1
|
Akhlaghi E, Salari E, Mansouri M, Shafiei M, Kalantar-Neyestanaki D, Aghassi H, Fasihi Harandi M. Identification and comparison of intestinal microbial diversity in patients at different stages of hepatic cystic echinococcosis. Sci Rep 2024; 14:18912. [PMID: 39143364 PMCID: PMC11324937 DOI: 10.1038/s41598-024-70005-5] [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: 04/22/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024] Open
Abstract
There is a significant focus on the role of the host microbiome in different outcomes of human parasitic diseases, including cystic echinococcosis (CE). This study was conducted to identify the intestinal microbiome of patients with CE at different stages of hydatid cyst compared to healthy individuals. Stool samples from CE patients as well as healthy individuals were collected. The samples were divided into three groups representing various stages of hepatic hydatid cyst: active (CE1 and CE2), transitional (CE3), and inactive (CE4 and CE5). One family member from each group was selected to serve as a control. The gut microbiome of patients with different stages of hydatid cysts was investigated using metagenomic next-generation amplicon sequencing of the V3-V4 region of the 16S rRNA gene. In this study, we identified 4862 Operational Taxonomic Units from three stages of hydatid cysts in CE patients and healthy individuals with a combined frequency of 2,955,291. The most abundant genera observed in all the subjects were Blautia, Agathobacter, Faecalibacterium, Bacteroides, Bifidobacterium, and Prevotella. The highest microbial frequency was related to inactive forms of CE, and the lowest frequency was observed in the group with active forms. However, the lowest OTU diversity was found in patients with inactive cysts compared with those with active and transitional cyst stages. The genus Agatobacter had the highest OTU frequency. Pseudomonas, Gemella, and Ligilactobacillus showed significant differences among the patients with different stages of hydatid cysts. Additionally, Anaerostipes and Candidatus showed significantly different reads in CE patients compared to healthy individuals. Our findings indicate that several bacterial genera can play a role in the fate of hydatid cysts in patients at different stages of the disease.
Collapse
Affiliation(s)
- Elham Akhlaghi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Salari
- Department of Plant Protection, Faculty of Plant Production, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mehdi Mansouri
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Mohammad Shafiei
- Research Center for Hydatid Disease in Iran, Department of Surgery, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Davood Kalantar-Neyestanaki
- Medical Mycology and Bacteriology Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Medical Microbiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Aghassi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Fasihi Harandi
- Research Center for Hydatid Disease in Iran, Department of Medical Parasitology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Rocchetti MT, Russo P, De Simone N, Capozzi V, Spano G, Fiocco D. Immunomodulatory Activity on Human Macrophages by Cell-Free Supernatants to Explore the Probiotic and Postbiotic Potential of Lactiplantibacillus plantarum Strains of Plant Origin. Probiotics Antimicrob Proteins 2024; 16:911-926. [PMID: 37202651 PMCID: PMC11126452 DOI: 10.1007/s12602-023-10084-4] [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] [Accepted: 05/02/2023] [Indexed: 05/20/2023]
Abstract
Upon dietary administration, probiotic microorganisms can reach as live cells the human gut, where they interact with the microbiota and host cells, thereby exerting a beneficial impact on host functions, mainly through immune-modulatory activities. Recently, attention has been drawn by postbiotics, i.e. non-viable probiotic microbes, including their metabolic products, which possess biological activities that benefit the host. Lactiplantibacillus plantarum is a bacterial species that comprises recognised probiotic strains. In this study, we investigated in vitro the probiotic (and postbiotic) potential of seven L. plantarum strains, including five newly isolated from plant-related niches. The strains were shown to possess some basic probiotic attributes, including tolerance to the gastrointestinal environment, adhesion to the intestinal epithelium and safety. Besides, their cell-free culture supernatants modulated cytokine patterns in human macrophages in vitro, promoting TNF-α gene transcription and secretion, while attenuating the transcriptional activation and secretion of both TNF-α and IL-8 in response to a pro-inflammatory signal, and enhancing the production of IL-10. Some strains induced a high IL-10/IL-12 ratio that may correlate to an anti-inflammatory capacity in vivo. Overall, the investigated strains are good probiotic candidates, whose postbiotic fraction exhibits immunomodulatory properties that need further in vivo studies. The main novelty of this work consists in the polyphasic characterisation of candidate beneficial L. plantarum strains obtained from relatively atypical plant-associated niches, by an approach that explores both probiotic and postbiotic potentials, in particular studying the effect of microbial culture-conditioned media on cytokine pattern, analysed at both transcriptional and secretion level in human macrophages.
Collapse
Affiliation(s)
| | - Pasquale Russo
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Nicola De Simone
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, Foggia, Italy
| | - Vittorio Capozzi
- Institute of Sciences of Food Production, National Research Council (CNR) of Italy, C/O CS-DAT, Foggia, Italy
| | - Giuseppe Spano
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, Foggia, Italy
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy.
| |
Collapse
|
3
|
Kaur G, Kumar A, Kurl S, Mittal N, Malik DS, Bassi P, Singh T, Khan AA, Alanazi AM, Kaur G. Leucaena leucocephala succinate based polyelectrolyte complexes for colon delivery of synbiotic in management of inflammatory bowel disease. Heliyon 2024; 10:e29429. [PMID: 38628770 PMCID: PMC11017066 DOI: 10.1016/j.heliyon.2024.e29429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 03/17/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Polyelectrolyte complexes (PECs) formed by the interaction between oppositely charged polymers have emerged as promising carriers for accomplishing colon-specific release. In this study, we have explored the potential of polyelectrolyte complexes between a succinate derivative of Leucaena leucocephala galactomannan and cationic guar gum for colon delivery of synbiotic. The PECs were prepared using a polyelectrolyte complexation method and characterized. The PECs exhibited excellent stability, with high encapsulation efficiency for both probiotics (95.53 %) and prebiotics (83.33 %). In vitro studies demonstrated enhanced survivability and proliferation of the encapsulated probiotics in the presence of prebiotics (93.29 %). The SEM images revealed a smooth and firm structure with reduced number of pores when both prebiotic and probiotic were encapsulated together. The treatment with synbiotic PECs in acetic acid induced IBD rats significantly relieves colitis symptoms as was evident from colon/body ratio, DAI score and histopathology studies. An increase in the protein and reduced glutathione levels and reduction in superoxide dismutase activity was observed in colitic rats that received synbiotic treatment as compared to colitic rats. Overall, this study highlights the potential of Leucaena leucocephala succinate-cationic guar gum PECs as a promising system for colon-specific synbiotic delivery, with implications for improved gut health and the treatment of various gastrointestinal disorders.
Collapse
Affiliation(s)
- Gaganpreet Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Punjab, 147002, India
| | - Aman Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Punjab, 147002, India
| | - Samridhi Kurl
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Punjab, 147002, India
| | - Neeraj Mittal
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | - Pallavi Bassi
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Tanveer Singh
- Department of Neuroscience and Experimental Therapeutics, Medical Research, College of Medicine, Bryan, 77807, USA
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Amer M. Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Gurpreet Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Punjab, 147002, India
| |
Collapse
|
4
|
Carbonne C, Chadi S, Kropp C, Molimard L, Chain F, Langella P, Martin R. Ligilactobacillus salivarius CNCM I-4866, a potential probiotic candidate, shows anti-inflammatory properties in vitro and in vivo. Front Microbiol 2023; 14:1270974. [PMID: 38094624 PMCID: PMC10716304 DOI: 10.3389/fmicb.2023.1270974] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 04/19/2024] Open
Abstract
INTRODUCTION The aim of this work was to characterize a new strain of Ligilactobacillus salivarius (CNCM I-4866) (CNCM I-4866) to address its potential as probiotic with a special focus on intestinal inflammation. Potential anti-inflammatory abilities of this strain were evaluated through in vivo and in vitro experiments. METHODS Firstly, the strain was tested in a murine acute inflammation colitis model induced by DNBS. In vitro characterization was then performed with diverse tests: modulation capability of intestinal permeability; study of the impact on immunity profile through cytokines dosage; capacity to inhibit pathogens and adhere to intestinal cells lines. Production of metabolites, antibiotic resistance and survival to gastro-intestinal tract conditions were also tested. RESULTS In vitro assay has shown a reduction of colonic damage and markers of inflammation after treatment with CNCM I-4866. Transcriptomic analysis performed on colons showed the capacity of the strain to down-regulate pro-inflammatory cytokines. L. salivarius CNCM I-4866 exerted anti-inflammatory profile by reducing IL-8 production by TNF-α stimulated cell and modulated cytokines profile on peripheral blood mononuclear cells (PBMC). It protected intestinal integrity by increasing trans-epithelial electrical resistance (TEER) on Caco-2 TNF-α inflamed cells. Additionally, L. salivarius CNCM I-4866 displayed inhibition capacity on several intestinal pathogens and adhered to eukaryotic cells. Regarding safety and technical concerns, CNCM I-4866 was highly resistant to 0.3% of bile salts and produced mainly L-lactate. Finally, strain genomic characterization allowed us to confirm safety aspect of our strain, with no antibiotic gene resistance found. DISCUSSION Taken together, these results indicate that L. salivarius CNCM I-4866 could be a good probiotic candidate for intestinal inflammation, especially with its steady anti-inflammatory profile.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rebeca Martin
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
5
|
Shang J, Yang S, Tang Z, Chen Y, Duan B, Meng X. Bifidobacterium bifidum H3-R2 and Its Molecular Communication within the Context of Ulcerative Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11678-11688. [PMID: 36095239 DOI: 10.1021/acs.jafc.2c02909] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bifidobacteria are important mediators of immune system development within the gastrointestinal system and immunological homeostasis. The present study explored the anti-colitic activity of Bifidobacterium bifidum H3-R2 in a murine dextran sulfate sodium (DSS)-induced model of ulcerative colitis (UC). Moreover, this study offers novel insight regarding the molecular basis for the probiotic properties of B. bifidum H3-R2 by analyzing the underlying mechanisms whereby B. bifidum H3-R2-derived proteins affect the intestinal barrier. B. bifidum H3-R2 administration was sufficient to alleviate clinical manifestations consistent with DSS-induced colitis, restoring aberrant inflammatory cytokine production, enhancing tight junction protein expression, and positively impacting overall intestinal microecological homeostasis in these animals. Moreover, the bifidobacteria-derived GroEL and transaldolase (TAL) proteins were found to regulate tight junction protein expression via the NF-κB, myosin light chain kinase (MLCK), RhoA/Rho-associated protein kinase (ROCK), and mitogen-activated protein kinase (MAPK) signaling pathways, preventing the lipopolysaccharide (LPS)-mediated disruption of the intestinal epithelial cell barrier.
Collapse
Affiliation(s)
- Jiacui Shang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuo Yang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Zongxin Tang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yuhan Chen
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bofan Duan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Food College, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
6
|
Li X, Su L, Zhang X, Chen Q, Wang Y, Shen Z, Zhong T, Wang L, Xiao Y, Feng X, Yu X. Recent Advances on the Function and Purification of Milk Exosomes: A Review. Front Nutr 2022; 9:871346. [PMID: 35757254 PMCID: PMC9219579 DOI: 10.3389/fnut.2022.871346] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/12/2022] [Indexed: 11/20/2022] Open
Abstract
Exosomes are nano-scale extracellular vesicles, which can be used as drug carriers, tumor treatment, intestinal development and immune regulator. That is why it has great potential in pharmacy, functional foods, nutritional supplements, especially those for infants, postoperative patients, chemotherapy patients and the elderly. In addition, abnormal exosome level is also related to diseases such as cardiovascular diseases, tumor, diabetes, neurodegenerative and autoimmune diseases, as well as infectious diseases. Despite its high biological significance, pharmaceutical and nutritional value, the low abundancy of exosomes in milk is one of the bottlenecks restricting its in-depth research and real-life application. At present, there is no unified standard for the extraction of breast milk exosomes. Therefore, choosing the proper extraction method is very critical for its subsequent research and development. Based on this, this paper reviewed the purification techniques, the function and the possible applications of milk exosomes based on 47 latest references. Humble advices on future directions, prospects on new ideas and methods which are useful for the study of exosomes are proposed at the end of the paper as well.
Collapse
Affiliation(s)
- Xiaoping Li
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Lan Su
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xinling Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Qi Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ying Wang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zhenwei Shen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Tian Zhong
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ling Wang
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ying Xiao
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- Guangdong-Hong Kong-Macau Joint Laboratory for Contaminants Exposure and Health, Guangzhou, China
| | - Xiao Feng
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Xi Yu
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
- Guangdong-Hong Kong-Macau Joint Laboratory for Contaminants Exposure and Health, Guangzhou, China
| |
Collapse
|
7
|
Li S, Li N, Wang C, Zhao Y, Cao J, Li X, Zhang Z, Li Y, Yang X, Wang X, Che C, Zhao Y, Wang L, Zhao L, Shen J. Gut Microbiota and Immune Modulatory Properties of Human Breast Milk Streptococcus salivarius and S. parasanguinis Strains. Front Nutr 2022; 9:798403. [PMID: 35273986 PMCID: PMC8901577 DOI: 10.3389/fnut.2022.798403] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/24/2022] [Indexed: 01/14/2023] Open
Abstract
Human breast milk Streptococcus spp. are transferred to infant guts via breast feeding, but their effects on the gut microbiota and immunity remain unclear. In this study, we characterized gut microbiota and immune modulatory properties of human breast milk S. salivarius F286 and S. parasanguinis F278 that had been shown to be able to colonize gut. The two Streptococcus strains were orally administered to mouse pups individually at 1 × 107 cells/day from postnatal Days 1 to 21. At postnatal week 3 (the weaning period), S. salivarius F286 reduced the colonic microbiota α-diversity, increased 21 amplicon sequence variants (ASVs), including bacteria from Akkermansia, Intestinimonas, and Lachnospiraceae, and decreased 52 ASVs, including bacteria from Eubacterium, Bifidobacterium, Escherichia-Shigella, and Turicibacter; however, S. parasanguinis F278 didn't change the colonic microbiota. Both Streptococcus strains reduced the ileal mRNA expression of cytokine/transcription factor representatives of T helper (Th) cells, including IFN-γ (Th1), Gata3 (Th2), and TGF-β (Treg) in 2-week-old suckling mice, and promoted the ileal expression of Foxp3 and TGF-β, which are representatives of anti-inflammatory Treg cells, in 3-week-old weaning mice. The two Streptococcus strains exhibited anti-inflammatory potential when incubated in vitro with human peripheral blood mononuclear cells and TNF-α-treated gut epithelial HT29 cells. In C. elegans, both strains activated immune response genes, which was associated with their lifespan-prolonging effects. Our results suggest that S. salivarius F286 and S. parasanguinis F278 may exert regulatory (anti-inflammatory) roles in gut immunity and S. salivarius F286 can modulate gut microbiota, and highlight the probiotic potential of milk S. salivarius and S. parasanguinis strains.
Collapse
Affiliation(s)
- Shuo Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Na Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenwei Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Cao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuejing Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxin Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanyan Che
- Department of Animal Sciences, Anhui Science and Technology University, Chuzhou, China
| | - Yufeng Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linghua Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liping Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Shen
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
Guerrero Sanchez M, Passot S, Campoy S, Olivares M, Fonseca F. Ligilactobacillus salivarius functionalities, applications, and manufacturing challenges. Appl Microbiol Biotechnol 2021; 106:57-80. [PMID: 34889985 DOI: 10.1007/s00253-021-11694-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022]
Abstract
Ligilactobacillus salivarius is a lactic acid bacteria that has been gaining attention as a promising probiotic. Numerous strains exhibit functional properties with health benefits such as antimicrobial activity, immunological effects, and the ability to modulate the intestinal microbiota. However, just a small number of them are manufactured at an industrial scale and included in commercial products. The under exploitation of L. salivarius strains that remain in the freezer of companies is due to their incapacity to overcome the environmental stresses induced by production and stabilization processes.The present study summarizes the functionalities and applications of L. salivarius reported to date. It aims also at providing a critical evaluation of the literature available on the manufacturing steps of L. salivarius concentrates, the bacterial quality after each step of the process, and the putative degradation and preservation mechanisms. Here, we highlight the principal issues and future research challenges for improving the production and long-term preservation at the industrial scale of this microorganism, and probably of other probiotics.Key points• L. salivarius beneficial properties and commercialized products.• Production conditions and viability of L. salivarius after stabilization processes.• Prospects for identifying preservation mechanisms to improve L. salivarius stability.
Collapse
Affiliation(s)
| | - S Passot
- Université Paris-Saclay, INRAE, AgroParisTech, UMR SayFood, 78850, Thiverval-Grignon, France
| | - S Campoy
- R&D Department, Biosearch Life, 18004, Granada, Spain
| | - M Olivares
- R&D Department, Biosearch Life, 18004, Granada, Spain
| | - F Fonseca
- Université Paris-Saclay, INRAE, AgroParisTech, UMR SayFood, 78850, Thiverval-Grignon, France.
| |
Collapse
|
9
|
Han T, Hu X, Li K, Zhang D, Zhang Y, Li J. Bifidobacterium infantis Maintains Genome Stability in Ulcerative Colitis via Regulating Anaphase-Promoting Complex Subunit 7. Front Microbiol 2021; 12:761113. [PMID: 34795654 PMCID: PMC8593188 DOI: 10.3389/fmicb.2021.761113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Probiotics represents a promising intestinal microbiota-targeted therapeutic method for the treatment of ulcerative colitis (UC). Several lines of evidence implicate that Bifidobacterium infantis serves as a probiotic strain with proven efficacy in maintaining the remission of UC. However, the exact mechanisms underlying the beneficial effects of B. infantis on UC progression have yet to be elucidated. Herein, we provide evidence that B. infantis acts as a key predisposing factor for the maintenance of host genome stability. First, we showed that the fecal microbiota transplantation (FMT) of UC-derived feces contributes to more severely DNA damage in dextran sodium sulfate (DSS)-induced mice likely due to mucosa-associated microbiota alterations, as reflected by the rapid appearance of DNA double strand breaks (DSBs), a typical marker of genome instability. Genomic DNA damage analysis of colon tissues derived from healthy controls, patients with UC or dysplasia, and colitis associated cancer (CAC) patients, revealed an enhanced level of DSBs with aggravation in the degree of the intestinal mucosal lesions. To evaluate whether B. infantis modulates the host genome stability, we employed the DSS-induced colitis model and a TNFα-induced intestinal epithelial cell model. Following the administration of C57BL/6 mice with B. infantis via oral gavage, we found that the development of DSS-induced colitis in mice was significantly alleviated, in contrast to the colitis model group. Notably, B. infantis administration decreased DSB levels in both DSS-induced colitis and TNF-treated colonial cell model. Accordingly, our bioinformatic and functional studies demonstrated that B. infantis altered signal pathways involved in ubiquitin-mediated proteolysis, transcriptional misregulation in cancer, and the bacterial invasion of epithelial cells. Mechanistically, B. infantis upregulated anaphase-promoting complex subunit 7 (APC7), which was significantly suppressed in colitis condition, to activate the DNA repair pathway and alter the genome stability, while downregulation of APC7 abolished the efficiency of B. infantis treatment to induce a decrease in the level of DSBs in TNFα-induced colonial cells. Collectively, our results support that B. infantis orchestrates a molecular network involving in APC7 and genome stability, to control UC development at the clinical, biological, and mechanistic levels. Supplying B. infantis and targeting its associated pathway will yield valuable insight into the clinical management of UC patients.
Collapse
Affiliation(s)
- Taotao Han
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaomin Hu
- Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kemin Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Di Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Piccioni A, Franza L, Vaccaro V, Saviano A, Zanza C, Candelli M, Covino M, Franceschi F, Ojetti V. Microbiota and Probiotics: The Role of Limosilactobacillus Reuteri in Diverticulitis. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57080802. [PMID: 34441008 PMCID: PMC8398895 DOI: 10.3390/medicina57080802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 02/05/2023]
Abstract
The microbiota is the set of commensal microorganisms, residing in the organism, helping proper functioning of organs and systems. The role that the microbiota plays in maintaining the health of vertebrates is widely accepted, particularly in the gastrointestinal system, where it is fundamental for immunity, development, and conversion of nutrients. Dysbiosis is an alteration of the microbiota which refers to a disturbed balance, which can cause a number of pathologies. Probiotics have proven to be effective in modulating the microbiota of the gastrointestinal system and, therefore, in promoting the health of the individual. In particular, Lactobacilli are a group of Gram-positive bacteria, which are able to produce lactic acid through glucose metabolism. They are present in different microenvironments, ranging from the vagina, to the mouth, to different tracts of the small intestine. In the present review, we will discuss the use of Limosilactobacillus in human health in general and more specifically in diverticulitis. In particular we analyze the role of Limosilactobacillus reuteri and its anti-inflammatory action. For this review, articles were identified using the electronic PubMed database through a comprehensive search, conducted by combining key terms such as "diverticulitis", "Limosilactobacillus reuteri", "human health and disease", "probiotics". We selected all the articles published in the last 10 years and screened 1017 papers. Articles referenced in the screened papers were evaluated if considered interesting for our topic. Probiotics have proven to be effective in modulating the microbiota of the gastrointestinal system and, therefore, in promoting the health of the individual. The importance of probiotics in treating diverticular disease and acute diverticulitis can be further understood if taking into consideration some pathophysiological aspects, associated to the microbiota. L. reuteri plays an important role in human health and disease. The effectiveness of L. reuteri in stimulating a correct bowl motility partly explains its effectiveness in treating diverticulitis. The most important action of L. reuteri is probably its immunomodulating activity. Levels of IL-6, IL-8, and Tumor necrosis factor (TNF-alpha) are reduced after supplementation with different strands of Lactobacilli, while T-regulatory cells increase in number and activity. Anyway, new mechanisms of action of probiotics come to light from the many investigations currently taking place in numerous centres around the world and to improve how exactly probiotic administration could make the difference in the management of diverticular disease and acute diverticulitis.
Collapse
Affiliation(s)
- Andrea Piccioni
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Roma, Italy; (M.C.); (M.C.); (F.F.)
- Correspondence:
| | - Laura Franza
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| | - Vanessa Vaccaro
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| | - Angela Saviano
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| | - Christian Zanza
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| | - Marcello Candelli
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Roma, Italy; (M.C.); (M.C.); (F.F.)
| | - Marcello Covino
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Roma, Italy; (M.C.); (M.C.); (F.F.)
| | - Francesco Franceschi
- Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Roma, Italy; (M.C.); (M.C.); (F.F.)
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| | - Veronica Ojetti
- Emergency Department, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (L.F.); (V.V.); (A.S.); (C.Z.); (V.O.)
| |
Collapse
|
11
|
Piccioni A, Franza L, Brigida M, Zanza C, Torelli E, Petrucci M, Nicolò R, Covino M, Candelli M, Saviano A, Ojetti V, Franceschi F. Gut Microbiota and Acute Diverticulitis: Role of Probiotics in Management of This Delicate Pathophysiological Balance. J Pers Med 2021; 11:jpm11040298. [PMID: 33919818 PMCID: PMC8070761 DOI: 10.3390/jpm11040298] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/05/2023] Open
Abstract
How can the knowledge of probiotics and their mechanisms of action be translated into clinical practice when treating patients with diverticular disease and acute diverticulitis? Changes in microbiota composition have been observed in patients who were developing acute diverticulitis, with a reduction of taxa with anti-inflammatory activity, such as Clostridium cluster IV, Lactobacilli and Bacteroides. Recent observations supported that a dysbiosis characterised by decreased presence of anti-inflammatory bacterial species might be linked to mucosal inflammation, and a vicious cycle results from a mucosal inflammation driving dysbiosis at the same time. An alteration in gut microbiota can lead to an altered activation of nerve fibres, and subsequent neuronal and muscular dysfunction, thus favoring abdominal symptoms' development. The possible role of dysbiosis and mucosal inflammation in leading to dysmotility is linked, in turn, to bacterial translocation from the lumen of the diverticulum to perivisceral area. There, a possible activation of Toll-like receptors has been described, with a subsequent inflammatory reaction at the level of the perivisceral tissues. Being aware that bacterial colonisation of diverticula is involved in the pathogenesis of acute diverticulitis, the rationale for the potential role of probiotics in the treatment of this disease becomes clearer. For this review, articles were identified using the electronic PubMed database through a comprehensive search conducted by combining key terms such as "gut microbiota", "probiotics and gut disease", "probiotics and acute diverticulitis", "probiotics and diverticular disease", "probiotics mechanism of action". However, the amount of data present on this matter is not sufficient to draw robust conclusions on the efficacy of probiotics for symptoms' management in diverticular disease.
Collapse
Affiliation(s)
- Andrea Piccioni
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Correspondence:
| | - Laura Franza
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Mattia Brigida
- Unit of Gastroenterology, Department of Systems Medicine, Tor Vergata University, 2-00133 Rome, Italy;
| | - Christian Zanza
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Enrico Torelli
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Martina Petrucci
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Rebecca Nicolò
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Marcello Covino
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
| | - Marcello Candelli
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
| | - Angela Saviano
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Veronica Ojetti
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| | - Francesco Franceschi
- Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, 1-00168 Rome, Italy; (M.C.); (M.C.); (V.O.); (F.F.)
- Università Cattolica del Sacro Cuore, 1-00168 Rome, Italy; (L.F.); (C.Z.); (E.T.); (M.P.); (R.N.); (A.S.)
| |
Collapse
|
12
|
Udayan S, Buttó LF, Rossini V, Velmurugan J, Martinez-Lopez M, Sancho D, Melgar S, O'Toole PW, Nally K. Macrophage cytokine responses to commensal Gram-positive Lactobacillus salivarius strains are TLR2-independent and Myd88-dependent. Sci Rep 2021; 11:5896. [PMID: 33723368 PMCID: PMC7961041 DOI: 10.1038/s41598-021-85347-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 02/26/2021] [Indexed: 01/31/2023] Open
Abstract
The mechanisms through which cells of the host innate immune system distinguish commensal bacteria from pathogens are currently unclear. Toll-like receptors (TLRs) are a class of pattern recognition receptors (PRRs) expressed by host cells which recognize microbe-associated molecular patterns (MAMPs) common to both commensal and pathogenic bacteria. Of the different TLRs, TLR2/6 recognize bacterial lipopeptides and trigger cytokines responses, especially to Gram-positive and Gram-negative pathogens. We report here that TLR2 is dispensable for triggering macrophage cytokine responses to different strains of the Gram-positive commensal bacterial species Lactobacillus salivarius. The L. salivarius UCC118 strain strongly upregulated expression of the PRRs, Mincle (Clec4e), TLR1 and TLR2 in macrophages while downregulating other TLR pathways. Cytokine responses triggered by L. salivarius UCC118 were predominantly TLR2-independent but MyD88-dependent. However, macrophage cytokine responses triggered by another Gram-positive commensal bacteria, Bifidobacterium breve UCC2003 were predominantly TLR2-dependent. Thus, we report a differential requirement for TLR2-dependency in triggering macrophage cytokine responses to different commensal Gram-positive bacteria. Furthermore, TNF-α responses to the TLR2 ligand FSL-1 and L. salivarius UCC118 were partially Mincle-dependent suggesting that PRR pathways such as Mincle contribute to the recognition of MAMPs on distinct Gram-positive commensal bacteria. Ultimately, integration of signals from these different PRR pathways and other MyD88-dependent pathways may determine immune responses to commensal bacteria at the host-microbe interface.
Collapse
Affiliation(s)
- Sreeram Udayan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | | | - Valerio Rossini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Maria Martinez-Lopez
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Silvia Melgar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Paul W O'Toole
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Ken Nally
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
13
|
Fernández L, Pannaraj PS, Rautava S, Rodríguez JM. The Microbiota of the Human Mammary Ecosystem. Front Cell Infect Microbiol 2020; 10:586667. [PMID: 33330129 PMCID: PMC7718026 DOI: 10.3389/fcimb.2020.586667] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk contains a dynamic and complex site-specific microbiome, which is not assembled in an aleatory way, formed by organized microbial consortia and networks. Presence of some genera, such as Staphylococcus, Streptococcus, Corynebacterium, Cutibacterium (formerly known as Propionibacterium), Lactobacillus, Lactococcus and Bifidobacterium, has been detected by both culture-dependent and culture-independent approaches. DNA from some gut-associated strict anaerobes has also been repeatedly found and some studies have revealed the presence of cells and/or nucleic acids from viruses, archaea, fungi and protozoa in human milk. Colostrum and milk microbes are transmitted to the infant and, therefore, they are among the first colonizers of the human gut. Still, the significance of human milk microbes in infant gut colonization remains an open question. Clinical studies trying to elucidate the question are confounded by the profound impact of non-microbial human milk components to intestinal microecology. Modifications in the microbiota of human milk may have biological consequences for infant colonization, metabolism, immune and neuroendocrine development, and for mammary health. However, the factors driving differences in the composition of the human milk microbiome remain poorly known. In addition to colostrum and milk, breast tissue in lactating and non-lactating women may also contain a microbiota, with implications in the pathogenesis of breast cancer and in some of the adverse outcomes associated with breast implants. This and other open issues, such as the origin of the human milk microbiome, and the current limitations and future prospects are addressed in this review.
Collapse
Affiliation(s)
- Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Pia S. Pannaraj
- Department of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine and Children’s Hospital, Los Angeles, CA, United States
| | - Samuli Rautava
- University of Helsinki and Helsinki University Hospital, New Children’s Hospital, Pediatric Research Center, Helsinki, Finland
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
14
|
Shang J, Wan F, Zhao L, Meng X, Li B. Potential Immunomodulatory Activity of a Selected Strain Bifidobacterium bifidum H3-R2 as Evidenced in vitro and in Immunosuppressed Mice. Front Microbiol 2020; 11:2089. [PMID: 32983062 PMCID: PMC7491056 DOI: 10.3389/fmicb.2020.02089] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/08/2020] [Indexed: 12/30/2022] Open
Abstract
The microbiota is directly involved in the development and modulation of the intestinal immune system. In particular, members of the genus Bifidobacterium play a primary role in immune regulation. In the present study, Bifidobacterium bifidum H3-R2 was screened from 15 bifidobacterium strains by in vitro experiment, showing a positive tolerance to digestive tract conditions, adhesion ability to intestinal epithelial cells and a regulatory effect on immune cell activity. Immunostimulatory activity of B. bifidum H3-R2 was also elucidated in vivo in cytoxan (CTX)-treated mice. The results showed that the administration of B. bifidum H3-R2 ameliorated the CTX-induced bodyweight loss and imbalanced expression of inflammatory cytokines, enhanced the production of secretory immunoglobulin A (SIgA), and promoted splenic lymphocyte proliferation, natural killer (NK) cell activity and phagocytosis of macrophages in immunosuppressed mice. In addition, B. bifidum H3-R2 restored injured intestinal mucosal, and increased the villus length and crypt depth in CTX-treated mice. The results could be helpful for understanding the functions of B. bifidum H3-R2, supporting its potential as a novel probiotic for immunoregulation.
Collapse
Affiliation(s)
- Jiacui Shang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Feng Wan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Le Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China.,School of Food Science, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China.,School of Food Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
15
|
Wang J, Ishfaq M, Guo Y, Chen C, Li J. Assessment of Probiotic Properties of Lactobacillus salivarius Isolated From Chickens as Feed Additives. Front Vet Sci 2020; 7:415. [PMID: 32766298 PMCID: PMC7379216 DOI: 10.3389/fvets.2020.00415] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022] Open
Abstract
The continued use of sub-therapeutic antibiotics as feed additives in the poultry industry improved health and growth performance. However, the resulting antibiotic resistance increasingly becomes a major threat to public health. Probiotics are promising alternatives for the antibiotics used in poultry industry. The aim of this study was to evaluate the probiotic properties of Lactobacillus salivarius as feed additive in chickens. White leghorn chickens were randomly assigned to experimental groups. Effects of Lactobacillus salivarius supplementation on growth performance, resistance to Escherichia coli O78 challenge and heat-stress, and immune response after vaccinated with attenuated infectious bursal disease virus (IBDV) vaccine were determined. The results showed that Lactobacillus salivarius supplementation improved growth performance, such as weight and longer shank length, increased relative weights of the immune organs and decreased concentrations of odor-causing compounds. In addition, Lactobacillus salivarius supplementation alleviated organ injury caused by Escherichia coli O78 challenge and heat stress. Furthermore, Lactobacillus salivarius results in enhanced immune response after IBDV vaccine immunization, enhanced specific antibody and IFN-γ production, and lymphocyte proliferation. Our results revealed a tremendous potential of Lactobacillus salivarius as antibiotics' substitute in poultry production.
Collapse
Affiliation(s)
- Jian Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Muhammad Ishfaq
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuquan Guo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chunli Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, China
| |
Collapse
|
16
|
Chen C, Ahn EH, Kang SS, Liu X, Alam A, Ye K. Gut dysbiosis contributes to amyloid pathology, associated with C/EBPβ/AEP signaling activation in Alzheimer's disease mouse model. SCIENCE ADVANCES 2020; 6:eaba0466. [PMID: 32832679 PMCID: PMC7439296 DOI: 10.1126/sciadv.aba0466] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/15/2020] [Indexed: 05/02/2023]
Abstract
The gut-brain axis is bidirectional, and gut microbiota influence brain disorders including Alzheimer's disease (AD). CCAAT/enhancer binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling spatiotemporally mediates AD pathologies in the brain via cleaving both β-amyloid precursor protein and Tau. We show that gut dysbiosis occurs in 5xFAD mice, and is associated with escalation of the C/EBPβ/AEP pathway in the gut with age. Unlike that of aged wild-type mice, the microbiota of aged 3xTg mice accelerate AD pathology in young 3xTg mice, accompanied by active C/EBPβ/AEP signaling in the brain. Antibiotic treatment diminishes this signaling and attenuates amyloidogenic processes in 5xFAD, improving cognitive functions. The prebiotic R13 inhibits this pathway and suppresses amyloid aggregates in the gut. R13-induced Lactobacillus salivarius antagonizes the C/EBPβ/AEP axis, mitigating gut leakage and oxidative stress. Our findings support the hypothesis that C/EBPβ/AEP signaling is activated by gut dysbiosis, implicated in AD pathologies in the gut.
Collapse
Affiliation(s)
- Chun Chen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Eun Hee Ahn
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | - Ashfaqul Alam
- Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Bifidobacterium animalis subsp. lactis 420 for Metabolic Health: Review of the Research. Nutrients 2020; 12:nu12040892. [PMID: 32218248 PMCID: PMC7230722 DOI: 10.3390/nu12040892] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
The growing worldwide epidemic of obesity and associated metabolic health comorbidities has resulted in an urgent need for safe and efficient nutritional solutions. The research linking obesity with gut microbiota dysbiosis has led to a hypothesis that certain bacterial strains could serve as probiotics helping in weight management and metabolic health. In the search for such strains, the effect of Bifidobacterium animalis subsp. lactis 420 (B420) on gut microbiota and metabolic health, and the mechanisms of actions, has been investigated in a variety of in vitro, pre-clinical, and clinical studies. In this review, we aim to highlight the research on B420 related to obesity, metabolic health, and the microbiota. Current research supports the hypothesis that gut dysbiosis leads to an imbalance in the inflammatory processes and loss of epithelial integrity. Bacterial components, like endotoxins, that leak out of the gut can invoke low-grade, chronic, and systemic inflammation. This imbalanced state is often referred to as metabolic endotoxemia. Scientific evidence indicates that B420 can slow down many of these detrimental processes via multiple signaling pathways, as supported by mechanistic in vitro and in vivo studies. We discuss the connection of these mechanisms to clinical evidence on the effect of B420 in controlling weight gain in overweight and obese subjects. The research further indicates that B420 may improve the epithelial integrity by rebalancing a dysbiotic state induced by an obesogenic diet, for example by increasing the prevalence of lean phenotype microbes such as Akkermansia muciniphila. We further discuss, in the context of delivering the health benefits of B420: the safety and technological aspects of the strain including genomic characterization, antibiotic resistance profiling, stability in the product, and survival of the live probiotic in the intestine. In summary, we conclude that the clinical and preclinical studies on metabolic health suggest that B420 may be a potential candidate in combating obesity; however, further clinical studies are needed.
Collapse
|
18
|
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease, ulcerative colitis, and pouchitis, are chronic, relapsing intestinal inflammatory disorders mediated by dysregulated immune responses to resident microbiota. Current standard therapies that block immune activation with oral immunosuppressives or biologic agents are generally effective, but each therapy induces a sustained remission in only a minority of patients. Furthermore, these approaches can have severe adverse events. Recent compelling evidence of a role of unbalanced microbiota (dysbiosis) driving immune dysfunction and inflammation in IBD supports the therapeutic rationale for manipulating the dysbiotic microbiota. Traditional approaches using currently available antibiotics, probiotics, prebiotics, and synbiotics have not produced optimal results, but promising outcomes with fecal microbiota transplant provide a proof of principle for targeting the resident microbiota. Rationally designed oral biotherapeutic products (LBPs) composed of mixtures of protective commensal bacterial strains demonstrate impressive preclinical results. Resident microbial-based and microbial-targeted therapies are currently being studied with increasing intensity for IBD primary therapy with favorable early results. This review presents current evidence and therapeutic mechanisms of microbiota modulation, emphasizing clinical studies, and outlines prospects for future IBD treatment using new approaches, such as LBPs, bacteriophages, bacterial function-editing substrates, and engineered bacteria. We believe that the optimal clinical use of microbial manipulation may be as adjuvants to immunosuppressive for accelerated and improved induction of deep remission and as potential safer solo approaches to sustained remission using personalized regimens based on an individual patient's microbial profile.
Collapse
Affiliation(s)
- Akihiko Oka
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.
- National Gnotobiotic Rodent Resource Center, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27514, USA.
| |
Collapse
|
19
|
Rather IA, Bajpai VK, Ching LL, Majumder R, Nam GJ, Indugu N, Singh P, Kumar S, Hajrah NH, Sabir JS, Kamli MR, Park YH. Effect of a bioactive product SEL001 from Lactobacillus sakei probio65 on gut microbiota and its anti-colitis effects in a TNBS-induced colitis mouse model. Saudi J Biol Sci 2020; 27:261-270. [PMID: 31889846 PMCID: PMC6933275 DOI: 10.1016/j.sjbs.2019.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/25/2019] [Accepted: 09/03/2019] [Indexed: 01/02/2023] Open
Abstract
This study underpins the therapeutic potential of SEL001, a bioactive product isolated from Lactobacillus sakei probio65, in terms of its anti-inflammatory properties and its effect on gut-microbiota in a TNBS-induced ulcerative colitis mouse model. Ulcerative colitis was developed in mice by intra rectal administration of trinitrobenzene sulfonic acid. Bioactive product SEL001 (50 mg/kg b.w.) was administered orally. Myeloperoxidase activity was measured using 3,3', 5,5'-tetramethylbenzidine. The entire colon was sampled for post-mortem clinical assessment. Colonic injury was assessed through histological and histomorphometric examinations. The 454 pyrosequencing and QIIME pipeline were used for gut microbiota analysis and statistical analysis were conducted using R. mRNA extraction from colon tissue and RT-PCR approaches were employed to determine the changes in the level of specific biomarker genes associated with UC. The results depict that SEL001 significantly lowered pro-inflammatory cytokines, including CD4, TNF-α, and interleukin-6. Examination of clinical and histopathological traits revealed that SEL001 was effective and potent in reducing the inflammatory signatures of UC to a similar extent as did by the standard drug mesalamine (5-ASA). Pyro-sequencing 16S data revealed that the reduction in the major member of phylum Firmicutes, which has been previously associated with a higher risk of UC. The SEL001, an anti-inflammatory bioactive product originated from a probiotic strain L. sakei probio65 could be an alternative therapeutic agent for treatment of UC.
Collapse
Affiliation(s)
- Irfan A. Rather
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Vivek K. Bajpai
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
- Department of Energy and Materials Engineering, Dongguk University-Seoul, 30 Pildong-ro 1-gil, Seoul 04620, Republic of Korea
| | - Lew L. Ching
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
| | - Rajib Majumder
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
| | - Gyeong-Jun Nam
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
| | - Nagaraju Indugu
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA 19348, USA
| | - Prashant Singh
- Department of Food Science, College of Human Science, Florida State University, Tallahassee, FL 32306, USA
| | - Sanjay Kumar
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Nahid H. Hajrah
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Jamal S.M. Sabir
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Majid Rasool Kamli
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University (KAU), Jeddah 21589, Saudi Arabia
| | - Yong-Ha Park
- Department of Applied Microbiology and Biotechnology, School of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of Korea
| |
Collapse
|
20
|
Sevencan NO, Isler M, Kapucuoglu FN, Senol A, Kayhan B, Kiztanir S, Kockar MC. Dose-dependent effects of kefir on colitis induced by trinitrobenzene sulfonic acid in rats. Food Sci Nutr 2019; 7:3110-3118. [PMID: 31572604 PMCID: PMC6766543 DOI: 10.1002/fsn3.1174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Evidence suggests that gut microbiota dysbiosis plays a critical role in the initiation and promotion of inflammatory bowel disease (IBD). Kefir is a fermented dairy product including yeast and bacterial species. We aimed to investigate the effect of kefir on trinitrobenzene sulfonic acid (TNBS)-induced colitis in rats using two different doses. Fifty-four Wistar rats were divided into six groups. For 14 days, the normal control and colitis control groups were given tap water, kefir10 control, kefir10 colitis, and kefir30 control, and the kefir30 colitis groups were given phosphate-buffered saline containing 10% or 30% kefir, respectively, instead of tap water. Colitis was induced by intracolonically administrating TNBS in the colitis control, kefir10 colitis, and kefir30 colitis groups. On the 14th day, the rats were sacrificed. The weights and lengths of the colons were measured and macroscopically evaluated, and the distal 10 cm segments were subjected to a histopathological examination. The incidence of bloody stool and diarrhea in the kefir10 colitis group was found to be less than the colitis control and kefir30 colitis groups. The colonic weight/length ratio in the kefir10 colitis group was lower than that in the colitis control and kefir30 colitis groups. We detected that the 10% kefir treatment reduced TNBS-induced macroscopic colonic damage, while it was exacerbated by the 30% kefir treatment. No significant difference was observed between the colitis groups in terms of microscopic colonic damage scoring. These results indicate that kefir, with a careful dose selection, may be a useful agent in the treatment of IBD.
Collapse
Affiliation(s)
- Nurhayat Ozkan Sevencan
- Department of Internal Medicine, Medical FacultyThe University of KarabukKarabukTurkey
- Department of Internal Medicine, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
| | - Mehmet Isler
- Department of Gastroenterology, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
- Department of GastroenterologyDavraz Yasam HospitalIspartaTurkey
| | - Fatma Nilgun Kapucuoglu
- Department of Pathology, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
- Department of Pathology, Medical FacultyThe University of KocIstanbulTurkey
| | - Altug Senol
- Department of Gastroenterology, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
| | - Burcak Kayhan
- Department of Internal Medicine, Medical FacultyThe University of KarabukKarabukTurkey
| | - Sefa Kiztanir
- Department of Internal Medicine, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
| | - Muhammed Cem Kockar
- Department of Gastroenterology, Medical FacultyThe University of Suleyman DemirelIspartaTurkey
| |
Collapse
|
21
|
Ottman N, Reunanen J, Meijerink M, Pietilä TE, Kainulainen V, Klievink J, Huuskonen L, Aalvink S, Skurnik M, Boeren S, Satokari R, Mercenier A, Palva A, Smidt H, de Vos WM, Belzer C. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS One 2017; 12:e0173004. [PMID: 28249045 PMCID: PMC5332112 DOI: 10.1371/journal.pone.0173004] [Citation(s) in RCA: 297] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/12/2017] [Indexed: 12/24/2022] Open
Abstract
Gut barrier function is key in maintaining a balanced response between the host and its microbiome. The microbiota can modulate changes in gut barrier as well as metabolic and inflammatory responses. This highly complex system involves numerous microbiota-derived factors. The gut symbiont Akkermansia muciniphila is positively correlated with a lean phenotype, reduced body weight gain, amelioration of metabolic responses and restoration of gut barrier function by modulation of mucus layer thickness. However, the molecular mechanisms behind its metabolic and immunological regulatory properties are unexplored. Herein, we identify a highly abundant outer membrane pili-like protein of A. muciniphila MucT that is directly involved in immune regulation and enhancement of trans-epithelial resistance. The purified Amuc_1100 protein and enrichments containing all its associated proteins induced production of specific cytokines through activation of Toll-like receptor (TLR) 2 and TLR4. This mainly leads to high levels of IL-10 similar to those induced by the other beneficial immune suppressive microorganisms such as Faecalibacterium prausnitzii A2-165 and Lactobacillus plantarum WCFS1. Together these results indicate that outer membrane protein composition and particularly the newly identified highly abundant pili-like protein Amuc_1100 of A. muciniphila are involved in host immunological homeostasis at the gut mucosa, and improvement of gut barrier function.
Collapse
Affiliation(s)
- Noora Ottman
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Biosciences, University of Helsinki, Helsinki, Finland
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Justus Reunanen
- Cancer and Translational Medicine Research Unit, Biocenter Oulu and Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Marjolein Meijerink
- Host-Microbe Interactomics, Animal Sciences, Wageningen University, Wageningen, The Netherlands
- Department Risk Analysis for Products in Development, TNO, Zeist, the Netherlands
| | - Taija E. Pietilä
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Veera Kainulainen
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Judith Klievink
- Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
- Hematology Research Unit Helsinki, University of Helsinki and Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Laura Huuskonen
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Steven Aalvink
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Mikael Skurnik
- Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
- Helsinki University Central Hospital Laboratory Diagnostics, Helsinki, Finland
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University, Wageningen, The Netherlands
| | - Reetta Satokari
- Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - Annick Mercenier
- Host-Microbe Interactomics, Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - Airi Palva
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Pastrelo MM, Dias Ribeiro CC, Duarte JW, Bioago Gollücke AP, Artigiani-Neto R, Ribeiro DA, Miszputen SJ, Fujiyama Oshima CT, Ribeiro Paiotti AP. Effect of Concentrated Apple Extract on Experimental Colitis Induced by Acetic Acid. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2017; 6:38-49. [PMID: 28868268 PMCID: PMC5568191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 02/05/2017] [Indexed: 10/25/2022]
Abstract
Reactive oxygen and nitrogen species (ROS/RNS) play a crucial role in inflammatory bowel disease (IBD) exacerbating the chronic inflammatory process. Endogenous and diet antioxidants can neutralize these compounds. The apple is widely consumed, with several antioxidant activity compounds. The present study evaluated the effects of concentrated apple extract (CAE) in acetic acid induced colitis. 29 Wistar male rats were randomized into 5 groups. G1-Sham/saline solution, G2-CAE/control, G3-acetic acid/control, G4-curative- CAE treatment and G5-preventive-CAE treatment. Eight days later, the animals were euthanized and the colonic segment resected for macroscopic and histological analysis. Gene expression was evaluated for inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), catalase and copper and zinc superoxide dismutase (CuZnSOD) by quantitative real time PCR, while protein expression was assessed for iNOS, COX-2 and 8-hydroxy-20-deoxyguanosine (8-OHdG) via immunohistochemistry. The groups G3, G4 and G5 had weight loss, while G5 had weight increase at the end of the experiment. The treatment with CAE reduced the macroscopic and microscopic injury, decreased iNOS mRNA expression and increased CuZnSOD mRNA expression in animals with induced acetic acid-colitis. The findings of the present study suggest that CAE treatment exerts an antioxidant role by downregulating iNOS and upregulating CuZnSOD.
Collapse
Affiliation(s)
- Maurício Mercaldi Pastrelo
- Department of Pathology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.
| | - Carla Caroline Dias Ribeiro
- Department of Medicine, Discipline of Gastroenterology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.
| | - Joselmo Willamys Duarte
- Department of Pathology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.
| | | | - Ricardo Artigiani-Neto
- Department of Pathology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.
| | - Daniel Araki Ribeiro
- Department of Pathology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Department of Biosciences, Universidade Federal de São Paulo, Santos, Brazil.
| | - Sender Jankiel Miszputen
- Department of Medicine, Discipline of Gastroenterology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.
| | | | - Ana Paula Ribeiro Paiotti
- Department of Pathology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Department of Medicine, Discipline of Gastroenterology, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil.,Corresponding author: Laboratory of Molecular and Experimental Pathology Department of Pathology Universidade Federal de São Paulo São Paulo, Brazil
| |
Collapse
|
23
|
Shi LH, Balakrishnan K, Thiagarajah K, Mohd Ismail NI, Yin OS. Beneficial Properties of Probiotics. Trop Life Sci Res 2016. [PMID: 27688852 DOI: 10.21315/tlsr2016.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Probiotics are live microorganisms that can be found in fermented foods and cultured milk, and are widely used for the preparation of infant food. They are well-known as "health friendly bacteria", which exhibit various health beneficial properties such as prevention of bowel diseases, improving the immune system, for lactose intolerance and intestinal microbial balance, exhibiting antihypercholesterolemic and antihypertensive effects, alleviation of postmenopausal disorders, and reducing traveller's diarrhoea. Recent studies have also been focused on their uses in treating skin and oral diseases. In addition to that, modulation of the gut-brain by probiotics has been suggested as a novel therapeutic solution for anxiety and depression. Thus, this review discusses on the current probiotics-based products in Malaysia, criteria for selection of probiotics, and evidences obtained from past studies on how probiotics have been used in preventing intestinal disorders via improving the immune system, acting as an antihypercholesterolemic factor, improving oral and dermal health, and performing as anti-anxiety and anti-depressive agents.
Collapse
Affiliation(s)
- Lye Huey Shi
- Department of Agricultural and Food Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Kunasundari Balakrishnan
- Faculty of Engineering Technology, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia
| | - Kokila Thiagarajah
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Nor Ismaliza Mohd Ismail
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Ooi Shao Yin
- Department of Agricultural and Food Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
24
|
Shi LH, Balakrishnan K, Thiagarajah K, Mohd Ismail NI, Yin OS. Beneficial Properties of Probiotics. Trop Life Sci Res 2016; 27:73-90. [PMID: 27688852 DOI: 10.21315/tlsr2016.27.2.6] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Probiotics are live microorganisms that can be found in fermented foods and cultured milk, and are widely used for the preparation of infant food. They are well-known as "health friendly bacteria", which exhibit various health beneficial properties such as prevention of bowel diseases, improving the immune system, for lactose intolerance and intestinal microbial balance, exhibiting antihypercholesterolemic and antihypertensive effects, alleviation of postmenopausal disorders, and reducing traveller's diarrhoea. Recent studies have also been focused on their uses in treating skin and oral diseases. In addition to that, modulation of the gut-brain by probiotics has been suggested as a novel therapeutic solution for anxiety and depression. Thus, this review discusses on the current probiotics-based products in Malaysia, criteria for selection of probiotics, and evidences obtained from past studies on how probiotics have been used in preventing intestinal disorders via improving the immune system, acting as an antihypercholesterolemic factor, improving oral and dermal health, and performing as anti-anxiety and anti-depressive agents.
Collapse
Affiliation(s)
- Lye Huey Shi
- Department of Agricultural and Food Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Kunasundari Balakrishnan
- Faculty of Engineering Technology, Universiti Malaysia Perlis (UniMAP), 01000 Kangar, Perlis, Malaysia
| | - Kokila Thiagarajah
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Nor Ismaliza Mohd Ismail
- Department of Biological Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| | - Ooi Shao Yin
- Department of Agricultural and Food Science, Faculty of Science, Universiti Tunku Abdul Rahman, Jalan Universiti, Bandar Barat, 31900 Kampar, Perak, Malaysia
| |
Collapse
|
25
|
Javed NH, Alsahly MB, Khubchandani J. Oral Feeding of Probiotic Bifidobacterium infantis: Colonic Morphological Changes in Rat Model of TNBS-Induced Colitis. SCIENTIFICA 2016; 2016:9572596. [PMID: 27127686 PMCID: PMC4834163 DOI: 10.1155/2016/9572596] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 06/05/2023]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease of unknown etiology. It has been proposed that modifying the bacterial flora in intestine with probiotics may decrease the inflammatory process and prevent relapses in UC. We investigated the possible protective and therapeutic effects of a single strand of probiotic, Bifidobacterium infantis (BI), on colonic inflammation, in rats with regular feedings. Two groups of Lewis rats were prepared (n = 8). The first group was the control, sham-fed group (n = 4). The other group was the experimental BI-fed group (n = 4). Colitis was induced in both groups by intrarectal administration of TNBS under light anesthesia. The sham-fed colitis induced groups received a daily oral gavage feeding of 1.0 mL distilled water, whereas the B. infantis-fed group received 0.205 g of B. infantis dissolved in 1.0 mL distilled water daily. The change in body weight and food and water intake was recorded over the course of each study and analyzed. The rats were euthanized and tissues from the descending colon were harvested and analyzed microscopically and histologically. Results of our study indicated significant reduction in inflammation, mucosal damage, and preservation of goblet cells, as compared to the control animals. Modulation of gastrointestinal (GI) flora suggests a promising field in developing strategies for prevention and treatment of inflammatory bowel diseases by dietary modifications.
Collapse
Affiliation(s)
- Najma H. Javed
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Musaad B. Alsahly
- Department of Physiology and Health Science, Ball State University, Muncie, IN 47306, USA
| | - Jagdish Khubchandani
- Department of Physiology and Health Science, Ball State University, Muncie, IN 47306, USA
| |
Collapse
|
26
|
Distinct effects of Lactobacillus plantarum KL30B and Escherichia coli 3A1 on the induction and development of acute and chronic inflammation. Cent Eur J Immunol 2016; 40:420-30. [PMID: 26862305 PMCID: PMC4737739 DOI: 10.5114/ceji.2015.56963] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/04/2015] [Indexed: 12/16/2022] Open
Abstract
Objective Enteric bacteria are involved in the pathogenesis of ulcerative colitis. In experimental colitis, a breakdown of the intestinal epithelial barrier results in inflow of various gut bacteria, induction of acute inflammation and finally, progression to chronic colitis. Material and methods In the present study we compared pro-inflammatory properties of two bacterial strains isolated from human microbiome, Escherichia coli 3A1 and Lactobacillus plantarum KL30B. The study was performed using two experimental models of acute inflammation: peritonitis in mice and trinitrobenzenesulfonic acid (TNBS)-induced colitis in rats. Results Both bacterial strains induced massive neutrophil infiltration upon injection into sterile peritoneal cavity. However, peritoneal exudate cells stimulated in vitro with E. coli 3A1, produced far more nitric oxide, than those stimulated with L. plantarum KL30B. Interestingly, distinct effect on the development of TNBS-induced colitis was observed after oral administration of the tested bacteria. Lactobacillus plantarum KL30B evoked strong acute colitis. On the contrary, the administration of E. coli 3A1 resulted in a progression of colitis to chronicity. Conclusions Our results show that distinct effects of bacterial administration on the development of ongoing inflammation is strain specific and depends on the final effect of cross-talk between bacteria and cells of the innate immune system.
Collapse
|
27
|
Th17 Cells as Potential Probiotic Therapeutic Targets in Inflammatory Bowel Diseases. Int J Mol Sci 2015; 16:20841-58. [PMID: 26340622 PMCID: PMC4613231 DOI: 10.3390/ijms160920841] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/19/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are characterized by wasting and chronic intestinal inflammation triggered by various cytokine-mediated pathways. In recent years, it was shown that T helper 17 (Th17) cells are involved in the pathogenesis of IBD, which makes them an attractive therapeutic target. Th17 cells preferentially produce interleukin (IL)-17A–F as signature cytokines. The role of the interplay between host genetics and intestinal microbiota in the pathogenesis of IBD was demonstrated. Probiotics are live microorganisms that when orally ingested in adequate amounts, confer a health benefit to the host by modulating the enteric flora or by stimulating the local immune system. Several studies indicated the effectiveness of probiotics in preventing and treating IBD (ulcerative colitis, and Crohn’s disease). Furthermore, there is mounting evidence of probiotics selectively targeting the Th17 lineage in the prevention and management of inflammatory and autoimmune diseases such as IBD. This review highlights critical roles of Th17 cells in the pathogenesis of IBD and the rationale for using probiotics as a novel therapeutic approach for IBD through manipulation of Th17 cells. The potential molecular mechanisms by which probiotics modulate Th17 cells differentiation and production are also discussed.
Collapse
|
28
|
Duary RK, Batish VK, Grover S. Immunomodulatory activity of two potential probiotic strains in LPS-stimulated HT-29 cells. GENES AND NUTRITION 2014; 9:398. [PMID: 24682881 DOI: 10.1007/s12263-014-0398-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/17/2014] [Indexed: 12/22/2022]
Abstract
The relative expression of mucin, pro- and anti-inflammatory genes besides other signaling molecules in HT-29 cells by two test probiotic strains of Lactobacillus plantarum Lp9 and Lp91 and the reference strain L. plantarum 5276 was evaluated by RT-qPCR using Relative Expression Software Tool qBase-Plus under in vitro simulated gut conditions. Ten house keeping genes were evaluated by using geNorm 3.4 excel based application. The most stable genes were RPL27, ACTB and B2M which were subsequently used for calculating the normalization factor. Under pretreatment conditions (4 h probiotic treatment, followed by lipopolysaccharide challenge for 3 h), all the three strains evoked downregulation of IL-8 expression by ~100 %, while in case of TNF-α, the downregulation of the relative gene expression was at the rate of 98.2, 93.8 and 98.0 % with Lp5276, Lp9 and Lp91, respectively, under the same set of conditions. Lp91 evoked maximum downregulation of IL12p35 and IFN-γ with corresponding fold reduction in relative expression of the two genes by 96.5 and 96.7 % during pre-treatment conditions. However, IL-10 and IFN-α were significantly upregulated to the extent of 8.13 ± 0.36 and 2.62 ± 0.14 fold by Lp91 under the same conditions. Lp9 and Lp91 were also quite effective in inducing the expression of Cox-1 and Cox-2 in HT-29 cells as can be reflected from their ratios, i.e., 5.90 and 6.50 (under pretreatment conditions); 3.79 and 4.36 (under co-culture conditions). Thus, the two putative indigenous L. plantarum strains Lp9 and Lp91 demonstrated immunomodulating functions in HT-29 cells at significant levels under different experimental conditions.
Collapse
Affiliation(s)
- Raj Kumar Duary
- Department of Food Engineering and Technology, Tezpur University, Napaam, 784028, Assam, India
| | | | | |
Collapse
|
29
|
Safety evaluation of Lactobacillus paracasei subsp. paracasei LC-01, a probiotic bacterium. J Microbiol 2013; 51:633-8. [PMID: 24173643 DOI: 10.1007/s12275-013-3336-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 08/01/2013] [Indexed: 10/26/2022]
Abstract
The safety of Lactobacillus paracasei subsp. paracasei LC-01 was evaluated for its use as a potential probiotic. In our in vitro study, the antibiotic resistance and the ability to produce biogenic amine were determined. The results showed that the strain was sensitive to all tested antibiotics and did not produce biogenic amine except for tyramine. The oral toxicity of this strain was evaluated in Balb/C mice. One hundred mice were divided into 10 groups. Four groups were administered 0, 10(8), 10(9), or 10(10) CFU/mouse per day dissolved in saline solution respectively, for 28 days. Three groups were injected intraperitoneally with 10(9) CFU/mouse dissolved in saline solution, and were killed 2, 5, and 10 days after injection. The last 3 groups were injected with the vehicle as controls respectively. The results showed that oral administration of the strain had no adverse effects on mouse body weight and that there was no treatment-associated bacterial translocation. Intraperitoneal administration caused a significant translocation to liver, spleen and kidney. However, this translocation did not cause illness or death throughout the experiment. The results suggest that L. paracasei subsp. paracasei LC-01 is likely to be safe for human consumption.
Collapse
|
30
|
Aparna Sudhakaran V, Panwar H, Chauhan R, Duary RK, Rathore RK, Batish VK, Grover S. Modulation of anti-inflammatory response in lipopolysaccharide stimulated human THP-1 cell line and mouse model at gene expression level with indigenous putative probiotic lactobacilli. GENES AND NUTRITION 2013; 8:637-48. [PMID: 23728791 DOI: 10.1007/s12263-013-0347-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 05/21/2013] [Indexed: 12/17/2022]
Abstract
The anti-inflammatory potential of eight indigenous probiotic Lactobacillus isolates was evaluated in vitro in terms of modulating the expression of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in human acute monocytic leukemia (THP-1) cells under inflammatory conditions. Amongst these, Lactobacillus plantarum Lp91 was the most potent anti-inflammatory strain as it evoked a significant (P < 0.001) down-regulation of TNF-α by -1.45-fold relative to the control in THP-1 cells. However, in terms of IL-6 expression, all the strains could up-regulate its expression considerably at different levels. Hence, based on in vitro expression of TNF-α, Lp91 was selected for in vivo study in lipopolysaccharide (LPS)-induced mouse model to look at the expression of TNF-α, IL-6, monocyte chemotactic protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule (ICAM-1) and E-selectin in mouse aorta. In LPS challenged (2 h) mice group fed with Lp91 for 10 days, TNF-α, IL-6, MCP-1, VCAM-1, ICAM-1 and E-selectin expressions were significantly down-regulated by 3.10-, 10.02-, 4.22-, -3.14-, 2.28- and 5.71-fold relative to control conditions. In conclusion, Lp91 could serve as a candidate probiotic strain to explore it as a possible biotherapeutic anti-inflammatory agent against inflammatory diseases including cardiovascular disease.
Collapse
Affiliation(s)
- V Aparna Sudhakaran
- Department of Dairy Microbiology, Molecular Biology Unit, National Dairy Research Institute, Karnal, 132001, Haryana, India
| | | | | | | | | | | | | |
Collapse
|
31
|
Rizzetto L, Giovannini G, Bromley M, Bowyer P, Romani L, Cavalieri D. Strain dependent variation of immune responses to A. fumigatus: definition of pathogenic species. PLoS One 2013; 8:e56651. [PMID: 23441211 PMCID: PMC3575482 DOI: 10.1371/journal.pone.0056651] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 01/11/2013] [Indexed: 11/19/2022] Open
Abstract
For over a century microbiologists and immunologist have categorized microorganisms as pathogenic or non-pathogenic species or genera. This definition, clearly relevant at the strain and species level for most bacteria, where differences in virulence between strains of a particular species are well known, has never been probed at the strain level in fungal species. Here, we tested the immune reactivity and the pathogenic potential of a collection of strains from Aspergillus spp, a fungus that is generally considered pathogenic in immuno-compromised hosts. Our results show a wide strain-dependent variation of the immune response elicited indicating that different isolates possess diverse virulence and infectivity. Thus, the definition of markers of inflammation or pathogenicity cannot be generalized. The profound understanding of the molecular mechanisms subtending the different immune responses will result solely from the comparative study of strains with extremely diverse properties.
Collapse
Affiliation(s)
- Lisa Rizzetto
- Department of Neuroscience, Pharmacology and Child’s Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Gloria Giovannini
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Michael Bromley
- National Aspergillosis Centre and Mycology Reference Centre, University Hospital of South Manchester, University of Manchester, Manchester, United Kingdom
- School of Translational Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Paul Bowyer
- National Aspergillosis Centre and Mycology Reference Centre, University Hospital of South Manchester, University of Manchester, Manchester, United Kingdom
- School of Translational Medicine, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Luigina Romani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Duccio Cavalieri
- Department of Neuroscience, Pharmacology and Child’s Health (NEUROFARBA), University of Florence, Florence, Italy
- Innovation and Research Center, Edmund Mach Fondation, San Michele all’Adige (TN), Italy
- * E-mail:
| |
Collapse
|
32
|
Soyturk M, Saygili SM, Baskin H, Sagol O, Yilmaz O, Saygili F, Akpinar H. Effectiveness of Saccharomyces boulardii in a rat model of colitis. World J Gastroenterol 2012; 18:6452-6460. [PMID: 23197891 PMCID: PMC3508640 DOI: 10.3748/wjg.v18.i44.6452] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [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 effects of Saccharomyces boulardii (S. boulardii) in an experimental rat model of trinitrobenzene sulfonic acid (TNBS)-induced colitis.
METHODS: Thirty-two Wistar albino female rats were categorized into five groups. On the first day of the study, 50 mg TNBS was administered via a rectal catheter in order to induce colitis in all rats, except those in the control group. For 14 d, the rats were fed a standard diet, without the administration of any additional supplements to either the control or TNBS groups, in addition to 1 mg/kg per day S. boulardii to the S. boulardii group, 1 mg/kg per day methyl prednisolone (MP) to the MP group. The animals in the S. boulardii + MP group were coadministered these doses of S. boulardii and MP. During the study, weight loss, stool consistency, and the presence of obvious blood in the stool were evaluated, and the disease activity index (DAI) for colitis was recorded. The intestines were examined and colitis was macro- and microscopically scored. The serum and tissue levels of tumor necrosis factor-α (TNF-α) and nitric oxide (NO) were determined, and fungemia was evaluated in the blood samples.
RESULTS: The mean DAI scores for the MP and S. boulardii + MP groups was significantly lower than the TNBS group (3.69 ± 0.61 vs 4.46 ± 0.34, P = 0.018 and 3.77 ± 0.73 vs 4.46 ± 0.34, P = 0.025, respectively). While no significant differences between the TNBS and the S. boulardii or MP groups could be determined in terms of serum NO levels, the level of serum NO in the S. boulardii + MP group was significantly higher than in the TNBS and S. boulardii groups (8.12 ± 4.25 μmol/L vs 3.18 ± 1.19 μmol/L, P = 0.013; 8.12 ± 4.25 μmol/L vs 3.47 ± 1.66 μmol/L, P = 0.012, respectively). The tissue NO levels in the S. boulardii, MP and S. boulardii + MP groups were significantly lower than the TNBS group (16.62 ± 2.27 μmol/L vs 29.72 ± 6.10 μmol/L, P = 0.002; 14.66 ± 5.18 μmol/L vs 29.72 ± 6.10 μmol/L, P = 0.003; 11.95 ± 2.34 μmol/L vs 29.72 ± 6.10 μmol/L, P = 0.002, respectively). The tissue NO levels in the S. boulardii, MP and S. boulardii + MP groups were similar. The mean serum and tissue TNF-α levels were determined to be 12.97 ± 18.90 pg/mL and 21.75 ± 15.04 pg/mL in the control group, 18.25 ± 15.44 pg/mL and 25.27 ± 11.95 pg/mL in the TNBS group, 20.59 ± 16.15 pg/mL and 24.39 ± 13.06 pg/mL in the S. boulardii group, 9.05 ± 5.13 pg/mL and 24.46 ± 10.85 pg/mL in the MP group, and 13.95 ± 10.17 pg/mL and 24.26 ± 10.37 pg/mL in the S. boulardii + MP group. Significant differences in terms of the levels of serum and tissue TNF-α and the macroscopic and microscopic scores were not found between the groups. S. boulardii fungemia was not observed in any of the rats. However, Candida fungemia was detected in one rat (14%) in the TNBS group, two rats (28%) in the S. boulardii group, three rats (50%) in the MP group, and three rats (42%) in S. boulardii + MP group.
CONCLUSION: S. boulardii does not demonstrate considerable effects on the DAI, pathological scores, or cytokine levels but does decrease the tissue NO levels.
Collapse
|
33
|
Pophaly SD, Singh R, Pophaly SD, Kaushik JK, Tomar SK. Current status and emerging role of glutathione in food grade lactic acid bacteria. Microb Cell Fact 2012; 11:114. [PMID: 22920585 PMCID: PMC3462692 DOI: 10.1186/1475-2859-11-114] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Accepted: 08/18/2012] [Indexed: 12/15/2022] Open
Abstract
Lactic acid bacteria (LAB) have taken centre stage in perspectives of modern fermented food industry and probiotic based therapeutics. These bacteria encounter various stress conditions during industrial processing or in the gastrointestinal environment. Such conditions are overcome by complex molecular assemblies capable of synthesizing and/or metabolizing molecules that play a specific role in stress adaptation. Thiols are important class of molecules which contribute towards stress management in cell. Glutathione, a low molecular weight thiol antioxidant distributed widely in eukaryotes and Gram negative organisms, is present sporadically in Gram positive bacteria. However, new insights on its occurrence and role in the latter group are coming to light. Some LAB and closely related Gram positive organisms are proposed to possess glutathione synthesis and/or utilization machinery. Also, supplementation of glutathione in food grade LAB is gaining attention for its role in stress protection and as a nutrient and sulfur source. Owing to the immense benefits of glutathione, its release by probiotic bacteria could also find important applications in health improvement. This review presents our current understanding about the status of glutathione and its role as an exogenously added molecule in food grade LAB and closely related organisms.
Collapse
Affiliation(s)
- Sarang Dilip Pophaly
- Dairy Microbiology Division, National Dairy Research Institute, Karnal, Haryana, India, 132001
| | - Rameshwar Singh
- Dairy Microbiology Division, National Dairy Research Institute, Karnal, Haryana, India, 132001
| | | | - Jai K Kaushik
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India, 132001
| | - Sudhir Kumar Tomar
- Dairy Microbiology Division, National Dairy Research Institute, Karnal, Haryana, India, 132001
| |
Collapse
|
34
|
Wu ZW, Lu HF, Wu J, Zuo J, Chen P, Sheng JF, Zheng SS, Li LJ. Assessment of the fecal lactobacilli population in patients with hepatitis B virus-related decompensated cirrhosis and hepatitis B cirrhosis treated with liver transplant. MICROBIAL ECOLOGY 2012; 63:929-937. [PMID: 21965156 DOI: 10.1007/s00248-011-9945-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 09/13/2011] [Indexed: 05/31/2023]
Abstract
This study aims to provide an overview of the diversity of intestinal Lactobacillus among Chinese patients with hepatitis B virus (HBV)-related decompensated cirrhosis and who received liver transplant for hepatitis B cirrhosis. Fecal samples were collected from 38 healthy volunteers, 61 patients with HBV-related decompensated cirrhosis (group LC) and 74 patients who had liver transplant for hepatitis B cirrhosis (group LT). Quantitative polymerase chain reaction technology with species-specific primers was applied to investigate lactobacilli 16S rDNA in crude DNA, extracted from fecal samples. Software package Statistical Package for the Social Sciences and Palaeontological Statistics for Windows was used to analyze the data. Lactobacilli population of the two patient groups was different from the healthy control subjects, principal differences being marked decrease in the population of Lactobacillus rhamnosus (p < 0.001 for both patient groups) and reduction in the frequency of Lactobacillus fermentus (p < 0.001 for group LC and p < 0.01 for group LT). Our findings on the frequency of lactobacilli population suggested decreased diversity in groups LC and LT (compared with the healthy controls (p < 0.001 and p < 0.01, respectively)). Patients tended to have less complex fecal lactobacilli composition than the healthy controls, especially in the group LC.
Collapse
Affiliation(s)
- Zhong-Wen Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Paiotti APR, Ribeiro DA, Silva RM, Marchi P, Oshima CTF, Neto RA, Miszputen SJ, Franco M. Effect of COX-2 inhibitor lumiracoxib and the TNF-α antagonist etanercept on TNBS-induced colitis in Wistar rats. J Mol Histol 2012; 43:307-17. [PMID: 22426941 DOI: 10.1007/s10735-012-9400-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Accepted: 03/05/2012] [Indexed: 12/18/2022]
Abstract
Crohn's disease (CD) is associated with gut barrier dysfunction. Besides the baseline barrier defect, a subgroup of patients also expresses an intestinal barrier hyperresponsiveness to nonsteroidal anti-inflammatory drugs. On the other hand, the anti-tumour necrosis factor alpha (TNF-α) treatment has brought benefits to these patients. Thus, this study aimed to evaluate the effect of lumiracoxib, a selective-cyclooxygenase-2 (COX-2) inhibitor, and Etanercept (ETC), a TNF-α antagonist on the 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced experimental colitis. A total of 47 Wistar rats were randomized into seven groups, as follows: (1) Sham: sham induced-colitis; (2) TNBS: nontreated induced-colitis; (3) Lumiracoxib control; (4) Lumiracoxib-treated induced-colitis; (5) ETC control; (6) ETC-treated induced-colitis; (7) Lumiracoxib-ETC-treated induced-colitis. Rats from groups 6 and 7 presented significant improvement of macroscopic and histopathological damages in the distal colon. The gene expression of COX-2 mRNA, as well of TNF-α mRNA, decreased significantly in groups 6 and 7 compared to the TNBS nontreated and lumiracoxib-treated groups. The treatment only with lumiracoxib did not reduce the inflammation on TNBS-induced experimental colitis. ETC attenuated the damage seen in the colon and reduced the inflammation caused by TNBS. Our results suggest that down-regulation of TNF-α and COX-2 resulted in a decrease in inflammation caused by TNBS and thus provided some protection from the colonic damage caused by TNBS.
Collapse
Affiliation(s)
- Ana Paula Ribeiro Paiotti
- Department of Pathology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, UNIFESP, Sao Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Riboulet-Bisson E, Sturme MHJ, Jeffery IB, O'Donnell MM, Neville BA, Forde BM, Claesson MJ, Harris H, Gardiner GE, Casey PG, Lawlor PG, O'Toole PW, Ross RP. Effect of Lactobacillus salivarius bacteriocin Abp118 on the mouse and pig intestinal microbiota. PLoS One 2012; 7:e31113. [PMID: 22363561 PMCID: PMC3281923 DOI: 10.1371/journal.pone.0031113] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 01/02/2012] [Indexed: 12/20/2022] Open
Abstract
Lactobacilli are Gram-positive bacteria that are a subdominant element in the human gastrointestinal microbiota, and which are commonly used in the food industry. Some lactobacilli are considered probiotic, and have been associated with health benefits. However, there is very little culture-independent information on how consumed probiotic microorganisms might affect the entire intestinal microbiota. We therefore studied the impact of the administration of Lactobacillus salivarius UCC118, a microorganism well characterized for its probiotic properties, on the composition of the intestinal microbiota in two model animals. UCC118 has anti-infective activity due to production of the bacteriocin Abp118, a broad-spectrum class IIb bacteriocin, which we hypothesized could impact the microbiota. Mice and pigs were administered wild-type (WT) L. salivarius UCC118 cells, or a mutant lacking bacteriocin production. The microbiota composition was determined by pyrosequencing of 16S rRNA gene amplicons from faeces. The data show that L. salivarius UCC118 administration had no significant effect on proportions of major phyla comprising the mouse microbiota, whether the strain was producing bacteriocin or not. However, L. salivarius UCC118 WT administration led to a significant decrease in Spirochaetes levels, the third major phylum in the untreated pig microbiota. In both pigs and mice, L. salivarius UCC118 administration had an effect on Firmicutes genus members. This effect was not observed when the mutant strain was administered, and was thus associated with bacteriocin production. Surprisingly, in both models, L. salivarius UCC118 administration and production of Abp118 had an effect on Gram-negative microorganisms, even though Abp118 is normally not active in vitro against this group of microorganisms. Thus L. salivarius UCC118 administration has a significant but subtle impact on mouse and pig microbiota, by a mechanism that seems at least partially bacteriocin-dependent.
Collapse
Affiliation(s)
| | - Mark H. J. Sturme
- School of Health Sciences, Life and Health Sciences Research Institute, University of Minho, Braga, Portugal
| | - Ian B. Jeffery
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Michelle M. O'Donnell
- Department of Microbiology, University College Cork, Cork, Ireland
- Moorepark Food Research Centre, Teagasc, Fermoy, Ireland
| | - B. Anne Neville
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Brian M. Forde
- Department of Microbiology, University College Cork, Cork, Ireland
| | | | - Hugh Harris
- Department of Microbiology, University College Cork, Cork, Ireland
| | - Gillian E. Gardiner
- Department of Chemical and Life Sciences, Waterford Institute of Technology, Waterford, Ireland
| | - Patrick G. Casey
- Department of Microbiology, University College Cork, Cork, Ireland
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - Peadar G. Lawlor
- Pig Development Department, Animal & Grassland Research & Innovation Centre, Teagasc, Fermoy, Ireland
| | - Paul W. O'Toole
- Department of Microbiology, University College Cork, Cork, Ireland
- * E-mail:
| | - R. Paul Ross
- Moorepark Food Research Centre, Teagasc, Fermoy, Ireland
| |
Collapse
|
37
|
Bron PA, van Baarlen P, Kleerebezem M. Emerging molecular insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol 2011; 10:66-78. [PMID: 22101918 DOI: 10.1038/nrmicro2690] [Citation(s) in RCA: 423] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Probiotic bacteria can modulate immune responses in the host gastrointestinal tract to promote health. The genomics era has provided novel opportunities for the discovery and characterization of bacterial probiotic effector molecules that elicit specific responses in the intestinal system. Furthermore, nutrigenomic analyses of the response to probiotics have unravelled the signalling and immune response pathways which are modulated by probiotic bacteria. Together, these genomic approaches and nutrigenomic analyses have identified several bacterial factors that are involved in modulation of the immune system and the mucosal barrier, and have revealed that a molecular 'bandwidth of human health' could represent a key determinant in an individual's physiological responsiveness to probiotics. These approaches may lead to improved stratification of consumers and to subpopulation-level probiotic supplementation to maintain or improve health, or to reduce the risk of disease.
Collapse
Affiliation(s)
- Peter A Bron
- Top Institute Food and Nutrition, Nieuwe Kanaal 9A, 6709 PA Wageningen, The Netherlands
| | | | | |
Collapse
|
38
|
Abstract
The intestinal tract, known for its capability for self-renew, represents the first barrier of defence between the organism and its luminal environment. The thiol/disulfide redox systems comprising the glutathione/glutathione disulfide (GSH/GSSG), cysteine/cystine (Cys/CySS) and reduced and oxidized thioredoxin (Trx/TrxSS) redox couples play important roles in preserving tissue redox homeostasis, metabolic functions, and cellular integrity. Control of the thiol-disulfide status at the luminal surface is essential for maintaining mucus fluidity and absorption of nutrients, and protection against chemical-induced oxidant injury. Within intestinal cells, these redox couples preserve an environment that supports physiological processes and orchestrates networks of enzymatic reactions against oxidative stress. In this review, we focus on the intestinal redox and antioxidant systems, their subcellular compartmentation, redox signalling and epithelial turnover, and contribution of luminal microbiota, key aspects that are relevant to understanding redox-dependent processes in gut biology with implications for degenerative digestive disorders, such as inflammation and cancer.
Collapse
Affiliation(s)
- Magdalena L Circu
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | |
Collapse
|
39
|
Abstract
Over the past decade it has become clear that lactobacilli and other probiotic and commensal organisms can interact with mucosal immune cells or epithelial cells lining the mucosa to modulate specific functions of the mucosal immune system. The most well understood signalling mechanisms involve the innate pattern recognition receptors such as Toll-like receptors, nucleotide oligomerization domain-like receptors and C-type lectin receptors. Binding of microbe-associated molecular patterns with these receptors can activate antigen presenting cells and modulate their function through the expression of surface receptors, secreted cytokines and chemokines. In vitro the cytokine response of human peripheral blood mononuclear cells and dendritic cells to lactobacilli can be strikingly different depending on both the bacterial species and the strain. Several factors have been identified in lactobacilli that influence the immune response in vitro and in vivo including cell surface carbohydrates, enzymes modifying the structure of lipoteichoic acids and metabolites. In mice mechanistic studies point to a role for the homeostatic control of inducible T regulatory cells in the mucosal tissues as one possible immunomodulatory mechanism. Increasing evidence also suggests that induction of epithelial signalling by intestinal lactobacilli can modulate barrier functions, defensin production and regulate inflammatory signalling. Other probiotic mechanisms include modulation of the T cell effector subsets, enhancement of humoral immunity and interactions with the epithelial-associated dendritic cells and macrophages. A major challenge for the future will be to gain more knowledge about the interactions occurring between lactobacilli and the host in vivo and to understand the molecular basis of innate signalling in response to whole bacteria which trigger multiple signalling pathways.
Collapse
Affiliation(s)
- Jerry M Wells
- Host-Microbe-Interactomics, University of Wageningen, Animal Sciences Department, P.O. Box 338, 6700 AH, Wageningen, The Netherlands.
| |
Collapse
|
40
|
Paiotti APR, Miszputen SJ, Oshima CTF, Artigiani Neto R, Ribeiro DA, Franco M. Etanercept attenuates TNBS-induced experimental colitis: role of TNF-α expression. J Mol Histol 2011; 42:443-50. [PMID: 21863329 DOI: 10.1007/s10735-011-9349-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 07/29/2011] [Indexed: 01/26/2023]
Abstract
Crohn's disease (CD) is associated with gut barrier dysfunction. Tumour necrosis factor-α (TNF-α) plays an important role into the pathogenesis of several inflammatory diseases because its expression is increased in inflamed mucosa of CD patients. Anti-TNF therapy improves significantly mucosal inflammation. Thus, this study aimed to evaluate the effect of Etanercept (ETC), a tumour necrosis factor alpha (TNF-α) antagonist on the 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced experimental colitis. A total of 18 Wistar rats were randomized into four groups, as follows: (1) Sham: sham induced-colitis; (2) TNBS: non-treated induced-colitis; (3) ETC control; (4) ETC-treated induced-colitis. Rats from group 4 presented significant improvement either of macroscopic or of histopathological damage in the distal colon. The gene expression of TNF-α mRNA, decreased significantly in this group compared to the TNBS non-treated group. The treatment with etanercept attenuated the colonic damages and reduced the inflammation caused by TNBS. Taken together, our results suggest that ETC attenuates intestinal colitis induced by TNBS in Wistar rats by TNF-α downregulation.
Collapse
Affiliation(s)
- Ana Paula Ribeiro Paiotti
- Department of Pathology, Universidade Federal de São Paulo-Escola Paulista de Medicina, UNIFESP, São Paulo, SP, Brazil.
| | | | | | | | | | | |
Collapse
|
41
|
The effect of exopolysaccharide-producing probiotic strains on gut oxidative damage in experimental colitis. Dig Dis Sci 2011; 56:707-14. [PMID: 20683661 DOI: 10.1007/s10620-010-1362-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 07/15/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND Oxidative stress plays a role in disease initiation and progression in inflammatory bowel disease (IBD) and manipulation of this pathway may attenuate disease progress. In this study, the effect of exopolysaccharide (EPS)-producing probiotic bacteria on gut oxidative damage was evaluated in a rat model of experimental colitis. METHODS Colitis was induced by intracolonic administration of acetic acid. Rats were treated daily with two probiotic strains, L. delbrueckii subsp. bulgaricus B3 strain (EPS of 211 mg/l; high-EPS group) or L. delbrueckii subsp. bulgaricus A13 strain (EPS of 27 mg/l; low-EPS group), which were given directly into the stomach. Non-colitis-fed control and preventative groups were only treated with the high-EPS producing strain. Antioxidant enzyme activities (superoxide dismutase, catalase, total glutathione, reduced glutathione, glutathione disulfide) and lipid peroxidation were measured in colonic tissue samples after a treatment period of 7 days. RESULTS Significant oxidative damage was associated with a higher level of malondialdehyde (MDA) activity and reduced antioxidant enzyme activities in the colitis model group. All antioxidant enzyme activities were higher in both probiotic-treated groups compared with those of the colitis model group (P < 0.001). Lipid peroxidation was significantly ameliorated in both probiotic groups. The improvement of oxidative stress parameters was significantly more in the high-EPS group than in the low-EPS group (P < 0.001). CONCLUSIONS EPS-producing probiotic bacteria significantly attenuate oxidative stress in experimental colitis. Increased EPS production gives rise to a better probiotic function. These results suggest that EPS molecules could revaluate probiotic strains and exert their beneficial effects on the host and this may have a therapeutic potential.
Collapse
|
42
|
Spyropoulos BG, Misiakos EP, Fotiadis C, Stoidis CN. Antioxidant properties of probiotics and their protective effects in the pathogenesis of radiation-induced enteritis and colitis. Dig Dis Sci 2011; 56:285-94. [PMID: 20632107 DOI: 10.1007/s10620-010-1307-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 06/14/2010] [Indexed: 12/20/2022]
Abstract
Radiation therapy has become one of the most important treatment modalities for human malignancy, but certain immediate and delayed side-effects on the normal surrounding tissues limit the amount of effective radiation that can be administered. After exposure of the abdominal region to ionizing radiation, nearly all patients experience transient symptoms of irradiation of the bowel. Acute-phase symptoms may persist for a short time, yet long-term complications can represent significant clinical conditions with high morbidity. Data from both experimental studies and clinical trials suggest the potential benefit for probiotics in radiation-induced enteritis and colitis. On the other hand, it is well evidenced that both useful and harmful effects of therapeutic applications of ionizing radiation upon living systems are ascribed to free-radical production. Therefore, the hypothesis that probiotics reinforce antioxidant defense systems of normal mucosal cells exposed to ionizing radiation may explain to an extent their beneficial action. The aim of this review is threefold: First, to make a short brief into the natural history of radiation injury to the intestinal tract. Second, to describe the primary interaction of ionizing radiation at the cellular level and demonstrate the participation of free radicals in the mechanisms of injury and, third, to try a more profound investigation into the antioxidant abilities of probiotics and prebiotics based on the available experimental and clinical data.
Collapse
Affiliation(s)
- Basileios G Spyropoulos
- 1st Department of Propaedeutic Surgery, University of Athens School of Medicine, Hippokration Hospital, Athens, Greece.
| | | | | | | |
Collapse
|
43
|
Promising immunomodulatory effects of selected strains of dairy propionibacteria as evidenced in vitro and in vivo. Appl Environ Microbiol 2010; 76:8259-64. [PMID: 20971874 DOI: 10.1128/aem.01976-10] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Immunomodulatory properties of 10 dairy propionibacteria, analyzed on human peripheral blood mononuclear cells (PBMCs), revealed a highly strain-dependent induction of anti-inflammatory cytokine interleukin 10 (IL-10). Two selected strains of Propionibacterium freudenreichii showed a protective effect against two models of colitis in mice, suggesting a probiotic potential predicted by immune-based selection criteria for these cheese starter bacteria.
Collapse
|
44
|
Grijó NN, Borra RC, Sdepanian VL. Proinflammatory and anti-inflammatory cytokines present in the acute phase of experimental colitis treated with Saccharomyces boulardii. Dig Dis Sci 2010; 55:2498-504. [PMID: 19997974 DOI: 10.1007/s10620-009-1072-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2009] [Accepted: 11/20/2009] [Indexed: 12/31/2022]
Abstract
PURPOSE To study the proinflammatory and anti-inflammatory cytokines present in the acute phase of trinitrobenzene sulfonic acid (TNBS)-induced experimental colitis treated with Saccharomyces boulardii. METHODS Thirty male Wistar rats were divided into three groups: (1) treated group--received Saccharomyces boulardii for 14 days; (2) non-treated group--received sodium chloride solution for 14 days; (3) control group. Colitis was induced on the seventh day of the study in the treated and the non-treated groups using TNBS (10 mg) dissolved in 50% ethanol. Quantification of cytokines, including interleukin (IL)-1beta (IL-1beta), IL-6, transforming growth factor-beta (TGF-beta), IL-10 and tumor necrosis factor-alpha (TNF-alpha), in the serum and colonic tissue collected on day 14 were carried out using an enzyme-linked immunosorbent assay (ELISA). RESULTS The mean concentrations of TGF-beta in both the serum and the colonic tissue of the treated group were statistically higher than that of the control group. The mean concentration of TGF-beta in the colonic tissue of the non-treated group was also statistically higher than the control group. CONCLUSION The group treated with Saccharomyces boulardii showed increased amounts of TGF-beta, an anti-inflammatory cytokine, during the acute phase of colitis. There were no differences in the amount of TNF-alpha, IL-1beta, IL-6, and IL-10 between the treated and the non-treated or the control groups during the acute phase of experimental colitis induced by TNBS.
Collapse
Affiliation(s)
- Nathália Nahas Grijó
- Division of Pediatric Gastroenterology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | | |
Collapse
|
45
|
Martín V, Maldonado A, Fernández L, Rodríguez JM, Connor RI. Inhibition of human immunodeficiency virus type 1 by lactic acid bacteria from human breastmilk. Breastfeed Med 2010; 5:153-8. [PMID: 20491589 PMCID: PMC2936252 DOI: 10.1089/bfm.2010.0001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Human breastmilk provides a rich source of commensal lactic acid bacteria (LAB) to the infant during breastfeeding and stimulates abundant growth and colonization of these bacteria at mucosal surfaces in the infant gastrointestinal tract. While conferring critical nutritional and immunologic support to the developing newborn, breastmilk also serves as a vehicle for human immunodeficiency virus type 1 (HIV-1) transmission from mother to child during breastfeeding. Whether breastmilk LAB confer protection against mucosal exposure to HIV-1 in breastfeeding infants is unknown. STUDY DESIGN In the present study, we sought to evaluate LAB isolated from the breastmilk of healthy women for the ability to inhibit HIV-1 infection in vitro. A total of 38 strains of breastmilk bacteria were evaluated in this study. Both heat-killed bacteria and cell-free conditioned supernatants from bacterial cultures were tested for the ability to inhibit infection with HIV-1 using viral isolates with tropism for CCR5 (R5), CXCR4 (X4), or R5/X4 dual-tropism. RESULTS Significant inhibition of R5-tropic HIV-1 was demonstrated using heat-killed bacteria, most notably among breastmilk strains of Lactobacillus and Pediococcus. Selected strains of breastmilk LAB also demonstrated significant inhibition of HIV-1 infection against virus with tropism for X4 and R5/X4. CONCLUSION These results demonstrate for the first time that commensal LAB from human breastmilk inhibit HIV-1 infection in vitro and suggest a possible role for these bacteria in mucosal protection against HIV-1 in the breastfeeding infant.
Collapse
Affiliation(s)
- Virginia Martín
- Department of Nutrition, Food Science, and Technology, Complutense University of Madrid, Spain
| | | | | | | | | |
Collapse
|
46
|
Carroll IM, Threadgill DW, Threadgill DS. The gastrointestinal microbiome: a malleable, third genome of mammals. Mamm Genome 2009; 20:395-403. [PMID: 19629594 PMCID: PMC4372805 DOI: 10.1007/s00335-009-9204-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Accepted: 06/26/2009] [Indexed: 12/13/2022]
Abstract
The nonpathogenic, mutualistic bacteria of the mammalian gastrointestinal tract provide a number of benefits to the host. Recent reports have shown how the aggregate genomes of gastrointestinal bacteria provide novel benefits by functioning as the third major genome in mammals along with the nuclear and mitochondrial genomes. Consequently, efforts are underway to elucidate the complexity of the organisms comprising the unique ecosystem of the gastrointestinal tract, as well as those associated with other epidermal surfaces. The current knowledge of the gastrointestinal microbiome, its relationship to human health and disease with a particular focus on mammalian physiology, and efforts to alter its composition as a novel therapeutic approach are reviewed.
Collapse
Affiliation(s)
- Ian M. Carroll
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David W. Threadgill
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Genetics, North Carolina State University, Raleigh, NC 27695, USA
| | - Deborah S. Threadgill
- Department of Microbiology, North Carolina State University, Campus Box 7615, Raleigh, NC 27695, USA
| |
Collapse
|
47
|
Effects of Lactobacillus salivarius 433118 on intestinal inflammation, immunity status and in vitro colon function in two mouse models of inflammatory bowel disease. Dig Dis Sci 2008; 53:2495-506. [PMID: 18157694 DOI: 10.1007/s10620-007-0157-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 11/26/2007] [Indexed: 01/07/2023]
Abstract
The probiotic, Lactobacillus salivarius subsp. salivarius 433118 (UCC118), was investigated for its potential to attenuate colitis, modulate immune responses and alter intestinal barrier dysfunction in two different mouse models of inflammatory bowel disease (IBD). Following oral treatment with UCC118, faecal microbial analysis indicated that viable intact bacteria reached the colons of interleukin (IL)-10(-/-) mice and dextran sodium sulphate (DSS)-treated mice. Neither prophylactic nor therapeutic UCC118 treatment significantly prevented or attenuated inflammation in either model. In all studies, the probiotic-treated mice had comparable cytokine responses as vehicle-treated animals. Mannitol permeability was increased across colonic mucosae mounted in Ussing chambers from DSS-treated mice, but not in IL-10(-/-) mice. However, colonic mucosae from UCC118-treated mice had unchanged transepithelial electrical resistance (TEER) values and mannitol fluxes compared to controls. In two different mouse colitis models examined under a range of histological and functional criteria, the data therefore suggest that this Lactobacillus subsp. has limited potential as a prophylactic or therapeutic treatment for inflammatory bowel disease. While several studies have shown therapeutic activity for this probiotic in mouse models of IBD, our data suggest that there are inter-study variables in formulation, study design, animal models and assessment criteria that may impact on interpretation of probiotic efficacy.
Collapse
|
48
|
Prakash S, Malgorzata Urbanska A. Colon-targeted delivery of live bacterial cell biotherapeutics including microencapsulated live bacterial cells. Biologics 2008; 2:355-78. [PMID: 19707368 PMCID: PMC2721377 DOI: 10.2147/btt.s2372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There has been an ample interest in delivery of therapeutic molecules using live cells. Oral delivery has been stipulated as best way to deliver live cells to humans for therapy. Colon, in particular, is a part of gastrointestinal (GI) tract that has been proposed to be an oral targeted site. The main objective of these oral therapy procedures is to deliver live cells not only to treat diseases like colorectal cancer, inflammatory bowel disease, and other GI tract diseases like intestinal obstruction and gastritis, but also to deliver therapeutic molecules for overall therapy in various diseases such as renal failure, coronary heart disease, hypertension, and others. This review provides a comprehensive summary of recent advancement in colon targeted live bacterial cell biotherapeutics. Current status of bacterial cell therapy, principles of artificial cells and its potentials in oral delivery of live bacterial cell biotherapeutics for clinical applications as well as biotherapeutic future perspectives are also discussed in our review.
Collapse
Affiliation(s)
- Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Departments of Biomedical Engineering and Physiology, Artificial Cells and Organs Research Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Aleksandra Malgorzata Urbanska
- Biomedical Technology and Cell Therapy Research Laboratory, Departments of Biomedical Engineering and Physiology, Artificial Cells and Organs Research Center, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Damaskos D, Kolios G. Probiotics and prebiotics in inflammatory bowel disease: microflora 'on the scope'. Br J Clin Pharmacol 2008; 65:453-67. [PMID: 18279467 DOI: 10.1111/j.1365-2125.2008.03096.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The intestinal microflora is a large bacterial community that colonizes the gut, with a metabolic activity equal to an organ and various functions that affect the physiology and pathology of the host's mucosal immune system. Intestinal bacteria are useful in promotion of human health, but certain components of microflora, in genetically susceptible individuals, contribute to various pathological disorders, including inflammatory bowel disease. Clinical and experimental observations indicate an imbalance in protective and harmful microflora components in these disorders. Manipulation of gut flora to enhance its protective and beneficial role represents a promising field of new therapeutic strategies of inflammatory bowel disease. In this review, we discuss the implication of gut flora in the intestinal inflammation that justifies the role of probiotics and prebiotics in the prevention and treatment of inflammatory bowel disease and we address the evidence for therapeutic benefits from their use in experimental models of colitis and clinical trials.
Collapse
Affiliation(s)
- Dimitrios Damaskos
- Second Department of Surgery, General Hospital of Nikea, Piraeus, Greece [corrected]
| | | |
Collapse
|
50
|
Rochat T, Bermúdez-Humarán L, Gratadoux JJ, Fourage C, Hoebler C, Corthier G, Langella P. Anti-inflammatory effects of Lactobacillus casei BL23 producing or not a manganese-dependant catalase on DSS-induced colitis in mice. Microb Cell Fact 2007; 6:22. [PMID: 17659075 PMCID: PMC1949835 DOI: 10.1186/1475-2859-6-22] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Accepted: 07/20/2007] [Indexed: 01/22/2023] Open
Abstract
Background Human immune cells generate large amounts of reactive oxygen species (ROS) throughout the respiratory burst that occurs during inflammation. In inflammatory bowel diseases, a sustained and abnormal activation of the immune system results in oxidative stress in the digestive tract and in a loss of intestinal homeostasis. We previously showed that the heterologous production of the Lactobacillus plantarum ATCC14431 manganese-dependant catalase (MnKat) in Lb. casei BL23 successfully enhances its survival when exposed to oxidative stress. In this study, we evaluated the preventive effects of this antioxidative Lb. casei strain in a murine model of dextran sodium sulfate (DSS)-induced moderate colitis. Results Either Lb. casei BL23 MnKat- or MnKat+ was administered daily to mice treated with DSS for 10 days. In contrast to control mice treated with PBS for which DSS induced bleeding diarrhea and mucosal lesions, mice treated with both Lb. casei strains presented a significant (p < 0.05) reduction of caecal and colonic inflammatory scores. Conclusion No contribution of MnKat to the protective effect from epithelial damage has been observed in the tested conditions. In contrast, these results confirm the high interest of Lb. casei as an anti-inflammatory probiotic strain.
Collapse
Affiliation(s)
- Tatiana Rochat
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| | - Luis Bermúdez-Humarán
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| | - Jean-Jacques Gratadoux
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| | - Christel Fourage
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| | - Christine Hoebler
- Physiologie Intestinale, Croissance et Nutrition Humaine, UMR INRA/Université de Nantes, Rue de la Géraudière – BP 71627, 44316 Nantes cedex 3, France
| | - Gérard Corthier
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| | - Philippe Langella
- Unité d'Ecologie et Physiologie du Système Digestif, Centre de Recherche INRA, Domaine de Vilvert, 78352 Jouy-en-Josas cedex, France
| |
Collapse
|