epithelial homeostasis. In addition, MIMBb23sg upregulated cytokine IL-10 in the ileum and downregulated the cyclooxygenase COX-2 in the colon, suggesting an anti-inflammatory/regulatory activity. MIMBb23sg significantly affected the expression of the main gene involved in serotonin synthesis (TPH1) and the gene coding for the serotonin reuptake protein (SERT) in the ileum and colon, suggesting a potential propulsive effect toward the distal part of the gut, whereas the impact of MIMLh5 and DG on serotonergic genes suggested an effect toward motility control. The three probiotics decreased the expression of the permeability marker zonulin in gut distal sites. This preliminary in vivo study demonstrated the safety of the tested probiotic strains and their common ability to modulate the intestinal microbiota. The probiotics affected host gene expression in a strain-specific manner. Notably, the observed effects in the gut were site dependent. This study provides a rationale for investigating the effects of probiotics on the serotonergic system, which is a topic still widely unexplored.
Collapse
Affiliation(s)
- Valentina Taverniti
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | - Valentina Cesari
- Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giorgio Gargari
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | - Umberto Rossi
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Biddau
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Stefania Arioli
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| | - Ivan Toschi
- Department of Agricultural and Environmental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Simone Guglielmetti
- Department of Food, Environmental and Nutritional Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
40
|
Hossain MI, Mizan MFR, Roy PK, Nahar S, Toushik SH, Ashrafudoulla M, Jahid IK, Lee J, Ha SD. Listeria monocytogenes biofilm inhibition on food contact surfaces by application of postbiotics from Lactobacillus curvatus B.67 and Lactobacillus plantarum M.2. Food Res Int 2021; 148:110595. [PMID: 34507740 DOI: 10.1016/j.foodres.2021.110595] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Owing to their preservative and antimicrobial effects, postbiotics (metabolic byproducts of probiotics) are promising natural components for the food industry. Therefore, the present study aimed to investigate the efficacy of postbiotics collected from isolated Lactobacillus curvatus B.67 and Lactobacillus plantarum M.2 against Listeria monocytogenes pathogens in planktonic cells, motility, and biofilm states. The analysis of the metabolite composition of the postbiotics revealed various organic acids, along with a few well-known bacteriocin-encoding genes with potential antimicrobial effects. Postbiotics maintained their residual antimicrobial activity over the pH range 1-6 but lost all activity at neutral pH (pH 7). Full antimicrobial activity (100%) was observed during heat treatment, even under the autoclaving condition.Minimum inhibitory concentration (MICs) of L. curvatus B.67 and L. plantarum M.2 against L. monocytogenes were 80 and 70 mg/mL, respectively. However, four sub-MICs of the postbiotics (1/2, 1/4, 1/8, and 1/16 MIC) were tested for inhibition efficacy against L. monocytogenes during different experiment in this study. Swimming motility, biofilm formation, and expression levels of target genes related to biofilm formation, virulence, and quorum-sensing were significantly inhibited with increasing postbiotics concentration. Postbiotics from L. plantarum M.2 exhibited a higher inhibitory effect than the postbiotics from L. curvatus B.67. Nonetheless, both these postbiotics from Lactobacillus spp. could be used as effective bio-interventions for controlling L. monocytogenes biofilm in the food industry.
Collapse
Affiliation(s)
- Md Iqbal Hossain
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Md Furkanur Rahaman Mizan
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Pantu Kumar Roy
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Shamsun Nahar
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Sazzad Hossen Toushik
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Md Ashrafudoulla
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea
| | - Iqbal Kabir Jahid
- Department of Microbiology, Jashore University of Science and Technology, Bangladesh
| | - Jihyun Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Sang-Do Ha
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, 72-1 Nae-Ri, Daedeok-Myun, Anseong, Gyunggido 456-756, Republic of Korea.
| |
Collapse
|
41
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Effects of a Postbiotic and Prebiotic Mixture on Suckling Rats' Microbiota and Immunity. Nutrients 2021; 13:2975. [PMID: 34578853 PMCID: PMC8469903 DOI: 10.3390/nu13092975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023] Open
Abstract
Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
42
|
Probiotics, Prebiotics, and Synbiotics in the Irritable Bowel Syndrome Treatment: A Review. Biomolecules 2021; 11:biom11081154. [PMID: 34439821 PMCID: PMC8412098 DOI: 10.3390/biom11081154] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/02/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome is not a life-threatening disease, yet it significantly affects the quality of life and contributes to economic loss. It is estimated that even up to 45% of the world's population can suffer from the disease. The first attempts to diagnose irritable bowel syndrome were made at the end of the 19th century; however, establishing appropriate diagnostic criteria and treatment methods is still ongoing. To date, little is known about the etiology of irritable bowel syndrome; however, growing attention is drawn to the intestinal microbiota as a factor in the disease development. For this reason, researchers have conducted many studies on therapies that modulate the microbiota, among which probiotics, prebiotics, and synbiotics are widely studied. To date, most studies have examined probiotics; however, there are also several studies demonstrating the efficacy of prebiotics and synbiotics. The aim of this review was to summarize findings on the usefulness of probiotics, prebiotics, and synbiotics in the treatment of irritable bowel syndrome.
Collapse
|
43
|
Jäger R, Zaragoza J, Purpura M, Iametti S, Marengo M, Tinsley GM, Anzalone AJ, Oliver JM, Fiore W, Biffi A, Urbina S, Taylor L. Probiotic Administration Increases Amino Acid Absorption from Plant Protein: a Placebo-Controlled, Randomized, Double-Blind, Multicenter, Crossover Study. Probiotics Antimicrob Proteins 2021; 12:1330-1339. [PMID: 32358640 PMCID: PMC7641926 DOI: 10.1007/s12602-020-09656-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The fate of dietary protein in the gut is determined by microbial and host digestion and utilization. Fermentation of proteins generates bioactive molecules that have wide-ranging health effects on the host. The type of protein can affect amino acid absorption, with animal proteins generally being more efficiently absorbed compared with plant proteins. In contrast to animal proteins, most plant proteins, such as pea protein, are incomplete proteins. Pea protein is low in methionine and contains lower amounts of branched-chain amino acids (BCAAs), which play a crucial role in muscle health. We hypothesized that probiotic supplementation results in favorable changes in the gut microbiota, aiding the absorption of amino acids from plant proteins by the host. Fifteen physically active men (24.2 ± 5.0 years; 85.3 ± 12.9 kg; 178.0 ± 7.6 cm; 16.7 ± 5.8% body fat) co-ingested 20 g of pea protein with either AminoAlta™, a multi-strain probiotic (5 billion CFU L. paracasei LP-DG® (CNCM I-1572) plus 5 billion CFU L. paracasei LPC-S01 (DSM 26760), SOFAR S.p.A., Italy) or a placebo for 2 weeks in a randomized, double-blind, crossover design, separated by a 4-week washout period. Blood samples were taken at baseline and at 30-, 60-, 120-, and 180-min post-ingestion and analyzed for amino acid content. Probiotic administration significantly increased methionine, histidine, valine, leucine, isoleucine, tyrosine, total BCAA, and total EAA maximum concentrations (Cmax) and AUC without significantly changing the time to reach maximum concentrations. Probiotic supplementation can be an important nutritional strategy to improve post-prandial changes in blood amino acids and to overcome compositional shortcomings of plant proteins. ClinicalTrials.gov Identifier: ISRCTN38903788.
Collapse
Affiliation(s)
| | - Javier Zaragoza
- Human Performance Laboratory, School of Exercise & Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | | | - Stefania Iametti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Mauro Marengo
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Grant M Tinsley
- Energy Balance & Body Composition Laboratory, Texas Tech University, Lubbock, TX, USA
| | | | | | | | | | - Stacie Urbina
- Human Performance Laboratory, School of Exercise & Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Lem Taylor
- Human Performance Laboratory, School of Exercise & Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| |
Collapse
|
44
|
Abstract
Epidemiologic data support that acute gastrointestinal infection is one of the strongest risk factors for development of irritable bowel syndrome (IBS). Risk of post-infection IBS (PI-IBS) seems to be greater with bacterial and protozoal than viral enterocolitis. Younger individuals, women, and those with severe enterocolitis are more likely to develop PI-IBS. Disease mechanisms in animal models and humans involve chronic perturbation of intestinal microbiome, epithelial and neuronal remodeling, and immune activation. These mechanisms can lead to luminal (increased proteolytic activity, altered bile acid composition) and physiologic (increased permeability, transit changes, and visceral hypersensitivity) alterations that can mediate PI-IBS symptoms.
Collapse
Affiliation(s)
- Antonio Berumen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Adam L Edwinson
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA; Department of Medicine and Physiology, Enteric NeuroScience Program, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
45
|
Person H, Keefer L. Psychological comorbidity in gastrointestinal diseases: Update on the brain-gut-microbiome axis. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110209. [PMID: 33326819 PMCID: PMC8382262 DOI: 10.1016/j.pnpbp.2020.110209] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
The high comorbidity of psychological disorders in both functional and organic gastrointestinal diseases suggests the intimate and complex link between the brain and the gut. Termed the brain-gut axis, this bidirectional communication between the central nervous system and enteric nervous system relies on immune, endocrine, neural, and metabolic pathways. There is increasing evidence that the gut microbiome is a key part of this system, and dysregulation of the brain-gut-microbiome axis (BGMA) has been implicated in disorders of brain-gut interaction, including irritable bowel syndrome, and in neuropsychiatric disorders, including depression, Alzheimer's disease, and autism spectrum disorder. Further, alterations in the gut microbiome have been implicated in the pathogenesis of organic gastrointestinal diseases, including inflammatory bowel disease. The BGMA is an attractive therapeutic target, as using prebiotics, probiotics, or postbiotics to modify the gut microbiome or mimic gut microbial signals could provide novel treatment options to address these debilitating diseases. However, despite significant advancements in our understanding of the BGMA, clinical data is lacking. In this article, we will review current understanding of the comorbidity of gastrointestinal diseases and psychological disorders. We will also review the current evidence supporting the key role of the BGMA in this pathology. Finally, we will discuss the clinical implications of the BGMA in the evaluation and management of psychological and gastrointestinal disorders.
Collapse
Affiliation(s)
- Hannibal Person
- Division of Pediatric Gastroenterology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Laurie Keefer
- Division of Pediatric Gastroenterology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
46
|
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: 19] [Impact Index Per Article: 6.3] [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
|
47
|
Cristofori F, Dargenio VN, Dargenio C, Miniello VL, Barone M, Francavilla R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front Immunol 2021; 12:578386. [PMID: 33717063 PMCID: PMC7953067 DOI: 10.3389/fimmu.2021.578386] [Citation(s) in RCA: 304] [Impact Index Per Article: 101.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/14/2021] [Indexed: 02/05/2023] Open
Abstract
Hosting millions of microorganisms, the digestive tract is the primary and most important part of bacterial colonization. On one side, in cases of opportunistic invasion, the abundant bacterial population inside intestinal tissues may face potential health problems such as inflammation and infections. Therefore, the immune system has evolved to sustain the host-microbiota symbiotic relationship. On the other hand, to maintain host immune homeostasis, the intestinal microflora often exerts an immunoregulatory function that cannot be ignored. A field of great interest is the association of either microbiota or probiotics with the immune system concerning clinical uses. This microbial community regulates some of the host's metabolic and physiological functions and drives early-life immune system maturation, contributing to their homeostasis throughout life. Changes in gut microbiota can occur through modification in function, composition (dysbiosis), or microbiota-host interplays. Studies on animals and humans show that probiotics can have a pivotal effect on the modulation of immune and inflammatory mechanisms; however, the precise mechanisms have not yet been well defined. Diet, age, BMI (body mass index), medications, and stress may confound the benefits of probiotic intake. In addition to host gut functions (permeability and physiology), all these agents have profound implications for the gut microbiome composition. The use of probiotics could improve the gut microbial population, increase mucus-secretion, and prevent the destruction of tight junction proteins by decreasing the number of lipopolysaccharides (LPSs). When LPS binds endothelial cells to toll-like receptors (TLR 2, 4), dendritic cells and macrophage cells are activated, and inflammatory markers are increased. Furthermore, a decrease in gut dysbiosis and intestinal leakage after probiotic therapy may minimize the development of inflammatory biomarkers and blunt unnecessary activation of the immune system. In turn, probiotics improve the differentiation of T-cells against Th2 and development of Th2 cytokines such as IL-4 and IL-10. The present narrative review explores the interactions between gut microflora/probiotics and the immune system starting from the general perspective of a biological plausibility to get to the in vitro and in vivo demonstrations of a probiotic-based approach up to the possible uses for novel therapeutic strategies.
Collapse
Affiliation(s)
- Fernanda Cristofori
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vanessa Nadia Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Costantino Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vito Leonardo Miniello
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Michele Barone
- Gastroenterology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Ruggiero Francavilla
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
48
|
Cai T, Gallelli L, Cione E, Perletti G, Ciarleglio F, Malossini G, De Pretis G, Palmieri A, Mirone V, Bartoletti R, Johansen TEB. The use of Lactobacillus casei DG® prevents symptomatic episodes and reduces the antibiotic use in patients affected by chronic bacterial prostatitis: results from a phase IV study. World J Urol 2021; 39:3433-3440. [PMID: 33442769 PMCID: PMC8510959 DOI: 10.1007/s00345-020-03580-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose To evaluate the efficacy of Lactobacillus paracasei CNCM I-1572 (L. casei DG®) in both prevention of symptomatic recurrences and improvement of quality of life in patients with chronic bacterial prostatitis (CBP). Methods Patients with CBP attending a single Urological Institution were enrolled in this phase IV study. At enrollment, all patients were treated with antibiotics in agreement with EAU guidelines and then were treated with L. casei DG® (2 capsules/day for 3 months). Clinical and microbiological analyses were carried out before (enrollment, T0) and 6 months (T2) after the treatment. Both safety and adherence to the treatment were evaluated 3 months (T1) after the enrollment. NIH Chronic Prostatitis Symptom Index (CPSI), International Prostate Symptom Score (IPSS) and Quality of Well-Being (QoL) questionnaires were used. The outcome measures were the rate of symptomatic recurrence, changes in questionnaire symptom scores and the reduction of antibiotic use. Results Eighty-four patients were included. At T2, 61 patients (72.6%) reported a clinical improvement of symptoms with a return to their clinical status before symptoms. A time dependent improvement in clinical symptoms with significant changes in NIH-CPSI, IPSS and QoL (mean difference T2 vs T0: 16.5 ± 3.58; − 11.0 ± 4.32; + 0.3 ± 0.09; p < 0.001), was reported. We recorded that L. casei DG® treatment induced a statistically significant decrease in both (p < 0.001) symptomatic recurrence [1.9/3 months vs 0.5/3 months] and antibiotic use [− 7938 UDD]. No clinically relevant adverse effects were reported. Conclusions L. casei DG® prevents symptomatic recurrences and improves the quality of life in patients with CBP, reducing the antibiotic use.
Collapse
Affiliation(s)
- Tommaso Cai
- Department of Urology, Santa Chiara Regional Hospital, Trento, Italy. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Luca Gallelli
- Department of Health Sciences, University of Catanzaro, Catanzaro, Italy.,Clinical Pharmacology and Pharmacovigilance Unit, Mater Domini Hospital, Catanzaro, Italy
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, Department of Excellence 2018-2022, University of Calabria, 87036, Rende, CS, Italy
| | - Gianpaolo Perletti
- Department of Biotechnology and Life Sciences, Università degli Studi dell'Insubria, Varese, Italy.,Department of Human Structure and Repair, Faculty of Medicine and Medical Sciences, Ghent University, Ghent, Belgium
| | | | - Gianni Malossini
- Department of Urology, Santa Chiara Regional Hospital, Trento, Italy
| | - Giovanni De Pretis
- Department of Gastroenterology, Santa Chiara Regional Hospital, Trento, Italy
| | | | - Vincenzo Mirone
- AOU Federico II - Urology Unit, University Federico II, Naples, Italy
| | | | - Truls E Bjerklund Johansen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Urology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Aarhus, Aarhus, Denmark
| |
Collapse
|
49
|
Mars RAT, Frith M, Kashyap PC. Functional Gastrointestinal Disorders and the Microbiome-What Is the Best Strategy for Moving Microbiome-based Therapies for Functional Gastrointestinal Disorders into the Clinic? Gastroenterology 2021; 160:538-555. [PMID: 33253687 PMCID: PMC8575137 DOI: 10.1053/j.gastro.2020.10.058] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
There have been numerous human studies reporting associations between the intestinal microbiome and functional gastrointestinal disorders (FGIDs), and independently animal studies have explored microbiome-driven mechanisms underlying FGIDs. However, there is often a disconnect between human and animal studies, which hampers translation of microbiome findings to the clinic. Changes in the microbiota composition of patients with FGIDs are generally subtle, whereas changes in microbial function, reflected in the fecal metabolome, appear to be more precise indicators of disease subtype-specific mechanisms. Although we have made significant progress in characterizing the microbiome, to effectively translate microbiome science in a timely manner, we need concurrent and iterative longitudinal studies in humans and animals to determine the precise microbial functions that can be targeted to address specific pathophysiological processes in FGIDs. A systems approach integrating multiple data layers rather than evaluating individual data layers of symptoms, physiological changes, or -omics data in isolation will allow for validation of mechanistic insights from animal studies while also allowing new discovery. Patient stratification for clinical trials based on functional microbiome alterations and/or pathophysiological measurements may allow for more accurate determination of efficacy of individual microbiome-targeted interventions designed to correct an underlying abnormality. In this review, we outline current approaches and knowledge, and identify gaps, to provide a potential roadmap for accelerating translation of microbiome science toward microbiome-targeted personalized treatments for FGIDs.
Collapse
Affiliation(s)
- Ruben A T Mars
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Mary Frith
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
50
|
Hernández-Granados MJ, Franco-Robles E. Postbiotics in human health: Possible new functional ingredients? Food Res Int 2020; 137:109660. [DOI: 10.1016/j.foodres.2020.109660] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023]
|