1
|
Girija S, Kolluri G, Tyagi JS, Kurva SK, Sahoo PR, Srivastava V, Singh T. Postbiotics: The Dynamic Biomolecules in Poultry Nutrition, Health, and Production. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10548-9. [PMID: 40358876 DOI: 10.1007/s12602-025-10548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
The interest in employing cell components as well as metabolites obtained from varied probiotic strains is increasing because of the growing concern regarding safety issues related to live microbial cells. Numerous benefits over conventional probiotics will facilitate the implementation of postbiotics as feed supplements in poultry nutrition. The beneficial characteristics, such as immunomodulation and antibacterial action, point to the possibility of postbiotics improving host health by altering the physiology of the host and ameliorating the disease condition in poultry. As we proceed forward with the ultimate aim of lowering antibiotic use in poultry, maximising performance and maintaining poultry production will be reliant on the best mixes of diverse alternatives as postbiotics, together with appropriate farm management practices. Trials with emphasis on validating the health claims made by these bioactive compounds are highly necessary. Furthermore, the employment of postbiotics under challenging conditions in poultry needs to be explored. This review focuses on highlighting the aspects about postbiotics and its various biological effects on meat and egg production in the poultry industry.
Collapse
Affiliation(s)
- Simi Girija
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Gautham Kolluri
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India.
| | - Jagbir Singh Tyagi
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Shiva Kumar Kurva
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Pravas Ranjan Sahoo
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Vivek Srivastava
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| | - Twinkle Singh
- Division of Avian Physiology and Reproduction, ICAR-Central Avian Research Institute, Izatnagar, 243122, Bareilly, U.P, India
| |
Collapse
|
2
|
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: 10] [Impact Index Per Article: 10.0] [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
|
3
|
Sepordeh S, Jafari AM, Bazzaz S, Abbasi A, Aslani R, Houshmandi S, Rad AH. Postbiotic as Novel Alternative Agent or Adjuvant for the Common Antibiotic Utilized in the Food Industry. Curr Pharm Biotechnol 2024; 25:1245-1263. [PMID: 37702234 DOI: 10.2174/1389201025666230912123849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 07/11/2023] [Accepted: 07/27/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Antibiotic resistance is a serious public health problem as it causes previously manageable diseases to become deadly infections that can cause serious disability or even death. Scientists are creating novel approaches and procedures that are essential for the treatment of infections and limiting the improper use of antibiotics in an effort to counter this rising risk. OBJECTIVES With a focus on the numerous postbiotic metabolites formed from the beneficial gut microorganisms, their potential antimicrobial actions, and recent associated advancements in the food and medical areas, this review presents an overview of the emerging ways to prevent antibiotic resistance. RESULTS Presently, scientific literature confirms that plant-derived antimicrobials, RNA therapy, fecal microbiota transplantation, vaccines, nanoantibiotics, haemofiltration, predatory bacteria, immunotherapeutics, quorum-sensing inhibitors, phage therapies, and probiotics can be considered natural and efficient antibiotic alternative candidates. The investigations on appropriate probiotic strains have led to the characterization of specific metabolic byproducts of probiotics named postbiotics. Based on preclinical and clinical studies, postbiotics with their unique characteristics in terms of clinical (safe origin, without the potential spread of antibiotic resistance genes, unique and multiple antimicrobial action mechanisms), technological (stability and feasibility of largescale production), and economic (low production costs) aspects can be used as a novel alternative agent or adjuvant for the common antibiotics utilized in the production of animal-based foods. CONCLUSION Postbiotic constituents may be a new approach for utilization in the pharmaceutical and food sectors for developing therapeutic treatments. Further metabolomics investigations are required to describe novel postbiotics and clinical trials are also required to define the sufficient dose and optimum administration frequency of postbiotics.
Collapse
Affiliation(s)
- Sama Sepordeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sara Bazzaz
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Abbasi
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramin Aslani
- Food Safety and Hygiene Division, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sousan Houshmandi
- Department of Midwifery, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Xia Q, Lei Y, Wang J, Wang Q. Probiotic management and inflammatory factors as a novel treatment in cirrhosis: A systematic review and meta-analysis. Open Life Sci 2023; 18:20220741. [PMID: 37872967 PMCID: PMC10590617 DOI: 10.1515/biol-2022-0741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 10/25/2023] Open
Abstract
The interaction between intestinal microecological dysregulation, altered inflammatory factors, and cirrhosis is unclear. The aim of this systematic review and meta-analysis was to synthesize the results of previous studies to assess the efficacy of probiotics in the treatment of cirrhosis and their effect on inflammatory factors, as well as to explore the relationship between gut microecological dysregulation and liver disease to gain a deeper understanding of this interaction. Up to December 2022, eligible studies were identified by searching the following databases: National Knowledge Infrastructure (CNKI), Wanfang Data, Web of Science, PubMed, Embase, Medline, and the Cochrane Library. Statistical analysis was performed using software RevMan Version 5.4. A total of 33 eligible randomized controlled trials were included in the study, and data on probiotic strains, duration of intervention, measures in the control group, and outcomes were extracted and evaluated. Compared to the control group, the experimental group had significant improvements in overall efficacy. The results of the meta-analysis revealed that probiotic use significantly decreased biochemical parameters for liver function, including aspartate transaminase, alanine aminotransferase, and total bilirubin. Similar result was obtained in interleukin-6, tumor necrosis factor-α, and endotoxin. However, probiotic intervention did not significantly affect interleukin-2 and interleukin-10. The current meta-analysis illustrates that probiotic supplementation reduces inflammatory markers and biochemical parameters for liver function in patients with cirrhosis, suggesting that probiotic management may be a novel treatment for cirrhosis. Furthermore, the interaction of the gut microbiota, associated metabolites, and inflammation factors with cirrhosis may provide a promising therapeutic target for the pharmacological and clinical treatment of cirrhosis.
Collapse
Affiliation(s)
- Qinglan Xia
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan430065, China
| | - Yumeng Lei
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan430065, China
| | - Jiadun Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan430065, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan430065, China
- Asia General Hospital Affiliated to Wuhan University of Science and Technology, Wuhan430056, China
| |
Collapse
|
5
|
Ma B, Gavzy SJ, Saxena V, Song Y, Piao W, Lwin HW, Lakhan R, Iyyathurai J, Li L, France M, Paluskievicz C, Shirkey MW, Hittle L, Munawwar A, Mongodin EF, Bromberg JS. Strain-specific alterations in gut microbiome and host immune responses elicited by tolerogenic Bifidobacterium pseudolongum. Sci Rep 2023; 13:1023. [PMID: 36658194 PMCID: PMC9852428 DOI: 10.1038/s41598-023-27706-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
The beneficial effects attributed to Bifidobacterium are largely attributed to their immunomodulatory capabilities, which are likely to be species- and even strain-specific. However, their strain-specificity in direct and indirect immune modulation remain largely uncharacterized. We have shown that B. pseudolongum UMB-MBP-01, a murine isolate strain, is capable of suppressing inflammation and reducing fibrosis in vivo. To ascertain the mechanism driving this activity and to determine if it is specific to UMB-MBP-01, we compared it to a porcine tropic strain B. pseudolongum ATCC25526 using a combination of cell culture and in vivo experimentation and comparative genomics approaches. Despite many shared features, we demonstrate that these two strains possess distinct genetic repertoires in carbohydrate assimilation, differential activation signatures and cytokine responses signatures in innate immune cells, and differential effects on lymph node morphology with unique local and systemic leukocyte distribution. Importantly, the administration of each B. pseudolongum strain resulted in major divergence in the structure, composition, and function of gut microbiota. This was accompanied by markedly different changes in intestinal transcriptional activities, suggesting strain-specific modulation of the endogenous gut microbiota as a key to immune modulatory host responses. Our study demonstrated a single probiotic strain can influence local, regional, and systemic immunity through both innate and adaptive pathways in a strain-specific manner. It highlights the importance to investigate both the endogenous gut microbiome and the intestinal responses in response to probiotic supplementation, which underpins the mechanisms through which the probiotic strains drive the strain-specific effect to impact health outcomes.
Collapse
Affiliation(s)
- Bing Ma
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| | - Samuel J Gavzy
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yang Song
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Wenji Piao
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hnin Wai Lwin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ram Lakhan
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Jegan Iyyathurai
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Michael France
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christina Paluskievicz
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Marina W Shirkey
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Lauren Hittle
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Arshi Munawwar
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Emmanuel F Mongodin
- Institute of Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Lung Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jonathan S Bromberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
- Department of Surgery, University of Maryland Medical Center, Baltimore, MD, 21201, USA.
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
6
|
Mohseni AH, Casolaro V, Bermúdez-Humarán LG, Keyvani H, Taghinezhad-S S. Modulation of the PI3K/Akt/mTOR signaling pathway by probiotics as a fruitful target for orchestrating the immune response. Gut Microbes 2022; 13:1-17. [PMID: 33615993 PMCID: PMC7899637 DOI: 10.1080/19490976.2021.1886844] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) and the phosphatidylinositol-3-kinase (PI3K)/protein kinase B or Akt (PKB/Akt) signaling pathways are considered as two but somewhat interconnected significant immune pathways which play complex roles in a variety of physiological processes as well as pathological conditions. Aberrant activation of PI3K/Akt/mTOR signaling pathways has been reported to be associated in a wide variety of human diseases. Over the past few years, growing evidence in in vitro and in vivo models suggest that this sophisticated and subtle cascade mediates the orchestration of the immune response in health and disease through exposure to probiotics. An expanding body of literature has highlighted the contribution of probiotics and PI3K/Akt/mTOR signaling pathways in gastrointestinal disorders, metabolic syndrome, skin diseases, allergy, salmonella infection, and aging. However, longitudinal human studies are possibly required to verify more conclusively whether the investigational tools used to understand the regulation of these pathways might provide effective approaches in the prevention and treatment of various disorders. In this Review, we summarize the experimental evidence from recent peer-reviewed studies and provide a brief overview of the causal relationship between the effects of probiotics and their metabolites on the components of PI3K/Akt/mTOR signaling pathways and human disease.
Collapse
Affiliation(s)
- Amir Hossein Mohseni
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Salerno, Italy
| | | | - Hossein Keyvani
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran,Hossein Keyvani Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran, Tel +98 21 88715350
| | - Sedigheh Taghinezhad-S
- Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran,CONTACT Sedigheh Taghinezhad-S Department of Microbiology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, 1477893855, Iran
| |
Collapse
|
7
|
Basir L, Moghimipour E, Saadatzadeh A, Cheraghian B, Khanehmasjedi S. Effect of postbiotic-toothpaste on salivary levels of IgA in 6- to 12-year-old children: Study protocol for a randomized triple-blind placebo-controlled trial. Front Pediatr 2022; 10:1042973. [PMID: 36578663 PMCID: PMC9790979 DOI: 10.3389/fped.2022.1042973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Children in mixed dentition are highly at risk for dental caries, which is a major health issue worldwide. Despite their effect in controlling dental caries, using probiotics can be challenging. Therefore, it has been advised to use their inanimate forms, called postbiotics. We hypothesize that postbiotics can enhance the oral immunity. METHODS The aim of this triple-blind, randomized, placebo-controlled trial is to investigate the effect of postbiotic-toothpaste (Bifidobacterium animalis subsp. animalis) on salivary levels of Immunoglobulin A (IgA) and pH in children. Using comparing two means formula to calculate the sample size, for this trial 80 healthy 6- to 12-year-old children during mixed dentition with no cavitated dental caries will be selected by convenience sampling method and randomly allocated to two groups, postbiotic-toothpaste or placebo-toothpaste. Saliva samples will be gathered at baseline and four weeks after the intervention. The level of salivary IgA will be determined by ELISA and salivary pH will be measured using a pH meter. Data will be compared within and between groups using independent t-test and paired t-test, in case of normality, with a p < 0.05 as statistically significant. DISCUSSION If postbiotics-toothpaste prove to be effective in improving the oral immunity, they can be used to prevent dental caries and other oral diseases. The result of this study can help researchers who are working on the immunomodulatory effects of postbiotics in children. TRIAL REGISTRATION NUMBER Iranian Registry of Clinical Trials (IRCT), IRCT20191016045128N2. Registered on 7 March 2022.
Collapse
Affiliation(s)
- Leila Basir
- Department of Pediatric Dentistry, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Eskandar Moghimipour
- Department of Pharmaceutics, School of Pharmacy, Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afrooz Saadatzadeh
- Department of Food and Drug Control, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Cheraghian
- Department of Biostatistics and Epidemiology, School of Public Health, Alimentary Tract Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samaneh Khanehmasjedi
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Scarpellini E, Rinninella E, Basilico M, Colomier E, Rasetti C, Larussa T, Santori P, Abenavoli L. From Pre- and Probiotics to Post-Biotics: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:37. [PMID: 35010297 PMCID: PMC8750841 DOI: 10.3390/ijerph19010037] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND AIMS gut microbiota (GM) is a complex ecosystem containing bacteria, viruses, fungi, and yeasts. It has several functions in the human body ranging from immunomodulation to metabolic. GM derangement is called dysbiosis and is involved in several host diseases. Pre-, probiotics, and symbiotics (PRE-PRO-SYMB) have been extensively developed and studied for GM re-modulation. Herein, we review the literature data regarding the new concept of postbiotics, starting from PRE-PRO-SYMB. METHODS we conducted a search on the main medical databases for original articles, reviews, meta-analyses, randomized clinical trials, and case series using the following keywords and acronyms and their associations: gut microbiota, prebiotics, probiotics, symbiotic, and postbiotics. RESULTS postbiotics account for PRO components and metabolic products able to beneficially affect host health and GM. The deeper the knowledge about them, the greater their possible uses: the prevention and treatment of atopic, respiratory tract, and inflammatory bowel diseases. CONCLUSIONS better knowledge about postbiotics can be useful for the prevention and treatment of several human body diseases, alone or as an add-on to PRE-PRO-SYMB.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
- TARGID, KU Leuven, 3000 Leuven, Belgium
| | - Emanuele Rinninella
- UOC di Nutrizione Clinica, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica Del Sacro Cuore, 00168 Rome, Italy
| | - Martina Basilico
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | | | - Carlo Rasetti
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Tiziana Larussa
- Department of Health Sciences, Magna Græcia University, 88100 Catanzaro, Italy
| | - Pierangelo Santori
- Internal Medicine Unit, "Madonna del Soccorso" General Hospital, 63074 San Benedetto del Tronto, Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, Magna Græcia University, 88100 Catanzaro, Italy
| |
Collapse
|
9
|
Kaur S, Thukral SK, Kaur P, Samota MK. Perturbations associated with hungry gut microbiome and postbiotic perspectives to strengthen the microbiome health. FUTURE FOODS 2021. [DOI: 10.1016/j.fufo.2021.100043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
10
|
Probiotic-Induced Tolerogenic Dendritic Cells: A Novel Therapy for Inflammatory Bowel Disease? Int J Mol Sci 2021; 22:ijms22158274. [PMID: 34361038 PMCID: PMC8348973 DOI: 10.3390/ijms22158274] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are immune-mediated, chronic relapsing diseases with a rising prevalence worldwide in both adult and pediatric populations. Treatment options for immune-mediated diseases, including IBDs, are traditional steroids, immunomodulators, and biologics, none of which are capable of inducing long-lasting remission in all patients. Dendritic cells (DCs) play a fundamental role in inducing tolerance and regulating T cells and their tolerogenic functions. Hence, modulation of intestinal mucosal immunity by DCs could provide a novel, additional tool for the treatment of IBD. Recent evidence indicates that probiotic bacteria might impact immunomodulation both in vitro and in vivo by regulating DCs’ maturation and producing tolerogenic DCs (tolDCs) which, in turn, might dampen inflammation. In this review, we will discuss this evidence and the mechanisms of action of probiotics and their metabolites in inducing tolDCs in IBDs and some conditions associated with them.
Collapse
|
11
|
Al-Sadi R, Dharmaprakash V, Nighot P, Guo S, Nighot M, Do T, Ma TY. Bifidobacterium bifidum Enhances the Intestinal Epithelial Tight Junction Barrier and Protects against Intestinal Inflammation by Targeting the Toll-like Receptor-2 Pathway in an NF-κB-Independent Manner. Int J Mol Sci 2021; 22:8070. [PMID: 34360835 PMCID: PMC8347470 DOI: 10.3390/ijms22158070] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023] Open
Abstract
Defective intestinal tight junction (TJ) barrier is a hallmark in the pathogenesis of inflammatory bowel disease (IBD). To date, there are no effective therapies that specifically target the intestinal TJ barrier. Among the various probiotic bacteria, Bifidobacterium, is one of the most widely studied to have beneficial effects on the intestinal TJ barrier. The main purpose of this study was to identify Bifidobacterium species that cause a sustained enhancement in the intestinal epithelial TJ barrier and can be used therapeutically to target the intestinal TJ barrier and to protect against or treat intestinal inflammation. Our results showed that Bifidobacterium bifidum caused a marked, sustained enhancement in the intestinal TJ barrier in Caco-2 monolayers. The Bifidobacterium bifidum effect on TJ barrier was strain-specific, and only the strain designated as BB1 caused a maximal enhancement in TJ barrier function. The mechanism of BB1 enhancement of intestinal TJ barrier required live bacterial cell/enterocyte interaction and was mediated by the BB1 attachment to Toll-like receptor-2 (TLR-2) at the apical membrane surface. The BB1 enhancement of the intestinal epithelial TJ barrier function was mediated by the activation of the p38 kinase pathway, but not the NF-κB signaling pathway. Moreover, the BB1 caused a marked enhancement in mouse intestinal TJ barrier in a TLR-2-dependent manner and protected against dextran sodium sulfate (DSS)-induced increase in mouse colonic permeability, and treated the DSS-induced colitis in a TJ barrier-dependent manner. These studies show that probiotic bacteria BB1 causes a strain-specific enhancement of the intestinal TJ barrier through a novel mechanism involving BB1 attachment to the enterocyte TLR-2 receptor complex and activation of p38 kinase pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thomas Y. Ma
- Department of Medicine, Penn State College of Medicine, Hershey Medical Center, Penn State University, Hershey, PA 17033, USA; (R.A.-S.); (V.D.); (P.N.); (S.G.); (M.N.); (T.D.)
| |
Collapse
|
12
|
Xia Q, Chen G, Ren Y, Zheng T, Shen C, Li M, Chen X, Zhai H, Li Z, Xu J, Gu A, Jin M, Fan L. Investigating efficacy of "microbiota modulation of the gut-lung Axis" combined with chemotherapy in patients with advanced NSCLC: study protocol for a multicenter, prospective, double blind, placebo controlled, randomized trial. BMC Cancer 2021; 21:721. [PMID: 34157996 PMCID: PMC8220724 DOI: 10.1186/s12885-021-08448-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/04/2021] [Indexed: 11/28/2022] Open
Abstract
Background Most NSCLCs metastasised out of the lungs at the time of diagnosis and cannot be surgically removed . Cytotoxic chemotherapy drugs have become the main treatment in recent decades, especially in patients with NSCLC without EGFR, ALK, and ROS gene mutations. The prognosis of lung cancer is poor, and the overall 5-year survival rate is only 9–13%. Therefore the treatment of advanced NSCLC remains a significant medical need. Recent studies have shown a significant relationship between the gut-lung axis microecology and malignant tumors. Intestinal probiotics are likely to play a role in inhibiting tumorigenesis through “intestinal-pulmonary axis microecological regulation”. This study will seek to investigate the efficacy of “Microbiota modulation of the Gut-Lung Axis” combined with chemotherapy in patients with advanced NSCLC. Methods The research is a multicenter, prospective, double blind, placebo controlled, randomized trial. Based on the theoretical basis of “intestinal and lung axis microecological adjustment”, combined with traditional platinum-containing two-drug chemotherapy, the efficacy of the new therapy on patients with advanced NSCLC was observed. Collect the basic information of the patient, and study the effect of platinum-based combined chemotherapy on the diversity of intestinal flora in patients with lung cancer after receiving chemotherapy treatment, feces before and after chemotherapy, and the status and extent of adverse reactions during chemotherapy . A total of 180 subjects were included, divided into a control group (platinum-containing dual-drug chemotherapy) and an intervention group (platinum-containing dual-drug chemotherapy combined with Bifico), and were randomly assigned to the group 1:1. Discussion As a result, intestinal-pulmonary microecological balance could become a new target for the treatment of lung cancer. This study explores the combination of intestinal microecological regulation and chemotherapy to provide new treatment strategies and basis for lung cancer patients. It can help prolong the survival time of lung cancer patients and improve the quality of life, thereby generating huge economic and social benefits. The results can be promoted and applied to units engaged in the treatment of lung cancer. Trial registration number NCT03642548, date: August 22, 2018, the first version protocol. The URL of trial registry record: https://clinicaltrials.gov/ct2/show/NCT03642548?term=NCT03642548&draw=2&rank=1.
Collapse
Affiliation(s)
- Qing Xia
- Department of Pulmonary and Critical Care Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Guojie Chen
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Yanbei Ren
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Tiansheng Zheng
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Changxing Shen
- Department of Pulmonary and Critical Care Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Ming Li
- Department of Pulmonary and Critical Care Medicine, Shanghai 10th People's Hospital, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China.
| | - Xiangyun Chen
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Hong Zhai
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Zhuang Li
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China
| | - Jianfang Xu
- Oncology Department, Shanghai Pulmonary Hospital, Tongji University School of Medicine, No.507, Zhengmin Rd, Shanghai, 200433, China
| | - Aiqin Gu
- Oncology Department, Shanghai Chest Hospital, Shanghai Jiaotong University, No.241, West Huaihai Rd, Shanghai, 200030, China
| | - Meiling Jin
- Department of Pulmonary and Critical Care Medicine, Shanghai Zhongshan Hospital, Fudan University School of Medicine, No.180, Fenglin Rd, Shanghai, 200032, China
| | - Lihong Fan
- Institute of Energy Metabolism and Health, Tongji University School of Medicine, No. 301, Middle Yangchang Rd, Shanghai, 200072, China.
| |
Collapse
|
13
|
Żółkiewicz J, Marzec A, Ruszczyński M, Feleszko W. Postbiotics-A Step Beyond Pre- and Probiotics. Nutrients 2020; 12:E2189. [PMID: 32717965 PMCID: PMC7468815 DOI: 10.3390/nu12082189] [Citation(s) in RCA: 354] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
As an imbalance in the intestinal microbiota can lead to the development of several diseases (e.g., type 1 diabetes, cancer, among others), the use of prebiotics, probiotics, and postbiotics to alter the gut microbiome has attracted recent interest. Postbiotics include any substance released by or produced through the metabolic activity of the microorganism, which exerts a beneficial effect on the host, directly or indirectly. As postbiotics do not contain live microorganisms, the risks associated with their intake are minimized. Here, we provided a critical review of postbiotics described in the literature, including their mechanisms of action, clinical characteristics, and potential therapeutic applications. We detailed the pleiotropic effects of postbiotics, including their immunomodulatory, anti-inflammatory, antioxidant, and anti-cancer properties. Although the use of postbiotics is an attractive strategy for altering the microbiome, further study into its efficacy and safety is warranted.
Collapse
Affiliation(s)
- Jakub Żółkiewicz
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| | - Aleksandra Marzec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| | - Marek Ruszczyński
- Department of Paediatrics, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland;
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Żwirki i Wigury 63A, 02-091 Warsaw, Poland; (J.Ż.); (A.M.)
| |
Collapse
|
14
|
Plaza-Diaz J, Ruiz-Ojeda FJ, Gil-Campos M, Gil A. Mechanisms of Action of Probiotics. Adv Nutr 2019; 10:S49-S66. [PMID: 30721959 PMCID: PMC6363529 DOI: 10.1093/advances/nmy063] [Citation(s) in RCA: 654] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/11/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Probiotics are living microorganisms that confer health benefits to the host when administered in adequate amounts; however, dead bacteria and their components can also exhibit probiotic properties. Bifidobacterium and strains of lactic acid bacteria are the most widely used bacteria that exhibit probiotic properties and are included in many functional foods and dietary supplements. Probiotics have been shown to prevent and ameliorate the course of digestive disorders such as acute, nosocomial, and antibiotic-associated diarrhea; allergic disorders such as atopic dermatitis (eczema) and allergic rhinitis in infants; and Clostridium difficile-associated diarrhea and some inflammatory bowel disorders in adults. In addition, probiotics may be of interest as coadjuvants in the treatment of metabolic disorders, including obesity, metabolic syndrome, nonalcoholic fatty liver disease, and type 2 diabetes. However, the mechanisms of action of probiotics, which are diverse, heterogeneous, and strain specific, have received little attention. Thus, the aim of the present work was to review the main mechanisms of action of probiotics, including colonization and normalization of perturbed intestinal microbial communities in children and adults; competitive exclusion of pathogens and bacteriocin production; modulation of fecal enzymatic activities associated with the metabolization of biliary salts and inactivation of carcinogens and other xenobiotics; production of short-chain and branched-chain fatty acids, which, in turn, have wide effects not only in the intestine but also in peripheral tissues via interactions with short-chain fatty acid receptors, modulating mainly tissue insulin sensitivity; cell adhesion and mucin production; modulation of the immune system, which results mainly in the differentiation of T-regulatory cells and upregulation of anti-inflammatory cytokines and growth factors, i.e., interleukin-10 and transforming growth factor; and interaction with the brain-gut axis by regulation of endocrine and neurologic functions. Further research to elucidate the precise molecular mechanisms of action of probiotics is warranted.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Armilla, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Armilla, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Armilla, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Armilla, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - Mercedes Gil-Campos
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Instituto de Salud Carlos III, Madrid, Spain
- Pediatric Research and Metabolism Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research, Cordoba, Spain
| | - Angel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Armilla, Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix,” Biomedical Research Center, University of Granada, Armilla, Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, Granada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Kamiya S, Yonezawa H, Osaki T. Role of Probiotics in Eradication Therapy for Helicobacter pylori Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:243-255. [PMID: 31016634 DOI: 10.1007/5584_2019_369] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Probiotics are defined as, "Live microorganisms that, when administered in adequate amounts, confer a health benefit on the host", and have various effects including inhibitory capabilities on pathogens, stimulation of organ functions and activation of immune responses in the human. Probiotics were reported to inhibit Helicobacter pylori not only in vitro, but also in vivo studies. The mechanisms by which probiotics inhibit H. pylori infection include competition for nutrients, production of bactericidal substances, competitive inhibition of adherence and stimulation of host functions and immunity. In addition, probiotics are clinically used for eradication therapy of H. pylori infection, and the effects of probiotics as single treatment and combination use with other drugs including proton pump inhibitors and antibiotics against H. pylori are reported. It has been testified that probiotics increase the eradication rate and prevent adverse events including diarrhea, nausea, vomiting and taste disorder. In the Maastrich V/Florence Consensus Report 2017, it was stated that some probiotics may have a beneficial effect on H. pylori eradication and are effective in reducing side effects of eradication therapy, but more research is needed to answer several questions concerning the mechanisms of probiotics action. In addition, strain specificity, dosages and duration times of probiotics used for H. pylori eradication therapy need to be clarified in future studies.
Collapse
Affiliation(s)
- Shigeru Kamiya
- Faculty of Health Sciences, Kyorin University, Tokyo, Japan. .,Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan.
| | - Hideo Yonezawa
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| | - Takako Osaki
- Department of Infectious Diseases, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Dicks LMT, Dreyer L, Smith C, van Staden AD. A Review: The Fate of Bacteriocins in the Human Gastro-Intestinal Tract: Do They Cross the Gut-Blood Barrier? Front Microbiol 2018; 9:2297. [PMID: 30323796 PMCID: PMC6173059 DOI: 10.3389/fmicb.2018.02297] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022] Open
Abstract
The intestinal barrier, consisting of the vascular endothelium, epithelial cell lining, and mucus layer, covers a surface of about 400 m2. The integrity of the gut wall is sustained by transcellular proteins forming tight junctions between the epithelial cells. Protected by three layers of mucin, the gut wall forms a non-permeable barrier, keeping digestive enzymes and microorganisms within the luminal space, separate from the blood stream. Microorganisms colonizing the gut may produce bacteriocins in an attempt to outcompete pathogens. Production of bacteriocins in a harsh and complex environment such as the gastro-intestinal tract (GIT) may be below minimal inhibitory concentration (MIC) levels. At such low levels, the stability of bacteriocins may be compromised. Despite this, most bacteria in the gut have the ability to produce bacteriocins, distributed throughout the GIT. With most antimicrobial studies being performed in vitro, we know little about the migration of bacteriocins across epithelial barriers. The behavior of bacteriocins in the GIT is studied ex vivo, using models, flow cells, or membranes resembling the gut wall. Furthermore, little is known about the effect bacteriocins have on the immune system. It is generally believed that the peptides will be destroyed by macrophages once they cross the gut wall. Studies done on the survival of neurotherapeutic peptides and their crossing of the brain-blood barrier, along with other studies on small peptides intravenously injected, may provide some answers. In this review, the stability of bacteriocins in the GIT, their effect on gut epithelial cells, and their ability to cross epithelial cells are discussed. These are important questions to address in the light of recent papers advocating the use of bacteriocins as possible alternatives to, or used in combination with, antibiotics.
Collapse
Affiliation(s)
- Leon M. T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leané Dreyer
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Anton D. van Staden
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
17
|
Vinusha KS, Deepika K, Johnson TS, Agrawal GK, Rakwal R. Proteomic studies on lactic acid bacteria: A review. Biochem Biophys Rep 2018; 14:140-148. [PMID: 29872746 PMCID: PMC5986552 DOI: 10.1016/j.bbrep.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/02/2018] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Probiotics are amongst the most common microbes in the gastro-intestinal tract of humans and other animals. Prominent among probiotics are Lactobacillus and Bifidobacterium. They offer wide-ranging health promoting benefits to the host which include reduction in pathological alterations, stimulation of mucosal immunity and interaction with mediators of inflammation among others. Proteomics plays a vital role in understanding biological functions of a cell. Proteomics is also slowly and steadily adding to the existing knowledge on role of probiotics. In this paper, the proteomics of probiotics, with special reference to lactic acid bacteria is reviewed with a view to understand i) proteome map, ii) mechanism of adaptation to harsh gut environment such as low pH and bile acid, iii) role of cell surface proteins in adhering to intestinal epithelial cells, and iv) as a tool to answer basic cell functions. We have also reviewed various analytical methods used to carry out proteome analysis, in which 2D-MS and LC-MS/MS approaches were found to be versatile methods to perform high-throughput sample analyses even for a complex gut samples. Further, we present future road map of understanding gut microbes combining meta-proteomics, meta-genomics, meta-transcriptomics and -metabolomics.
Collapse
Affiliation(s)
- K Sri Vinusha
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - K Deepika
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - T Sudhakar Johnson
- Department of Biotechnology, K. L. E. F. deemed University, Guntur District, Vaddeswaram, Andhra Pradesh 522502, India
| | - Ganesh K Agrawal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal
| | - Randeep Rakwal
- Research Laboratory for Biotechnology and Biochemistry (RLABB), GPO Box 13265, Kathmandu, Nepal.,GRADE Academy Private Limited, Adarsh Nagar-13, Birgunj, Nepal.,Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan.,Global Research Center for Innovative Life Science, Peptide Drug Innovation, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 4-41 Ebara 2-chome, Shinagawa, Tokyo 142-8501, Japan
| |
Collapse
|
18
|
Effect of synbiotic supplementation in children and adolescents with cystic fibrosis: a randomized controlled clinical trial. Eur J Clin Nutr 2017; 72:736-743. [PMID: 29277839 DOI: 10.1038/s41430-017-0043-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 10/02/2017] [Accepted: 10/17/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND/OBJECTIVES Cystic fibrosis (CF) is characterized by excessive activation of immune processes. The aim of this study was to evaluate the effect of synbiotic supplementation on the inflammatory response in children/adolescents with CF. SUBJECTS/METHODS A randomized, placebo-controlled, double-blind, clinical-trial was conducted with control group (CG, n = 17), placebo-CF-group (PCFG, n = 19), synbiotic CF-group (SCFG, n = 22), PCFG negative (n = 8) and positive (n = 11) bacteriology, and SCFG negative (n = 12) and positive (n = 10) bacteriology. Markers of lung function (FEV1), nutritional status [body mass index-for age (BMI/A), height-for-age (H/A), weight-for-age (W/A), upper-arm fat area (UFA), upper-arm muscle area (UMA), body fat (%BF)], and inflammation [interleukin (IL)-12, tumor necrosis factor-alpha (TNF-α), IL-10, IL-6, IL-1β, IL-8, myeloperoxidase (MPO), nitric oxide metabolites (NOx)] were evaluated before and after 90-day of supplementation with a synbiotic. RESULTS No significance difference was found between the baseline and end evaluations of FEV1 and nutricional status markers. A significant interaction (time vs. group) was found for IL-12 (p = 0.010) and myeloperoxidase (p = 0.036) between PCFG and SCFG, however, the difference was not maintained after assessing the groups individually. NOx diminished significantly after supplementation in the SCFG (p = 0.030). In the SCFG with positive bacteriology, reductions were found in IL-6 (p = 0.033) and IL-8 (p = 0.009) after supplementation. CONCLUSIONS Synbiotic supplementation shown promise at diminishing the pro-inflammatory markers IL-6, IL-8 in the SCFG with positive bacteriology and NOx in the SCFG in children/adolescents with CF.
Collapse
|
19
|
Ruiz L, Delgado S, Ruas-Madiedo P, Sánchez B, Margolles A. Bifidobacteria and Their Molecular Communication with the Immune System. Front Microbiol 2017; 8:2345. [PMID: 29255450 PMCID: PMC5722804 DOI: 10.3389/fmicb.2017.02345] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/15/2017] [Indexed: 12/16/2022] Open
Abstract
Bifidobacterium represents a genus within the phylum Actinobacteria which is one of the major phyla in the healthy intestinal tract of humans. Bifidobacterium is one of the most abundant genera in adults, but its predominance is even more pronounced in infants, especially during lactation, when they can constitute the majority of the total bacterial population. They are one of the pioneering colonizers of the early gut microbiota, and they are known to play important roles in the metabolism of dietary components, otherwise indigestible in the upper parts of the intestine, and in the maturation of the immune system. Bifidobacteria have been shown to interact with human immune cells and to modulate specific pathways, involving innate and adaptive immune processes. In this mini-review, we provide an overview of the current knowledge on the immunomodulatory properties of bifidobacteria and the mechanisms and molecular players underlying these processes, focusing on the corresponding implications for human health. We deal with in vitro models suitable for studying strain-specific immunomodulatory activities. These include peripheral blood mononuclear cells and T cell-mediated immune responses, both effector and regulatory cell responses, as well as the modulation of the phenotype of dendritic cells, among others. Furthermore, preclinical studies, mainly germ-free, gnotobiotic, and conventional murine models, and human clinical trials, are also discussed. Finally, we highlight evidence supporting the immunomodulatory effects of bifidobacterial molecules (proteins and peptides, exopolysaccharides, metabolites, and DNA), as well as the role of bifidobacterial metabolism in maintaining immune homeostasis through cross-feeding mechanisms.
Collapse
Affiliation(s)
- Lorena Ruiz
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Susana Delgado
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Patricia Ruas-Madiedo
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Borja Sánchez
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| | - Abelardo Margolles
- Dairy Research Institute, Spanish National Research Council (Instituto de Productos Lácteos de Asturias - CSIC), Villaviciosa, Spain
| |
Collapse
|
20
|
Song L, Dong G, Guo L, Graves DT. The function of dendritic cells in modulating the host response. Mol Oral Microbiol 2017; 33:13-21. [PMID: 28845602 DOI: 10.1111/omi.12195] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2017] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that capture, process, and present antigens to lymphocytes to initiate and regulate the adaptive immune response. DCs detect bacteria in skin and mucosa and migrate into regional lymph nodes, where they stimulate antigen-specific T and B lymphocyte activation and proliferation. DCs direct CD4 T cells to differentiate to T-cell subsets such as T helper cells types 1, 2, and 17, and regulatory T cells. The periodontium is chronically exposed to oral bacteria that stimulate an inflammatory response to induce gingivitis or periodontitis. DCs play both protective and destructive roles through activation of the acquired immune response and are also reported to be a source of osteoclast precursors that promote bone resorption. FOXO1, a member of the forkhead box O family of transcription factors, plays a significant role in the activation of DCs. The function of DCs in periodontal inflammation has been investigated in a mouse model by lineage-specific deletion of FOXO1 in these cells. Deletion of FOXO1 reduces DC protective function and enhances susceptibility to periodontitis. The kinase Akt, phosphorylates FOXO1 to inhibit FOXO activity. Hence the Akt-FOXO1 axis may play a key role in regulating DCs to have a significant impact on periodontal disease.
Collapse
Affiliation(s)
- L Song
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - G Dong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - L Guo
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Molecular Laboratory for Gene Therapy and Tooth Regeneration and Department of Orthodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - D T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
21
|
Westfall S, Lomis N, Kahouli I, Dia SY, Singh SP, Prakash S. Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell Mol Life Sci 2017; 74:3769-3787. [PMID: 28643167 PMCID: PMC11107790 DOI: 10.1007/s00018-017-2550-9] [Citation(s) in RCA: 347] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/05/2017] [Accepted: 05/29/2017] [Indexed: 02/07/2023]
Abstract
The gut microbiota is essential to health and has recently become a target for live bacterial cell biotherapies for various chronic diseases including metabolic syndrome, diabetes, obesity and neurodegenerative disease. Probiotic biotherapies are known to create a healthy gut environment by balancing bacterial populations and promoting their favorable metabolic action. The microbiota and its respective metabolites communicate to the host through a series of biochemical and functional links thereby affecting host homeostasis and health. In particular, the gastrointestinal tract communicates with the central nervous system through the gut-brain axis to support neuronal development and maintenance while gut dysbiosis manifests in neurological disease. There are three basic mechanisms that mediate the communication between the gut and the brain: direct neuronal communication, endocrine signaling mediators and the immune system. Together, these systems create a highly integrated molecular communication network that link systemic imbalances with the development of neurodegeneration including insulin regulation, fat metabolism, oxidative markers and immune signaling. Age is a common factor in the development of neurodegenerative disease and probiotics prevent many harmful effects of aging such as decreased neurotransmitter levels, chronic inflammation, oxidative stress and apoptosis-all factors that are proven aggravators of neurodegenerative disease. Indeed patients with Parkinson's and Alzheimer's diseases have a high rate of gastrointestinal comorbidities and it has be proposed by some the management of the gut microbiota may prevent or alleviate the symptoms of these chronic diseases.
Collapse
Affiliation(s)
- Susan Westfall
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Nikita Lomis
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Imen Kahouli
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Si Yuan Dia
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Surya Pratap Singh
- Department of Biochemistry, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
| |
Collapse
|
22
|
Inaba Y, Ueno N, Numata M, Zhu X, Messer JS, Boone DL, Fujiya M, Kohgo Y, Musch MW, Chang EB. Soluble bioactive microbial mediators regulate proteasomal degradation and autophagy to protect against inflammation-induced stress. Am J Physiol Gastrointest Liver Physiol 2016; 311:G634-G647. [PMID: 27514476 PMCID: PMC5142193 DOI: 10.1152/ajpgi.00092.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/07/2016] [Indexed: 01/31/2023]
Abstract
Bifidobacterium breve and other Gram-positive gut commensal microbes protect the gastrointestinal epithelium against inflammation-induced stress. However, the mechanisms whereby these bacteria accomplish this protection are poorly understood. In this study, we examined soluble factors derived from Bifidobacterium breve and their impact on the two major protein degradation systems within intestinal epithelial cells, proteasomes and autophagy. Conditioned media from gastrointestinal Gram-positive, but not Gram-negative, bacteria activated autophagy and increased expression of the autophagy proteins Atg5 and Atg7 along with the stress response protein heat shock protein 27. Specific examination of media conditioned by the Gram-positive bacterium Bifidobacterium breve (Bb-CM) showed that this microbe produces small molecules (<3 kDa) that increase expression of the autophagy proteins Atg5 and Atg7, activate autophagy, and inhibit proteasomal enzyme activity. Upregulation of autophagy by Bb-CM was mediated through MAP kinase signaling. In vitro studies using C2BBe1 cells silenced for Atg7 and in vivo studies using mice conditionally deficient in intestinal epithelial cell Atg7 showed that Bb-CM-induced cytoprotection is dependent on autophagy. Therefore, this work demonstrates that Gram-positive bacteria modify protein degradation programs within intestinal epithelial cells to promote their survival during stress. It also reveals the therapeutic potential of soluble molecules produced by these microbes for prevention and treatment of gastrointestinal disease.
Collapse
Affiliation(s)
- Yuhei Inaba
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Nobuhiro Ueno
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Masatsugu Numata
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois; Division of Life Style and Digestive Diseases, Kagoshima Medical University, Kagoshima, Japan
| | - Xiaorong Zhu
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - Jeannette S Messer
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - David L Boone
- Department of Microbiology and Immunology, Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Mikihiro Fujiya
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Yutaka Kohgo
- Division of Gastroenterology and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, Asahikawa, Japan
| | - Mark W Musch
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Inflammatory Bowel Disease Research Center, The University of Chicago, Chicago, Illinois;
| |
Collapse
|
23
|
Kwak MJ, Kwon SK, Yoon JK, Song JY, Seo JG, Chung MJ, Kim JF. Evolutionary architecture of the infant-adapted group of Bifidobacterium species associated with the probiotic function. Syst Appl Microbiol 2016; 39:429-439. [PMID: 27524178 DOI: 10.1016/j.syapm.2016.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 01/11/2023]
Abstract
Bifidobacteria, often associated with the gastrointestinal tract of animals, are well known for their roles as probiotics. Among the dozens of Bifidobacterium species, Bifidobacterium bifidum, B. breve, and B. longum are the ones most frequently isolated from the feces of infants and known to help the digestion of human milk oligosaccharides. To investigate the correlation between the metabolic properties of bifidobacteria and their phylogeny, we performed a phylogenomic analysis based on 452 core genes of forty-four completely sequenced Bifidobacterium species. Results show that a major evolutionary event leading to the clade of the infant-adapted species is linked to carbohydrate metabolism, but it is not the only factor responsible for the adaptation of bifidobacteria to the gut. The genome of B. longum subsp. infantis, a typical bifidobacterium in the gut of breast-fed infants, encodes proteins associated with several kinds of species-specific metabolic pathways, including urea metabolism and biosynthesis of riboflavin and lantibiotics. Our results demonstrate that these metabolic features, which are associated with the probiotic function of bifidobacteria, are species-specific and highly correlate with their phylogeny.
Collapse
Affiliation(s)
- Min-Jung Kwak
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Soon-Kyeong Kwon
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae-Kyung Yoon
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Ju Yeon Song
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jae-Gu Seo
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - Myung Jun Chung
- R&D Center, Cell Biotech Co., Ltd., 50, Aegibong-ro 409 beon-gil, Wolgot-myeon, Gimpo-si, Gyeonggi-do 10003, Republic of Korea
| | - Jihyun F Kim
- Department of Systems Biology and Division of Life Sciences, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Strategic Initiative for Microbiomes in Agriculture and Food, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
24
|
Saw S, Arora N. PI3K and ERK1/2 kinase inhibition potentiate protease inhibitor to attenuate allergen induced Th2 immune response in mouse. Eur J Pharmacol 2016; 776:176-84. [PMID: 26905476 DOI: 10.1016/j.ejphar.2016.02.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 12/14/2022]
Abstract
Proteases affect immune response by activating PI3K, ERK1/2 and p38 kinase. In present study, therapeutic effect of PI3K, ERK1/2 and p38 kinase inhibitor in combination with serine protease inhibitor was evaluated in cockroach extract (CE) induced airway inflammatory disease. Mice were sensitized on day 0, 7 and 14 and challenged on day 27, 28 and 29 with CE. Mice were given PI3K, ERK1/2 and the p38 kinase inhibitor (iPI3K, iERK1/2 and the ip38) alone or with serine protease inhibitor 4-(2-Aminoethyl) benzenesulfonyl fluoride hydrochloride (AEBSF), 1h before challenge. On day 30 airway resistance of mice were determined and euthanized to collect blood, BAL fluid and lung for analysis. CE immunized mice showed PI3K, ERK1/2 and p38 kinase activation, increased airway resistance, cellular infiltration, Th2 cytokines IgE and IgG1. AEBSF given to mice reduced the CE induced allergic response. AEBSF given in combination of iPI3K/iERK1/2 reduced cellular infiltration in lungs. Furthermore, iPI3K/iERK1/2 with AEBSF significantly reduced the CE induced Th2 cytokines in comparison to monotherapy of kinase inhibitor and AEBSF (P<0.05). The combination of iPI3K/iERK1/2 with AEBSF enhanced IL-12 level that could further provide a mean of Th2 reduction. Best effect in reduction of allergic response in mice was observed on administration of AEBSF with iPI3K. Conclusively, the combination of PI3K kinase inhibitor with AEBSF reduced allergen induced airway response and has therapeutic potential for add-on therapy in allergic airway disease.
Collapse
Affiliation(s)
- Sanjay Saw
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110007, India
| | - Naveen Arora
- CSIR-Institute of Genomics and Integrative Biology, Delhi University Campus, Mall Road, Delhi 110007, India.
| |
Collapse
|
25
|
In silico mining and characterization of bifidobacterial lipoprotein with CHAP domain secreted in an aggregated form. Int J Biol Macromol 2016; 82:653-62. [DOI: 10.1016/j.ijbiomac.2015.10.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 10/02/2015] [Accepted: 10/07/2015] [Indexed: 12/13/2022]
|
26
|
Ghoneum M, Felo N, Agrawal S, Agrawal A. A novel kefir product (PFT) activates dendritic cells to induce CD4+T and CD8+T cell responses in vitro. Int J Immunopathol Pharmacol 2015; 28:488-96. [PMID: 26384392 DOI: 10.1177/0394632015599447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Lactobacilli have been widely studied for their probiotic effects and have been reported to function as antiviral and anticancer agents. However, the underlying mechanisms via immune modulation are poorly understood. PFT is a freeze dried compound of Lactobacillus kefiri P-IF with a unique composition and functionality. In this study, we examined the potential stimulatory effects of two concentrations (50 µg and 100 µg/mL) of PFT on human monocyte-derived dendritic cell (DC) function in vitro. Results showed that PFT upregulated the expression of DC surface co-stimulatory and maturation markers CD80, CD86, and HLADR in a concentration dependent manner. PFT at 100 µg/mL markedly increased the secretion of IL-6, IL-10, TNF-α, and IL-1β by DCs. This concentration of PFT also stimulated the production of antiviral cytokines, IFN-α and IFN-λ(IL29) in DCs. Additionally, PFT at 100 µg/mL activated moDCs prime CD4(+)T cells and significantly increased the levels of IL-10, IFN-γ, and TNF-α by 1.7, four, three-fold, respectively. Furthermore PFT-stimulated DCs were also effective in enhancing the cytotoxic potential of CD8(+)T cells via the induction of Granzyme-B and upregulation of CD107a, and CD103 expression, a marker of resident/regulatory CD8(+)T cells. These data suggest that PFT functions as a natural adjuvant for DC activation and thus may be used in DC-based vaccine strategies against viral infections and cancer.
Collapse
Affiliation(s)
- Mamdooh Ghoneum
- Department of Otolaryngology, Charles Drew University of Medicine and Science, 1621 E. 120th Street, Los Angeles, CA 90059, USA
| | - Nouran Felo
- Department of Otolaryngology, Charles Drew University of Medicine and Science, 1621 E. 120th Street, Los Angeles, CA 90059, USA
| | - Sudhanshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California Irvine, Irvine, CA 92697
| | - Anshu Agrawal
- Department of Medicine, Division of Basic and Clinical Immunology, University of California Irvine, Irvine, CA 92697
| |
Collapse
|
27
|
Lee TM, Lin SZ, Chang NC. Inhibition of glycogen synthase kinase-3β prevents sympathetic hyperinnervation in infarcted rats. Exp Biol Med (Maywood) 2015; 240:979-92. [PMID: 25576342 DOI: 10.1177/1535370214564746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 10/29/2014] [Indexed: 11/16/2022] Open
Abstract
We have demonstrated that nerve growth factor (NGF) expression in the myocardium is selectively increased during chronic stage of myocardial infarction, resulting in sympathetic hyperinnervation. Glycogen synthase kinase-3 (GSK-3) signal has been shown to play key roles in the regulation of cytoskeletal assembly during axon regeneration. We assessed whether lithium, a GSK-3 inhibitor, attenuates cardiac sympathetic reinnervation after myocardial infarction through attenuated NGF expression and Tau expression. Twenty-four hours after ligation of the anterior descending artery, male Wistar rats were randomized to either LiCl or SB216763, chemically unrelated inhibitors of GSK-3β, a combination of LiCl and SB216763, or vehicle for four weeks. Myocardial norepinephrine levels revealed a significant elevation in vehicle-treated rats compared with sham-operated rats, consistent with excessive sympathetic reinnervation after infarction. Immunohistochemical analysis for sympathetic nerve also confirmed the change of myocardial norepinephrine. This was paralleled by a significant upregulation of NGF protein and mRNA in the vehicle-treated rats, which was reduced after administering either LiCl, SB216763, or combination. Arrhythmic scores during programmed stimulation in the vehicle-treated rats were significantly higher than those treated with GSK-3 inhibitors. Addition of SB216763 did not have additional beneficial effects compared with those seen in rats treated with LiCl alone. Furthermore, lithium treatment increased Tau1 and decreased AT8 and AT180 levels. Chronic use of lithium after infarction, resulting in attenuated sympathetic reinnervation by GSK-3 inhibition, may modify the arrhythmogenic response to programmed electrical stimulation.
Collapse
Affiliation(s)
- Tsung-Ming Lee
- Department of Medicine, Cardiology Section, China Medical University-An Nan Hospital, Tainan 709, Taiwan Department of Medicine, China Medical University, Taichung 40447, Taiwan Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Shinn-Zong Lin
- Neuropsychiatry Center, China Medical University Hospital, Taichung 40447, Taiwan Graduate Institute of Immunology, China Medical University, Taichung 40447, Taiwan Department of Neurosurgery, China Medical University Beigan Hospital, Yunlin 651, Taiwan Department of Neurosurgery, China Medical University-An Nan Hospital, Tainan 40447, Taiwan
| | - Nen-Chung Chang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
| |
Collapse
|
28
|
Plé C, Adouard N, Breton J, Dewulf J, Pot B, Bonnarme P, Foligné B. Designing specific cheese-ripening ecosystems to shape the immune effects of dairy products? J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.11.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
29
|
In vitro characterization of the digestive stress response and immunomodulatory properties of microorganisms isolated from smear-ripened cheese. Int J Food Microbiol 2014; 197:98-107. [PMID: 25589362 DOI: 10.1016/j.ijfoodmicro.2014.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 12/09/2014] [Accepted: 12/14/2014] [Indexed: 11/20/2022]
Abstract
Thirty-six microorganisms (twenty-one bacteria, twelve yeasts and three fungi) were isolated from surface-ripened cheeses and subjected to in vitro digestive stress. The approach mimicked gastric and/or duodenal digestion. Lactobacillus rhamnosus GG, Escherichia coli Nissle 1917 and Saccharomyces boulardii were used as reference strains. We studied the microorganisms grown separately in culture medium and then included (or not) in a rennet gel. The microorganisms' immunomodulatory abilities were also assessed by profiling cytokine induction in human peripheral blood mononuclear cells (PBMCs). The loss of viability was less than 1 log CFU/mL for yeasts under all conditions. In contrast, Gram-negative bacteria survived gastric and/or duodenal stress well but most of the Gram-positive bacteria were more sensitive (especially to gastric stress). Inclusion of sensitive Gram-positive bacteria in rennet gel dramatically improved gastric survival, when compared with a non-included cultured (with a 4 log CFU/mL change in survival). However, the rennet gel did not protect the bacteria against duodenal stress. The PBMC cytokine assay tests showed that the response to yeasts was usually anti-inflammatory, whereas the response to bacteria varied from one strain to another.
Collapse
|
30
|
Scuotto A, Djorie S, Colavizza M, Romond PC, Romond MB. Bifidobacterium breve C50 secretes lipoprotein with CHAP domain recognized in aggregated form by TLR2. Biochimie 2014; 107 Pt B:367-75. [PMID: 25457102 DOI: 10.1016/j.biochi.2014.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/10/2014] [Indexed: 10/24/2022]
Abstract
Extracellular components secreted by Bifidobacterium breve C50 can induce maturation, high IL-10 production and prolonged survival of dendritic cells via a TLR2 pathway. In this study, the components were isolated from the supernatant by gel filtration chromatography. Antibodies raised against the major compounds with molecular weight above 600 kDa (Bb C50BC) also recognized compounds of lower molecular weight (200–600 kDa). TLR2 and TLR6 bound to the components already recognized by the antibodies. Trypsin digestion of Bb C50BC released three major peptides whose sequences displayed close similarities to a putative secreted protein with a CHAP amidase domain from B. breve. The 1300-bp genomic region corresponding to the hypothetical protein was amplified by PCR. The deduced polypeptide started with an N-terminal signal sequence of 45 amino acids, containing the lipobox motif (LAAC) with the cysteine in position 25, and 2 positively charged residues within the first 14 residues of the signal sequence. Lipid detection in Bb C50BC by GC/MS further supported the implication of a lipoprotein. Sugars were also detected in Bb C50BC. Close similarity with the glucan-binding protein B from Bifidobacterium animalis of two released peptides from Bb C50BC protein suggested that glucose moieties, possibly in glucan form, could be bound to the lipoprotein. Finally, heating at 100 °C for 5 min led to the breakdown of Bb C50BC in compounds of molecular weight below 67 kDa, which suggested that Bb C50BC was an aggregate. One might assume that a basic unit was formed by the lipoprotein bound putatively to glucan. Besides the other sugars and hexosamines recognized by galectin 1 were localized at the surface of the Bb C50BC aggregate. In conclusion, the extracellular components secreted by B. breve C50 were constituted of a lipoprotein putatively associated with glucose moieties and acting in an aggregating form as an agonist of TLR2/TLR6.
Collapse
|
31
|
Villena J, Aso H, Kitazawa H. Regulation of toll-like receptors-mediated inflammation by immunobiotics in bovine intestinal epitheliocytes: role of signaling pathways and negative regulators. Front Immunol 2014; 5:421. [PMID: 25228903 PMCID: PMC4151153 DOI: 10.3389/fimmu.2014.00421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/19/2014] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) detect bacterial and viral associated molecular patterns via germline-encoded pattern-recognition receptors (PRRs) and are responsible for maintaining immune tolerance to the communities of resident commensal bacteria while being also capable to mount immune responses against pathogens. Toll-like receptors (TLRs) are a major class of PRRs expressed on IECs and immune cells, which are involved in the induction of both tolerance and inflammation. In the last decade, experimental and clinical evidence was generated to support the application of probiotics with immunoregulatory capacities (immunobiotics) for the prevention and treatment of several gastrointestinal inflammatory disorders in which TLRs exert a significant role. The majority of these studies were performed in mouse and human cell lines, and despite the growing interest in the bovine immune system due to the economic importance of cattle as livestock, only few studies have been conducted on cattle. In this regard, our group has established a bovine intestinal epithelial (BIE) cell line originally derived from fetal bovine intestinal epitheliocytes and used this cell line to evaluate the impact of immunobiotics in TLR-mediated inflammation. This review aims to summarize the current knowledge of the beneficial effects of immunobiotics in the regulation of intestinal inflammation/infection in cattle. Especially, we discuss the role of TLRs and their negative regulators in both the inflammatory response and the beneficial effects of immunobiotics in bovine IECs. This review article emphasizes the cellular and molecular interactions of immunobiotics with BIE cells through TLRs and gives the scientific basis for the development of immunomodulatory feed for bovine healthy development.
Collapse
Affiliation(s)
- Julio Villena
- Immunobiotics Research Group , Tucuman , Argentina ; Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET) , Tucuman , Argentina
| | - Hisashi Aso
- Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University , Sendai , Japan
| |
Collapse
|
32
|
Zhong SS, Li SM, Zhang ZS. Effect of bifidobacterial adhesin on intestinal microflora and bacterial translocation induced by ischemia-reperfusion injury in rats. Shijie Huaren Xiaohua Zazhi 2014; 22:3632-3638. [DOI: 10.11569/wcjd.v22.i24.3632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the effect of bifidobacterial adhesin on intestinal microflora and bacterial translocation induced by ischemia-reperfusion (I/R) injury in rats.
METHODS: Seventy-two SD rats were randomly divided into a sham operation group (n = 24), an I/R model group (n = 24) and an experiment group (n = 24; treated with bifidobacterial adhesin). The rats were sacrificed 6 h, 1, 4 and 7 d after inducing I/R. The changes of intestinal microflora and bacterial translocation were observed, and the blood level of endotoxin was measured.
RESULTS: In the I/R model group, the numbers of Enterrococci (6.63 lgN/g ± 1.06 lgN/g vs 5.26 lgN/g ± 1.08 lgN/g, 9.44 lgN/g ± 1.37 lgN/g vs 5.30 lgN/g ± 1.12 lgN/g, 8.56 lgN/g ± 1.35 lgN/g vs 4.99 lgN/g ± 0.96 lgN/g, 8.23 lgN/g ± 1.01 lgN/g vs 5.18 lgN/g ± 1.03 lgN/g, P < 0.05) and Enterobacilli (7.86 lgN/g ± 1.17 lgN/g vs 6.39 lgN/g ± 0.85 lgN/g, 9.49 lgN/g ± 1.23 lgN/g vs 6.64 lgN/g ± 1.44 lgN/g, 8.76 lgN/g ± 0.86 lgN/g vs 6.52 lgN/g ± 1.13 lgN/g, 8.89 lgN/g ± 1.09 lgN/g vs 6.71 lgN/g ± 0.98 lgN/g, P < 0.05) in the feces of rats increased significantly at all time points, the numbers of Clostridium perfringens increased significantly on days 1 (6.47 lgN/g ± 1.43 lgN/g vs 4.51 lgN/g ± 1.22 lgN/g, P < 0.05), 4 (6.70 lgN/g ± 1.16 lgN/g vs 4.71 lgN/g ± 0.89 lgN/g, P < 0.05) and 7 (6.55 lgN/g ± 1.29 lgN/g vs 4.46 lgN/g ± 0.79 lgN/g, P < 0.05), while the numbers of Bifidobacterium (6.13 lgN/g ± 1.28 lgN/g vs 9.02 lgN/g ± 1.10 lgN/g, 5.59 lgN/g ± 1.22 lgN/g vs 8.66 lgN/g ± 0.99 lgN/g, P < 0.05) and Lactobacillus (6.07 lgN/g ± 1.09 lgN/g vs 9.08 lgN/g ± 1.04 lgN/g, 5.35 lgN/g ± 1.26 lgN/g vs 8.89 lgN/g ± 0.97 lgN/g, P < 0.05) decreased significantly on days 4 and 7 compared with those in the control subjects. The numbers of Enterrococci (6.37 lgN/g ± 1.04 lgN/g vs 8.56 lgN/g ± 1.35 lgN/g, 5.42 lgN/g ± 0.92 lgN/g vs 8.23 lgN/g ± 1.01 lgN/g, P < 0.05) and Enterobaci (7.55 lgN/g ± 1.03 lgN/g vs 8.76 lgN/g ± 0.86 lgN/g, 7.16 lgN/g ± 0.86 lgN/g vs 8.89 lgN/g ± 1.09 lgN/g, P < 0.05) in the experiment group were significantly lower than those in the I/R model group on days 4 and 7. The numbers of Clostridium perfringens on days 1, 4 and 7 (5.95 lgN/g ± 1.24 lgN/g vs 4.51 lgN/g ± 1.22 lgN/g, 6.08 lgN/g ± 1.07 lgN/g vs 4.71 lgN/g ± 0.89 lgN/g, 5.87 lgN/g ± 0.82 lgN/g vs 4.46 lgN/g ± 0.79 lgN/g, P< 0.05) were significantly higher in the experiment group than in the control group, but had declined compared to those in the I/R group. The numbers of Bifidobacterium (8.56 lgN/g ± 0.85 lgN/g vs 8.45 lgN/g ± 0.86 lgN/g, 7.89 lgN/g ± 1.47 lgN/g vs 8.78 lgN/g ± 1.06 lgN/g, 8.67 lgN/g ± 1.13 lgN/g vs 9.02 lgN/g ± 1.10 lgN/g, 8.75 lgN/g ± 0.96 lgN/g vs 8.66 lgN/g ± 0.99 lgN/g, P > 0.05) and Lactobacillus (9.16 lgN/g ± 0.94 lgN/g vs 8.91 lgN/g ± 1.06 lgN/g, 8.56 lgN/g ± 1.21 lgN/g vs 9.11 lgN/g ± 1.13 lgN/g, 9.16 lgN/g ± 1.08 lgN/g vs 9.08 lgN/g ± 1.04 lgN/g, 9.01 lgN/g ± 0.95 lgN/g vs 8.89 lgN/g ± 0.97 lgN/g, P > 0.05) showed no significant changes at all time points in the experiment group. The values of endotoxin (1.43 EU/mL ± 0.32 EU/mL vs 0.21 EU/mL ± 0.18 EU/mL,1.84 EU/mL ± 0.24 EU/mL vs 0.30 EU/mL ± 0.23 EU/mL,1.69 EU/mL ± 0.35 EU/mL vs 0.26 EU/mL ± 0.21 EU/mL,1.73 EU/mL ± 0.31 EU/mL vs 0.28 EU/mL ± 0.19 EU/mL, P < 0.05) and bacterial translocation rates in the liver (50% vs 0, 66.67% vs 16.67%, 83.33% vs 0, 83.33% vs 0, P < 0.05), spleen (33.33% vs 0, 50% vs 0, 66.67% vs 0, 66.67% vs 0, P < 0.05) and mesenteric lymph nodes (66.67% vs 0, 83.33% vs 0, 100% vs 16.67%, 100% vs 0, P < 0.05) in the I/R model group were significantly higher than those in the control group, while these parameters were significantly lower in the experiment group than in the I/R model group at all time points (endotoxin: 0.57 EU/mL ± 0.23 EU/mL vs 1.43 EU/mL ± 0.32 EU/mL, 0.71 EU/mL ± 0.16 EU/mL vs 1.84 EU/mL ± 0.24 EU/mL, 0.41 EU/mL ± 0.22 EU/mL vs 1.69 EU/mL ± 0.35 EU/mL, 0.35 EU/mL ± 0.12 EU/mL vs 1.73 EU/mL ± 0.31 EU/mL, P < 0.05; bacterial translocation in the liver: 16.67% vs 50%, 33.33% vs 66.67%, 50% vs 83.33%, 33.33% vs 83.33%, P < 0.05; spleen: 0 vs 33.33%, 16.67% vs 50%, 33.33% vs 66.67%, 33.33% vs 66.67%, P < 0.05; mesenteric lymph nodes: 50% vs 66.67%, 50% vs 83.33%, 50% vs 100%, 33.33% vs 100%, P < 0.05).
CONCLUSION: Bifidobacterial adhesin can improve the intestinal flora imbalance, reduce bacterial translocation and endotoxemia occurrence, and protect intestinal mucosal barrier function in rats after intestinal I/R injury.
Collapse
|
33
|
Immune modulating capability of two exopolysaccharide-producing Bifidobacterium strains in a Wistar rat model. BIOMED RESEARCH INTERNATIONAL 2014; 2014:106290. [PMID: 24971309 PMCID: PMC4058098 DOI: 10.1155/2014/106290] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/28/2014] [Indexed: 12/14/2022]
Abstract
Fermented dairy products are the usual carriers for the delivery of probiotics to humans, Bifidobacterium and Lactobacillus being the most frequently used bacteria. In this work, the strains Bifidobacterium animalis subsp. lactis IPLA R1 and Bifidobacterium longum IPLA E44 were tested for their capability to modulate immune response and the insulin-dependent glucose homeostasis using male Wistar rats fed with a standard diet. Three intervention groups were fed daily for 24 days with 10% skimmed milk, or with 109 cfu of the corresponding strain suspended in the same vehicle. A significant increase of the suppressor-regulatory TGF-β cytokine occurred with both strains in comparison with a control (no intervention) group of rats; the highest levels were reached in rats fed IPLA R1. This strain presented an immune protective profile, as it was able to reduce the production of the proinflammatory IL-6. Moreover, phosphorylated Akt kinase decreased in gastroctemius muscle of rats fed the strain IPLA R1, without affecting the glucose, insulin, and HOMA index in blood, or levels of Glut-4 located in the membrane of muscle and adipose tissue cells. Therefore, the strain B. animalis subsp. lactis IPLA R1 is a probiotic candidate to be tested in mild grade inflammation animal models.
Collapse
|
34
|
Al‐Hassi HO, Mann ER, Sanchez B, English NR, Peake ST, Landy J, Man R, Urdaci M, Hart AL, Fernandez‐Salazar L, Lee GH, Garrote JA, Arranz E, Margolles A, Stagg AJ, Knight SC, Bernardo D. Altered human gut dendritic cell properties in ulcerative colitis are reversed by
Lactobacillus plantarum
extracellular encrypted peptide STp. Mol Nutr Food Res 2013; 58:1132-43. [PMID: 24347371 DOI: 10.1002/mnfr.201300596] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/09/2013] [Accepted: 10/18/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Hafid O. Al‐Hassi
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
| | - Elizabeth R. Mann
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
| | - Borja Sanchez
- Nutrition and Bromatology Group Department of Analytical and Food Chemistry Food Science and Technology Faculty University of Vigo Ourense Campus Ourense Spain
| | - Nicholas R. English
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
| | - Simon T.C. Peake
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
- St. Mark's Hospital North West London Hospitals NHS Trust Harrow UK
| | - Jonathan Landy
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
- St. Mark's Hospital North West London Hospitals NHS Trust Harrow UK
| | - Ripple Man
- St. Mark's Hospital North West London Hospitals NHS Trust Harrow UK
| | - Maria Urdaci
- Laboratoire de Microbiologie et Biochimie Appliquee Ecole Nationale Superieure des Sciences Agronomiques de Bordeaux Gradignan France
| | - Ailsa L. Hart
- St. Mark's Hospital North West London Hospitals NHS Trust Harrow UK
| | | | - Gui Han Lee
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
- St. Mark's Hospital North West London Hospitals NHS Trust Harrow UK
| | - Jose A. Garrote
- Department of Genetics and Molecular Biology Clinical Laboratory Service Hospital Universitario Rio Hortega Valladolid Spain
| | - Eduardo Arranz
- Department of Paediatrics and Immunology Mucosal Immunology Service Universidad de Valladolid IBGM‐CSIC Valladolid Spain
| | - Abelardo Margolles
- Departmento de Microbiologia y Bioquimica de Productos Lacteos Instituto de Productos Lacteos de Asturias Consejo Superior de Investigaciones Cientificas Villaviciosa Spain
| | - Andrew J. Stagg
- Centre for Immunology and Infectious Disease Barts and the London School of Medicine and Dentistry Blizard Institute of Cell and Molecular Science Queen Mary University of London UK
| | - Stella C. Knight
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
| | - David Bernardo
- Antigen Presentation Research Group Imperial College London Northwick Park and St. Mark's Campus Harrow UK
| |
Collapse
|
35
|
Fermentation products: immunological effects on human and animal models. Pediatr Res 2013; 74:238-44. [PMID: 23670282 DOI: 10.1038/pr.2013.76] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 12/29/2012] [Indexed: 01/06/2023]
Abstract
Infant formulas have been shown to influence the development of the gut microbiota. Besides the probiotic- and prebiotic-containing formulas, fermented milk-based infant formulas offer an additional means for modulation of gut immunity and/or gut microbiota. These formulas are produced by the fermentation of cow's milk with specific lactic acid bacteria strains, followed by heat treatment; they do not contain viable bacteria or added prebiotic oligosaccharides but contain specific products resulting from the fermentation process. This review is focused on the effects of fermentation products, distinguishing them from those of living bacteria and prebiotic compounds on the immune system. Besides the possible modulation of gut microbiota composition, in vitro and in vivo studies suggest that specific fermentation products can actively participate in the establishment of immune balance and oral tolerance. Although further research is needed to confirm the clinical benefits observed in infants to better characterize the active fermentation compounds and to delineate the involved pathways, these fermented formulas appear to deserve interest.
Collapse
|
36
|
Trojandt S, Knies D, Pektor S, Ritz S, Mailänder V, Grabbe S, Reske-Kunz AB, Bros M. The chemotherapeutic agent topotecan differentially modulates the phenotype and function of dendritic cells. Cancer Immunol Immunother 2013; 62:1315-26. [PMID: 23666509 PMCID: PMC11029351 DOI: 10.1007/s00262-013-1431-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 04/28/2013] [Indexed: 11/24/2022]
Abstract
The camptothecin analogue topotecan (TPT) induces tumor cell apoptosis due to interference with topoisomerase I and is clinically used as a second-line chemotherapeutic in the treatment for metastasizing ovarian and small cell lung carcinoma. Based on the more recent finding of TPT-mediated inhibition of the transcription factor hypoxia-induced factor-1α, a hallmark of solid tumors, TPT, is currently tested in clinical trials for its suitability as a first-line chemotherapeutic for the treatment for various types of tumors. Due to the gained clinical interest in TPT and in light of its modulatory effect on signaling pathways, which are also of importance for immune cell functions, we asked for potential effects of TPT on dendritic cells (DCs), the main antigen-presenting cell population of the immune system. Here, we show that TPT at a therapeutically relevant dose partially activated monocyte-derived DCs as reflected by enhanced migratory activity, elevated expression of HLA-DR and costimulatory/maturation markers, and accordingly an increased allogenic CD4(+) T cell stimulation. In marked contrast, TPT prevented full maturation of DCs stimulated with a cocktail of proinflammatory mediators, accompanied by somewhat lower upregulation of NF-κB factors p65 and RelB.
Collapse
Affiliation(s)
- Stefanie Trojandt
- Department of Dermatology, Clinical Research Unit Allergology, Medical Center of the Johannes Gutenberg-University, Obere Zahlbacher-Str. 63, 55131, Mainz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Harb H, van Tol EAF, Heine H, Braaksma M, Gross G, Overkamp K, Hennen M, Alrifai M, Conrad ML, Renz H, Garn H. Neonatal supplementation of processed supernatant fromLactobacillus rhamnosusGG improves allergic airway inflammation in mice later in life. Clin Exp Allergy 2013; 43:353-64. [DOI: 10.1111/cea.12047] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 09/25/2012] [Accepted: 10/16/2012] [Indexed: 01/20/2023]
Affiliation(s)
- H. Harb
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| | | | - H. Heine
- Section of Immunoregulation; Research Center Borstel - Leibniz-Center for Biosciences and Medicine; Borstel; Germany
| | - M. Braaksma
- Microbiology and Systems Biology; TNO; Zeist; The Netherlands
| | | | - K. Overkamp
- Microbiology and Systems Biology; TNO; Zeist; The Netherlands
| | - M. Hennen
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| | - M. Alrifai
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| | - M. L. Conrad
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| | - H. Renz
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| | - H. Garn
- Institute for Laboratory Medicine and Pathobiochemistry - Molecular Diagnostics; Philipps University of Marburg; Marburg; Germany
| |
Collapse
|
38
|
Intestinal dendritic cells: their role in intestinal inflammation, manipulation by the gut microbiota and differences between mice and men. Immunol Lett 2013; 150:30-40. [PMID: 23352670 DOI: 10.1016/j.imlet.2013.01.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 01/08/2013] [Accepted: 01/08/2013] [Indexed: 02/06/2023]
Abstract
The intestinal immune system maintains a delicate balance between immunogenicity against invading pathogens and tolerance of the commensal microbiota and food antigens. Dendritic cells (DC) generate primary T-cell responses, and determine whether these responses are immunogenic or tolerogenic. The regulatory role of DC is of particular importance in the gut due to the high antigenic load. Intestinal DC act as sentinels, sampling potentially pathogenic antigens but also harmless antigens including the commensal microbiota. Following antigen acquisition, intestinal DC migrate to secondary lymphoid organs to activate naive T-cells. DC also imprint specific homing properties on T-cells that they stimulate; gut DC specifically induce gut-homing properties on T-cells upon activation, enabling T-cell migration back to intestinal sites. Data regarding properties on gut DC in humans is scarce, although evidence now supports the role of DC as important players in intestinal immunity in humans. Here, we review the role of intestinal DC in shaping mucosal immune responses and directing tissue-specific T-cell responses, with a special focus on the importance of distinguishing DC subsets from macrophages at intestinal sites. We compare and contrast human DC with their murine counterparts, and discuss the ability of the gut microbiota to shape intestinal DC function, and how this may be dysregulated in inflammatory bowel disease (IBD). Lastly, we describe recent advances in the study of probiotics on intestinal DC function, including the use of soluble secreted bacterial products.
Collapse
|
39
|
Murata K, Villena J, Tomosada Y, Hara R, Chiba E, Shimazu T, Aso H, Suda Y, Iwabuchi N, Xiao JZ, Saito T, Kitazawa H. Bifidobacteria Upregulate Expression of Toll-Like Receptor Negative Regulators Counteracting Enterotoxigenic <i>Escherichia coli</i> Mediated Inflammation in Bovine Intestinal Epitheliocytes. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojvm.2013.32023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Pagnini C, Fave GD, Bamias G. Probiotics in inflammatory bowel disease: Pathophysiological background and clinical applications. World J Immunol 2013; 3:31. [DOI: 10.5411/wji.v3.i3.31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/06/2013] [Accepted: 08/13/2013] [Indexed: 02/05/2023] Open
|
41
|
A human coronavirus responsible for the common cold massively kills dendritic cells but not monocytes. J Virol 2012; 86:7577-87. [PMID: 22553325 DOI: 10.1128/jvi.00269-12] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human coronaviruses are associated with upper respiratory tract infections that occasionally spread to the lungs and other organs. Although airway epithelial cells represent an important target for infection, the respiratory epithelium is also composed of an elaborate network of dendritic cells (DCs) that are essential sentinels of the immune system, sensing pathogens and presenting foreign antigens to T lymphocytes. In this report, we show that in vitro infection by human coronavirus 229E (HCoV-229E) induces massive cytopathic effects in DCs, including the formation of large syncytia and cell death within only few hours. In contrast, monocytes are much more resistant to infection and cytopathic effects despite similar expression levels of CD13, the membrane receptor for HCoV-229E. While the differentiation of monocytes into DCs in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4 requires 5 days, only 24 h are sufficient for these cytokines to sensitize monocytes to cell death and cytopathic effects when infected by HCoV-229E. Cell death induced by HCoV-229E is independent of TRAIL, FasL, tumor necrosis factor alpha, and caspase activity, indicating that viral replication is directly responsible for the observed cytopathic effects. The consequence of DC death at the early stage of HCoV-229E infection may have an impact on the early control of viral dissemination and on the establishment of long-lasting immune memory, since people can be reinfected multiple times by HCoV-229E.
Collapse
|
42
|
Benson KF, Redman KA, Carter SG, Keller D, Farmer S, Endres JR, Jensen GS. Probiotic metabolites from Bacillus coagulans GanedenBC30 TM support maturation of antigen-presenting cells in vitro. World J Gastroenterol 2012; 18:1875-83. [PMID: 22563167 PMCID: PMC3337562 DOI: 10.3748/wjg.v18.i16.1875] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 12/20/2011] [Accepted: 04/01/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the effects of probiotic metabolites on maturation stage of antigen-presenting immune cells.
METHODS: Ganeden Bacillus coagulans 30 (GBC30) bacterial cultures in log phase were used to isolate the secreted metabolite (MET) fraction. A second fraction was made to generate a crude cell-wall-enriched fraction, by centrifugation and lysis, followed by washing. A preparation of MET was subjected to size exclusion centrifugation, generating three fractions: < 3 kDa, 3-30 kDa, and 30-200 kDa and activities were tested in comparison to crude MET and cell wall in primary cultures of human peripheral blood mononuclear cell (PBMC) as a source of antigen-presenting mononuclear phagocytes. The maturation status of mononuclear phagocytes was evaluated by staining with monoclonal antibodies towards CD14, CD16, CD80 and CD86 and analyzed by flow cytometry.
RESULTS: Treatment of PBMC with MET supported maturation of mononuclear phagocytes toward both macrophage and dendritic cell phenotypes. The biological activity unique to the metabolites included a reduction of CD14+ CD16+ pro-inflammatory cells, and this property was associated with the high molecular weight metabolite fraction. Changes were also seen for the dendritic cell maturation markers CD80 and CD86. On CD14dim cells, an increase in both CD80 and CD86 expression was seen, in contrast to a selective increase in CD86 expression on CD14bright cells. The co-expression of CD80 and CD86 indicates effective antigen presentation to T cells and support of T helper cell differentiation. The selective expression of CD86 in the absence of CD80 points to a role in generating T regulatory cells.
CONCLUSION: The data show that a primary mechanism of action of GBC30 metabolites involves support of more mature phenotypes of antigen-presenting cells, important for immunological decision-making.
Collapse
|
43
|
Chen YP, Hsiao PJ, Hong WS, Dai TY, Chen MJ. Lactobacillus kefiranofaciens M1 isolated from milk kefir grains ameliorates experimental colitis in vitro and in vivo. J Dairy Sci 2012; 95:63-74. [PMID: 22192184 DOI: 10.3168/jds.2011-4696] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/25/2011] [Indexed: 12/13/2022]
Abstract
Lactobacillus kefiranofaciens M1, isolated from and identified in Taiwanese milk kefir grain, has demonstrated immune-modulating activity. In the present study, we further investigated the effects of Lb. kefiranofaciens M1 on intestinal epithelial cells in vitro and on dextran sodium sulfate (DSS)-induced colitis in vivo. The possible mechanisms regarding the cytokine products and intestinal epithelial barrier restoration as well as the putative receptor for the protective effects of Lb. kefiranofaciens M1 were investigated. In vitro results indicated that Lb. kefiranofaciens M1 could strengthen the epithelial barrier function in vitro by increasing the transepithelial electrical resistance (TEER) and significantly upregulated the level of the chemokine CCL-20 at both the apical and basolateral sites. The in vivo effects of Lb. kefiranofaciens M1 on the regulation of intestinal physiology indicate that this strain could ameliorate DSS-induced colitis with a significant attenuation of the bleeding score and colon length shortening. Production of proinflammatory cytokines was decreased and that of the antiinflammatory cytokine IL-10 was increased in the DSS-treated mice given Lb. kefiranofaciens M1. The putative receptor for the protective effects of Lb. kefiranofaciens M1 was toll-like receptor 2 (TLR2), which was involved in probiotic-induced cytokine production in vitro and in attenuation of the bleeding score and colon length shortening in vivo. In this study, the kefir lactobacillus Lb. kefiranofaciens M1 clearly demonstrated an anticolitis effect. Based on these results, Lb. kefiranofaciens M1 has the potential to be applied in fermented dairy products as an alternative therapy for intestinal disorders.
Collapse
Affiliation(s)
- Y P Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
44
|
Bergmann C, Akhmetshina A, Dees C, Palumbo K, Zerr P, Beyer C, Zwerina J, Distler O, Schett G, Distler JHW. Inhibition of glycogen synthase kinase 3β induces dermal fibrosis by activation of the canonical Wnt pathway. Ann Rheum Dis 2011; 70:2191-8. [PMID: 21873331 DOI: 10.1136/ard.2010.147140] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Glycogen synthase kinase 3β (GSK-3) regulates the phosphorylation and subsequent degradation of β-catenin, thereby preventing aberrant activation of the canonical Wnt pathway. A study was undertaken to define the role of GSK-3 in fibroblast activation and in experimental models of systemic sclerosis (SSc). METHODS siRNA and specific inhibitors were used to inhibit GSK-3 in cultured fibroblasts and in mice. Activation of the canonical Wnt signalling was analysed by determining the levels of nuclear β-catenin and by measuring the mRNA levels of the Wnt target gene Axin2. The effects of GSK-3 on the release of collagen were evaluated in human dermal fibroblasts and in the mouse model of bleomycin-induced skin fibrosis in tight-skin-1 (tsk-1) mice. RESULTS Targeting GSK-3 potently activated the canonical Wnt pathway in fibroblasts in vitro and in vivo. Inactivation of GSK-3 dose-dependently stimulated the release of collagen from cultured fibroblasts in a β-catenin-dependent manner and further resulted in progressive accumulation of collagen and dermal thickening in mice. Inhibition of GSK-3 aggravated experimental fibrosis in bleomycin-challenged mice and in tsk-1 mice. CONCLUSION Inhibition of GSK-3 activates the canonical Wnt pathway in fibroblasts, stimulates the release of collagen from fibroblasts, exacerbates experimental fibrosis and is sufficient to induce fibrosis. GSK-3 is therefore a key regulator of the canonical Wnt signalling in fibroblasts and inhibition of GSK-3 results in fibroblast activation and increased release of collagen.
Collapse
Affiliation(s)
- Christina Bergmann
- Department of Internal Medicine III and Institute for Clinical Immunology, University of Erlangen-Nuremberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Beurel E. Regulation by glycogen synthase kinase-3 of inflammation and T cells in CNS diseases. Front Mol Neurosci 2011; 4:18. [PMID: 21941466 PMCID: PMC3171068 DOI: 10.3389/fnmol.2011.00018] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Accepted: 08/09/2011] [Indexed: 11/13/2022] Open
Abstract
Elevated markers of neuroinflammation have been found to be associated with many psychiatric and neurodegenerative diseases, such as mood disorders, Alzheimer's disease, and multiple sclerosis (MS). Since neuroinflammation is thought to contribute to the pathophysiology of these diseases and to impair responses to therapeutic interventions and recovery, it is important to identify mechanisms that regulate neuroinflammation and potential targets for controlling neuroinflammation. Recent findings have demonstrated that glycogen synthase kinase-3 (GSK3) is an important regulator of both the innate and adaptive immune systems' contributions to inflammation. Studies of the innate immune system have shown that inhibitors of GSK3 profoundly alter the repertoire of cytokines that are produced both by peripheral and central cells, reducing pro-inflammatory cytokines, and increasing anti-inflammatory cytokines. Furthermore, inhibitors of GSK3 promote tolerance to inflammatory stimuli, reducing inflammatory cytokine production upon repeated exposure. Studies of the adaptive immune system have shown that GSK3 regulates the production of cytokines by T cells and the differentiation of T cells to subtypes, particularly Th17 cells. Regulation of transcription factors by GSK3 appears to play a prominent role in its regulation of immune responses, including of NF-κB, cyclic AMP response element binding protein, and signal transducer and activator of transcription-3. Invivo studies have shown that GSK3 inhibitors ameliorate clinical symptoms of both peripheral and central inflammatory diseases, particularly experimental autoimmune encephalomyelitis, the animal model of MS. Therefore, the development and application of GSK3 inhibitors may provide a new therapeutic strategy to reduce neuroinflammation associated with many central nervous system diseases.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
46
|
Lian GH, Chen MH, Peng XB, Ruan WS. Treatment with Lactobacillus acidophilus cell wall extract and DNA improves experimental acute colitis in mice. Shijie Huaren Xiaohua Zazhi 2011; 19:78-83. [DOI: 10.11569/wcjd.v19.i1.78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of treatment with Lactobacillus acidophilus (L.acidophilus) cell wall extract (BCW) and DNA on experimental colitis in mice.
METHODS: Forty 6-8-wk-old specific pathogen-free female BALB/c mice were randomly divided into four groups: normal group, L.acidophilus BCW-treated group, L.acidophilus DNA-treated group, and control group. Except the normal group, other groups of mice were subjected to induction of experimental colitis with 1.5% DSS in drinking water for 7 d and then treated with L.acidophilus BCW (20 μg/10 g), L.acidophilus DNA (0.2 μg/10 g), and physiological saline (0.2 mL/10 g), respectively. The changes in body weight, fecal traits, and faecal occult bleeding were recorded each day. All animals were killed on day 8 to isolate the whole colon for examination of length and wet weight. Hematoxylin and eosin staining of colonic sections was performed. The effect of treatment with L.acidophilus BCW and DNA on experimental colitis was then evaluated.
RESULTS: Compared with the control group, weight loss was significantly improved (2.94 g ± 0.78 g, 3.37 g ± 1.08 g vs 6.96 g ± 1.39 g, both P < 0.05) and DAI score was significantly decreased (4.27 ± 0.41, 4.62 ± 0.56 vs 6.85 ± 0.94, both P < 0.05) in mice treated with both L.acidophilus BCW and DNA. Treatment with L.acidophilus BCW and DNA prevented the shortening of colon length (8.62 cm ± 1.31 cm, 8.15 cm ± 0.97 cm vs 6.63 cm ± 1.38 cm, both P < 0.05), increased the colon/body weight index (1.63% ± 0.27%, 1.68% ± 0.29% vs 2.12% ± 0.22%, both P < 0.05), improved mucosal damage and inflammatory infiltration, and protected mucosal glands and villi.
CONCLUSION: Treatment with L.acidophilus cell wall extract and DNA can improve DSS-induced experimental colitis in mice.
Collapse
|
47
|
Wang H, Brown J, Martin M. Glycogen synthase kinase 3: a point of convergence for the host inflammatory response. Cytokine 2010; 53:130-40. [PMID: 21095632 DOI: 10.1016/j.cyto.2010.10.009] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 09/29/2010] [Accepted: 10/26/2010] [Indexed: 02/07/2023]
Abstract
The phosphatidylinositol 3-kinase (PI3K) pathway has been shown to play a central role in regulating the host inflammatory response. Recent studies characterizing the downstream effector molecules within the PI3K pathway have identified that the serine/threonine kinase, glycogen synthase kinase 3 (GSK3), plays a pivotal role in regulating the production of pro- and anti-inflammatory cytokines. In innate immune cells, GSK3 inactivation augments anti-inflammatory cytokine production while concurrently suppressing the production of pro-inflammatory cytokines. The role of GSK3 in T cell biology has also been studied in detail and is involved in regulating multiple downstream signaling processes mediated by the T cell receptor (TCR), the co-stimulatory molecule CD28, and the IL-17 receptor. In vivo studies assessing the therapeutic properties of GSK3 inhibitors have shown that the inactivation of GSK3 can protect the host from immune-mediated pathology and death. This review will highlight the immunological importance GSK3 plays within different signal transduction pathways of the immune system, the cellular mechanisms regulating the activity of GSK3, the role of GSK3 in innate and adaptive immune responses, and the in vivo use of GSK3 inhibitors to treat inflammatory mediated diseases in animals.
Collapse
Affiliation(s)
- Huizhi Wang
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | | | | |
Collapse
|
48
|
Ng SC, Kamm MA, Stagg AJ, Knight SC. Intestinal dendritic cells: their role in bacterial recognition, lymphocyte homing, and intestinal inflammation. Inflamm Bowel Dis 2010; 16:1787-807. [PMID: 20222140 DOI: 10.1002/ibd.21247] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) play a key role in discriminating between commensal microorganisms and potentially harmful pathogens and in maintaining the balance between tolerance and active immunity. The regulatory role of DC is of particular importance in the gut where the immune system lies in intimate contact with the highly antigenic external environment. Intestinal DC constantly survey the luminal microenvironment. They act as sentinels, acquiring antigens in peripheral tissues before migrating to secondary lymphoid organs to activate naive T cells. They are also sensors, responding to a spectrum of environmental cues by extensive differentiation or maturation. Recent studies have begun to elucidate mechanisms for functional specializations of DC in the intestine that may include the involvement of retinoic acid and transforming growth factor-β. Specialized CD103(+) intestinal DC can promote the differentiation of Foxp3(+) regulatory T cells via a retinoic acid-dependent process. Different DC outcomes are, in part, influenced by their exposure to microbial stimuli. Evidence is also emerging of the close interaction between bacteria, epithelial cells, and DC in the maintenance of intestinal immune homeostasis. Here we review recent advances of functionally specialized intestinal DC and their mechanisms of antigen uptake and recognition. We also discuss the interaction of DC with intestinal microbiota and their ability to orchestrate protective immunity and immune tolerance in the host. Lastly, we describe how DC functions are altered in intestinal inflammation and their emerging potential as a therapeutic target in inflammatory bowel disease.
Collapse
Affiliation(s)
- S C Ng
- Antigen Presentation Research Group, Faculty of Medicine, Imperial College London, Northwick Park and St Mark's Campus, Harrow, UK
| | | | | | | |
Collapse
|
49
|
Sánchez B, Urdaci MC, Margolles A. Extracellular proteins secreted by probiotic bacteria as mediators of effects that promote mucosa-bacteria interactions. MICROBIOLOGY-SGM 2010; 156:3232-3242. [PMID: 20864471 DOI: 10.1099/mic.0.044057-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
During the last few years, a substantial body of scientific evidence has accumulated suggesting that certain surface-associated and extracellular components produced by probiotic bacteria could be responsible for some of their mechanisms of action. These bacterial components would be able to directly interact with the host mucosal cells; they include exopolysaccharides, bacteriocins, lipoteichoic acids and surface-associated and extracellular proteins. Extracellular proteins include proteins that are actively transported to the bacterial surroundings through the cytoplasmic membrane, as well as those that are simply shed from the bacterial surface. Compared to the other bacterial components, the interactive ability of extracellular proteins/peptides has been less extensively studied. In this review, current findings supporting an interaction between extracellular proteins/peptides produced by probiotic bacteria (strains of the genera Bifidobacterium, Lactobacillus and Escherichia) and host mucosal cells are discussed. Research needs and future trends are also considered.
Collapse
Affiliation(s)
- Borja Sánchez
- Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300 Villaviciosa, Asturias, Spain
| | - María C Urdaci
- Université de Bordeaux, UMR 5248 CNRS, UBX1-ENITAB, ENITAB, 1 cours du Général de Gaulle, 33175 Gradignan Cedex, France
| | - Abelardo Margolles
- Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Ctra. Infiesto s/n, 33300 Villaviciosa, Asturias, Spain
| |
Collapse
|
50
|
Abstract
Since the discovery in 1899 of bifidobacteria as numerically dominant microbes in the feces of breast-fed infants, there have been numerous studies addressing their role in modulating gut microflora as well as their other potential health benefits. Because of this, they are frequently incorporated into foods as probiotic cultures. An understanding of their full interactions with intestinal microbes and the host is needed to scientifically validate any health benefits they may afford. Recently, the genome sequences of nine strains representing four species of Bifidobacterium became available. A comparative genome analysis of these genomes reveals a likely efficient capacity to adapt to their habitats, with B. longum subsp. infantis exhibiting more genomic potential to utilize human milk oligosaccharides, consistent with its habitat in the infant gut. Conversely, B. longum subsp. longum exhibits a higher genomic potential for utilization of plant-derived complex carbohydrates and polyols, consistent with its habitat in an adult gut. An intriguing observation is the loss of much of this genome potential when strains are adapted to pure culture environments, as highlighted by the genomes of B. animalis subsp. lactis strains, which exhibit the least potential for a gut habitat and are believed to have evolved from the B. animalis species during adaptation to dairy fermentation environments.
Collapse
Affiliation(s)
- Ju-Hoon Lee
- Department of Food Science and Nutrition, Microbial and Plant Genomics Institute, University of Minnesota, 1500 Gortner Ave., St. Paul, Minnesota 55108
| | - Daniel J. O'Sullivan
- Department of Food Science and Nutrition, Microbial and Plant Genomics Institute, University of Minnesota, 1500 Gortner Ave., St. Paul, Minnesota 55108
| |
Collapse
|