51
|
Camelo-Silva C, Mota E Souza B, Vicente R, Arend GD, Sanches MAR, Barreto PLM, Ambrosi A, Verruck S, Di Luccio M. Polyfunctional sugar-free white chocolate fortified with Lacticaseibacillus rhamnosus GG co-encapsulated with beet residue extract (Beta vulgaris L.). Food Res Int 2024; 179:114016. [PMID: 38342537 DOI: 10.1016/j.foodres.2024.114016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/28/2023] [Accepted: 01/11/2024] [Indexed: 02/13/2024]
Abstract
Chocolate is a worldwide consumed food. This study investigated the fortification of sugar-free white chocolate with Lacticaseibacillus rhamnosus GG microcapsule co-encapsulated with beet residue extract. The chocolates were evaluated for moisture, water activity, texture, color properties, melting, physicochemical, and probiotic stability during storage. Furthermore, the survival of L. rhamnosus GG and the bioaccessibility of phenolic compounds were investigated under in vitro simulated gastrointestinal conditions. Regarding the characterization of probiotic microcapsules, the encapsulation efficiency of L. rhamnosus GG was > 89 % while the encapsulation efficiency of phenolic compounds was > 62 %. Chocolates containing probiotic microcapsules were less hard and resistant to breakage. All chocolates had a similar melting behavior (endothermic peaks between 32.80 and 34.40 °C). After 120 days of storage at 4 °C, probiotic populations > 6.77 log CFU/g were detected in chocolate samples. This result demonstrates the potential of this matrix to carry L. rhamnosus GG cells. Regarding the resistance of probiotic strains during gastric simulation, the co-encapsulation of L. rhamnosus GG with beet extract contributed to high counts during gastrointestinal transit, reaching the colon (48 h) with viable cell counts equal to 11.80 log CFU/g. Finally, one of our main findings was that probiotics used phenolic compounds as a substrate source, which may be an observed prebiotic effect.
Collapse
Affiliation(s)
- Callebe Camelo-Silva
- Laboratory of Membrane Processes, Department of Chemical and Food Engineering, Federal University of Santa Catarina, 88040-970 Florianópolis, SC, Brazil.
| | - Bianca Mota E Souza
- Department of Food Science and Technology, Agricultural Sciences Center, Federal University of Santa Catarina, 88034-001 Florianópolis, SC, Brazil
| | - Renata Vicente
- Laboratory of Membrane Processes, Department of Chemical and Food Engineering, Federal University of Santa Catarina, 88040-970 Florianópolis, SC, Brazil
| | - Giordana Demaman Arend
- Laboratory of Membrane Processes, Department of Chemical and Food Engineering, Federal University of Santa Catarina, 88040-970 Florianópolis, SC, Brazil
| | - Marcio Augusto Ribeiro Sanches
- Department of Food Engineering and Technology, State University of São Paulo, 15054-000 São José do Rio Preto, SP, Brazil
| | - Pedro Luiz Manique Barreto
- Department of Food Science and Technology, Agricultural Sciences Center, Federal University of Santa Catarina, 88034-001 Florianópolis, SC, Brazil
| | - Alan Ambrosi
- Laboratory of Membrane Processes, Department of Chemical and Food Engineering, Federal University of Santa Catarina, 88040-970 Florianópolis, SC, Brazil.
| | - Silvani Verruck
- Department of Food Science and Technology, Agricultural Sciences Center, Federal University of Santa Catarina, 88034-001 Florianópolis, SC, Brazil.
| | - Marco Di Luccio
- Laboratory of Membrane Processes, Department of Chemical and Food Engineering, Federal University of Santa Catarina, 88040-970 Florianópolis, SC, Brazil.
| |
Collapse
|
52
|
Ren D, Ding M, Su J, Ye J, He X, Zhang Y, Shang X. Stachyose in combination with L. rhamnosus GG ameliorates acute hypobaric hypoxia-induced intestinal barrier dysfunction through alleviating inflammatory response and oxidative stress. Free Radic Biol Med 2024; 212:505-519. [PMID: 38211833 DOI: 10.1016/j.freeradbiomed.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
High altitude is closely related to intestinal mucosal damage and intestinal microbiota imbalance, and there is currently no effective prevention and treatment measures. In this study, the effects of stachyose (STA), L. rhamnosus GG (LGG) and their combination on inflammatory response, oxidatve stress and intestinal barrier function in mice exposed to acute hypobaric hypoxia were investigated. Our results indicated the combination of STA and LGG could more effectively regulate intestinal microbiota disorders caused by hypobaric hypoxia than STA or LGG alone. When mice were administered with STA + LGG, the content of short chain fatty acids (SCFAs) especially butyric acid significantly increased, which helped intestinal cells to form tight connections, improve the level of anti-inflammatory cytokine (TGF-β) and antioxidant enzymes (SOD, CAT, GSH-Px), and decrease the expression of pro-inlammatory cytokines and hypoxia-inducing factors (IFN-γ, IL-1β, IL-6, TNF-α and HIF-1α), thereby enhance the strong intestinal barrier function. Furthermore, the synbiotics significantly reduced the ratio of Firmicutes to Bacteroidetes, while significantly increased the relative abundance of Rikenella, Bacteroides, Odoribacter, Ruminiclostridium_5 and Gordonibacter, which were correlated with production of SCFAs and anti-inflammatory role. Correlation analysis showed that the protective effect of synbiotics on intestinal barrier function was associated with its anti-inflammatory activity and antioxidant capacity. It provided a strong foundation for further research on the role of STA and LGG in maintaining normal intestinal function at high altitude. Our study has identified and demonstrated a new synbiotic that may be one of the ideal intervention measures for preventing and treating intestinal dysfunction at high altitude.
Collapse
Affiliation(s)
- Dingxin Ren
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Mengying Ding
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Junqing Su
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Jianzhou Ye
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Xiaoqin He
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China
| | - Yafeng Zhang
- No. 889, Xi'an Institute for Food and Drug, Cangtai West Road, Chang'an District, Xi'an, Shaanxi, 710700, PR China
| | - Xiaoya Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, PR China.
| |
Collapse
|
53
|
Hu R, Yang T, Ai Q, Shi Y, Ji Y, Sun Q, Tong B, Chen J, Wang Z. Autoinducer-2 promotes the colonization of Lactobacillus rhamnosus GG to improve the intestinal barrier function in a neonatal mouse model of antibiotic-induced intestinal dysbiosis. J Transl Med 2024; 22:177. [PMID: 38369503 PMCID: PMC10874557 DOI: 10.1186/s12967-024-04991-5] [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: 12/05/2023] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Human health is seriously threatened by antibiotic-induced intestinal disorders. Herein, we aimed to determine the effects of Autoinducer-2 (AI-2) combined with Lactobacillus rhamnosus GG (LGG) on the intestinal barrier function of antibiotic-induced intestinal dysbiosis neonatal mice. METHODS An antibiotic-induced intestinal dysbiosis neonatal mouse model was created using antibiotic cocktails, and the model mice were randomized into the control, AI-2, LGG, and LGG + AI-2 groups. Intestinal short-chain fatty acids and AI-2 concentrations were detected by mass spectrometry and chemiluminescence, respectively. The community composition of the gut microbiota was analyzed using 16S rDNA sequencing, and biofilm thickness and bacterial adhesion in the colon were assessed using scanning electron microscopy. Transcriptome RNA sequencing of intestinal tissues was performed, and the mRNA and protein levels of HCAR2 (hydroxycarboxylic acid receptor 2), claudin3, and claudin4 in intestinal tissues were determined using quantitative real-time reverse transcription PCR and western blotting. The levels of inflammatory factors in intestinal tissues were evaluated using enzyme-linked immunosorbent assays (ELISAs). D-ribose, an inhibitor of AI-2, was used to treat Caco-2 cells in vitro. RESULTS Compared with the control, AI-2, and LGG groups, the LGG + AI-2 group showed increased levels of intestinal AI-2 and proportions of Firmicutes and Lacticaseibacillus, but a reduced fraction of Proteobacteria. Specifically, the LGG + AI-2 group had considerably more biofilms and LGG on the colon surface than those of other three groups. Meanwhile, the combination of AI-2 and LGG markedly increased the concentration of butyric acid and promoted Hcar2, claudin3 and claudin4 expression levels compared with supplementation with LGG or AI-2 alone. The ELISAs revealed a significantly higher tumor necrosis factor alpha (TNF-α) level in the control group than in the LGG and LGG + AI-2 groups, whereas the interleukin 10 (IL-10) level was significantly higher in the LGG + AI-2 group than in the other three groups. In vitro, D-ribose treatment dramatically suppressed the increased levels of Hcar2, claudin3, and claudin4 in Caco-2 cells induced by AI-2 + LGG. CONCLUSIONS AI-2 promotes the colonization of LGG and biofilm formation to improve intestinal barrier function in an antibiotic-induced intestinal dysbiosis neonatal mouse model.
Collapse
Affiliation(s)
- Riqiang Hu
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ting Yang
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Qing Ai
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yanchun Ji
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Sun
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Tong
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Jie Chen
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
| | - Zhengli Wang
- Children Nutrition Research Center, Chongqing Key Laboratory of Child Neurodevelopmental and Cognitive Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Chongqing, China.
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Jiangxi Hospital Affiliated Children's Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
54
|
Lin M, Yanjun C. Research progress on the mechanism of probiotics regulating cow milk allergy in early childhood and its application in hypoallergenic infant formula. Front Nutr 2024; 11:1254979. [PMID: 38419849 PMCID: PMC10900986 DOI: 10.3389/fnut.2024.1254979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 01/22/2024] [Indexed: 03/02/2024] Open
Abstract
Some infants and young children suffer from cow's milk allergy (CMA), and have always mainly used hypoallergenic infant formula as a substitute for breast milk, but some of these formulas can still cause allergic reactions. In recent years, it has been found that probiotic nutritional interventions can regulate CMA in children. Scientific and reasonable application of probiotics to hypoallergenic infant formula is the key research direction in the future. This paper discusses the mechanism and clinical symptoms of CMA in children. This review critically ex- amines the issue of how probiotics use intestinal flora as the main vector to combine with the immune system to exert physiological functions to intervene CMA in children, with a particular focus on four mechanisms: promoting the early establishment of intestinal microecological balance, regulating the body's immunity and alleviating allergic response, enhancing the intestinal mucosal barrier function, and destroying allergen epitopes. Additionally, it overviews the development process of hypoallergenic infant formula and the research progress of probiotics in hypoallergenic infant formula. The article also offers suggestions and outlines potential future research directions and ideas in this field.
Collapse
Affiliation(s)
| | - Cong Yanjun
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, College of Food and Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
55
|
Mantel M, Durand T, Bessard A, Pernet S, Beaudeau J, Guimaraes-Laguna J, Maillard MB, Guédon E, Neunlist M, Le Loir Y, Jan G, Rolli-Derkinderen M. Propionibacterium freudenreichii CIRM-BIA 129 mitigates colitis through S layer protein B-dependent epithelial strengthening. Am J Physiol Gastrointest Liver Physiol 2024; 326:G163-G175. [PMID: 37988603 DOI: 10.1152/ajpgi.00198.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 11/23/2023]
Abstract
The growing incidence of human diseases involving inflammation and increased gut permeability makes the quest for protective functional foods more crucial than ever. Propionibacterium freudenreichii (P. freudenreichii) is a beneficial bacterium used in the dairy and probiotic industries. Selected strains exert anti-inflammatory effects, and the present work addresses whether the P. freudenreichii CIRM-BIA129, consumed daily in a preventive way, could protect mice from acute colitis induced by dextran sodium sulfate (DSS), and more precisely, whether it could protect from intestinal epithelial breakdown induced by inflammation. P. freudenreichii CIRM-BIA129 mitigated colitis severity and inhibited DSS-induced permeability. It limited crypt length reduction and promoted the expression of zonula occludens-1 (ZO-1), without reducing interleukin-1β mRNA (il-1β) expression. In vitro, P. freudenreichii CIRM-BIA129 prevented the disruption of a Caco-2 monolayer induced by proinflammatory cytokines. It increased transepithelial electrical resistance (TEER) and inhibited permeability induced by inflammation, along with an increased ZO-1 expression. Extracellular vesicles (EVs) from P. freudenreichii CIRM-BIA129, carrying the surface layer protein (SlpB), reproduced the protective effect of P. freudenreichii CIRM-BIA129. A mutant strain deleted for slpB (ΔslpB), or EVs from this mutant strain, had lost their protective effects and worsened both DSS-induced colitis and inflammation in vivo. These results shown that P. freudenreichii CIRM-BIA129 daily consumption has the potential to greatly alleviate colitis symptoms and, particularly, to counter intestinal epithelial permeability induced by inflammation by restoring ZO-1 expression through mechanisms involving S-layer protein B. They open new avenues for the use of probiotic dairy propionibacteria and/or postbiotic fractions thereof, in the context of gut permeability.NEW & NOTEWORTHY Propionibacterium freudenreichii reduces dextran sodium sulfate (DSS)-induced intestinal permeability in vivo. P. freudenreichii does not inhibit inflammation but damages linked to inflammation. P. freudenreichii inhibits intestinal epithelial breakdown through S-layer protein B. The protective effects of P. freudenreichii depend on S-layer protein B. Extracellular vesicles from P. freudenreichii CB 129 mimic the protective effect of the probiotic.
Collapse
Affiliation(s)
- Marine Mantel
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Tony Durand
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Anne Bessard
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Ségolène Pernet
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Julie Beaudeau
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
- Centres de Recherche en Nutrition Humaine-Ouest, Nantes, France
| | - Juliana Guimaraes-Laguna
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Marie-Bernadette Maillard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Eric Guédon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Michel Neunlist
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| | - Yves Le Loir
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Gwénaël Jan
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Unité Mixte de Recherche, L'Institut Agro, Rennes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, IMAD, Institut National de la Santé et de la Recherche Médicale, Nantes Université, Nantes, France
| |
Collapse
|
56
|
Zhang K, Peng P, Huang J, Chen M, Liu F, Zhu C, Lu Q, Wang M, Lin C. Integrating plasma metabolomics and gut microbiome to reveal the mechanisms of Huangqi Guizhi Wuwu Decoction intervene diabetic peripheral neuropathy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117301. [PMID: 37820997 DOI: 10.1016/j.jep.2023.117301] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/01/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huangqi Guizhi Wuwu Decoction (HGWD) is a classic traditional Chinese herbal formula from "Synopsis of Golden Chamber," which is used to treat blood stagnation and has been used for alleviating diabetic peripheral neuropathy (DPN) in the clinic. However, the mechanisms of HGWD intervention DPN are still to be discovered. AIM OF THE STUDY This study aims to explore the mechanism of HGWD intervention DPN by integrating plasma metabolomics and gut microbiome. MATERIALS AND METHODS BKS Cg-m+/+Leprdb/J (db/db) mice with DPN were at 16 weeks of age. The indices of DPN phenotypes in db/db mice, pathomorphology of the sciatic nerve, intraepithelial nerve fibers (IENF) of the foot pad, levels of blood lipids and oxidative stress, and inflammatory reaction were used to appraise the HGWD efficacy. Finally, the pharmacological mechanisms of HGWD intervening DPN were explored by metabolomics and 16S rRNA gene sequencing. RESULTS HGWD reversed DPN phenotypes in db/db mice, improved peripheral nerve structure, ameliorated the level of blood lipids and nerve growth factor in plasma, enhanced antioxidant capacity, and alleviated inflammatory responses. Plasma metabolomics disclosed that HGWD remarkably regulated the unusual levels of thirty-seven metabolites involved in sphingolipid metabolism, biosynthesis of unsaturated fatty acids, arachidonic acid metabolism, and amino acid biosynthesis pathways. The gut microbiome showed that nine bacteria were highly correlated with the efficacy of HGWD in DPN. Integrating analysis of microbiome and metabolomics demonstrated that the interaction of four bacteria with four metabolic pathways might be the significant mechanism of HGWD intervention in DPN. CONCLUSIONS The mediation of gut microbiota and plasma metabolism may be the potential mechanism of HGWD ameliorating DPN in db/db mice. The interaction of Lactobacillus, Alloprevotella, Bacteroides, and Desulfovibio with four metabolic pathways might be the critical mechanism for HGWD treating DPN.
Collapse
Affiliation(s)
- Kaihui Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Peng Peng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Jinhao Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Ming Chen
- Guangzhou BaiYunShan PanGaoShou Pharmaceutical Company Limited, Guangzhou, 511400, PR China
| | - Fangle Liu
- The First Affiliated Hospital, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510405, PR China
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Qifu Lu
- Guangzhou BaiYunShan PanGaoShou Pharmaceutical Company Limited, Guangzhou, 511400, PR China.
| | - Meiqi Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
57
|
Sosnowski K, Przybyłkowski A. Ethanol-induced changes to the gut microbiome compromise the intestinal homeostasis: a review. Gut Microbes 2024; 16:2393272. [PMID: 39224006 PMCID: PMC11376419 DOI: 10.1080/19490976.2024.2393272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The intestine is the largest organ in terms of surface area in the human body. It is responsible not only for absorbing nutrients but also for protection against the external world. The gut microbiota is essential in maintaining a properly functioning intestinal barrier, primarily through producing its metabolites: short-chain fatty acids, bile acids, and tryptophan derivatives. Ethanol overconsumption poses a significant threat to intestinal health. Not only does it damage the intestinal epithelium, but, maybe foremostly, it changes the gut microbiome. Those ethanol-driven changes shift its metabolome, depriving the host of the protective effect the physiological gut microbiota has. This literature review discusses the impact of ethanol consumption on the gut, the gut microbiota, and its metabolome, providing a comprehensive overview of the mechanisms through which ethanol disrupts intestinal homeostasis and discussing potential avenues for new therapeutic intervention.
Collapse
Affiliation(s)
- Konrad Sosnowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
58
|
Gan W, Xiang Y, Wei B, Liu S, Liu F. The inflammatory microenvironment of nasal polyps in patients with chronic rhinosinusitis and the relationship of this microenvironment with the nasal microbiome. Asian J Surg 2024; 47:124-133. [PMID: 37659949 DOI: 10.1016/j.asjsur.2023.08.096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/19/2023] [Accepted: 08/21/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND We investigated the characteristics of the microbial community of the nasal sinuses in patients with chronic rhinosinusitis with nasal polyps (CRSwNP) and identified the correlations of the nasal microbiome with the inflammatory microenvironment of the nasal cavity. METHODOLOGY We collected matched nasal secretion and polyp tissue samples from 77 CRSwNP patients. Then, we extracted microbial DNA from cotton swabs, used high-throughput sequencing technology based on 16S ribosomal RNA (rRNA) to detect the bacterial community composition, and detected cytokines such as interleukin (IL)-5, IL-8, IL-17a, IL-17e, IL-18, IL-27 and interferon (INF)-gamma in the polyp tissue samples using Luminex. Eosinophils and neutrophils in the peripheral blood and polyp tissue were counted, and the relationships between inflammatory factors or inflammatory cell counts and nasal microbial diversity were analyzed. RESULTS Among the inflammatory factors evaluated, IL-5 had a positive rate of 32.47%, IFN-γ had a positive rate of 84.42%, IL-17A and IL-17E had positive rates of 75.32%, IL-18 had a positive rate of 94.81%, IL-27 had a positive rate of 68.83%, and IL-8 had a positive rate of 100%. IL-17a and IL-27 were negatively correlated with both Enterobacter and Anaerococcus, IL-8 was negatively correlated with both Enterobacter and Staphylococcus, IL-18 was positively correlated with Candidatus Arthromitus and negatively correlated with Haemophilus, and IL-27 was positively correlated with Faecalibaculum. Lactobacillus and Enterococcus were positively correlated with the degree of neutrophil infiltration in nasal polyp tissue. CONCLUSIONS In Southwest China, inflammation of the nasal polyps exhibits a variety of patterns. Enterobacteria and anaerobic bacteria may be correlated with the inflammatory pattern of nasal polyps. The neutrophil-mediated inflammatory response plays an important role in patients with CRSwNP in Southwest China.
Collapse
Affiliation(s)
- Weigang Gan
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yu Xiang
- Morphometric Research Laboratory, North Sichuan Medical College, Nanchong, Sichuan, PR China
| | - Bo Wei
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Shixi Liu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Feng Liu
- Department of Otolaryngology-Head & Neck Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
59
|
Park C, Ji SY, Hwangbo H, Shin SY, Kim MY, Lee K, Kim DH, Cho BR, Lee H, Choi YH, You HJ. Enhancement of Immune Functions by Limosilactobacillus reuteri KBL346: In Vitro and In Vivo Studies. Int J Mol Sci 2023; 25:141. [PMID: 38203313 PMCID: PMC10779160 DOI: 10.3390/ijms25010141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/30/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Lactobacilli have been widely used as probiotics because of their benefits for intestinal health and physiological functions. Among a variety of Lactobacillus genera, Limosilactobacillus reuteri has been studied for its ability to exert anti-inflammatory functions and its role in controlling metabolic disorders, as well as the production of the antimicrobial compound reuterin. However, the effects and mechanisms of L. reuteri on enhancing immune responses in the immunosuppressed states have been relatively understudied. In this study, we isolated an immunomodulatory strain, namely, L. reuteri KBL346 (KBL346), from a fecal sample of a 3-month-old infant in Korea. We evaluated the immunostimulatory activity and hematopoietic function of KBL346 in macrophages and cyclophosphamide (CPA)-induced immunosuppressed mice. KBL346 increased the phagocytic activity against Candida albicans MYA-4788 in macrophages, and as biomarkers for this, increased secretions of nitric oxide (NO) and prostaglandin E2 (PGE2) were confirmed. Also, the secretions of innate cytokines (TNF-α, IL-1β, and IL-6) were increased. In CPA-induced immunosuppressed mice, KBL346 at a dosage of 1010 CFU/kg protected against spleen injury and suppressed levels of immune-associated parameters, including NK cell activity, T and B lymphocyte proliferation, CD4+ and CD8+ T cell abundance, cytokines, and immunoglobulins in vivo. The effects were comparable or superior to those in the Korean red ginseng positive control group. Furthermore, the safety assessment of KBL346 as a probiotic was conducted by evaluating its antibiotic resistance, hemolytic activity, cytotoxicity, and metabolic characteristics. This study demonstrated the efficacy and safety of KBL346, which could potentially be used as a supplement to enhance the immune system.
Collapse
Affiliation(s)
- Chanseop Park
- KoBioLabs Inc., Seoul 08826, Republic of Korea (K.L.); (B.-R.C.)
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea (D.H.K.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea (D.H.K.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Seung-yeon Shin
- KoBioLabs Inc., Seoul 08826, Republic of Korea (K.L.); (B.-R.C.)
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea (D.H.K.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Kiuk Lee
- KoBioLabs Inc., Seoul 08826, Republic of Korea (K.L.); (B.-R.C.)
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea (D.H.K.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Bo-Ram Cho
- KoBioLabs Inc., Seoul 08826, Republic of Korea (K.L.); (B.-R.C.)
| | - Hyesook Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea;
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47227, Republic of Korea (D.H.K.)
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hyun Ju You
- Department of Food and Nutrition, Research Institute of Human Ecology, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
60
|
Savitskaya I, Zhantlessova S, Kistaubayeva A, Ignatova L, Shokatayeva D, Sinyavskiy Y, Kushugulova A, Digel I. Prebiotic Cellulose-Pullulan Matrix as a "Vehicle" for Probiotic Biofilm Delivery to the Host Large Intestine. Polymers (Basel) 2023; 16:30. [PMID: 38201695 PMCID: PMC10780842 DOI: 10.3390/polym16010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 01/12/2024] Open
Abstract
This study describes the development of a new combined polysaccharide-matrix-based technology for the immobilization of Lactobacillus rhamnosus GG (LGG) bacteria in biofilm form. The new composition allows for delivering the bacteria to the digestive tract in a manner that improves their robustness compared with planktonic cells and released biofilm cells. Granules consisting of a polysaccharide matrix with probiotic biofilms (PMPB) with high cell density (>9 log CFU/g) were obtained by immobilization in the optimized nutrient medium. Successful probiotic loading was confirmed by fluorescence microscopy and scanning electron microscopy. The developed prebiotic polysaccharide matrix significantly enhanced LGG viability under acidic (pH 2.0) and bile salt (0.3%) stress conditions. Enzymatic extract of feces, mimicking colon fluid in terms of cellulase activity, was used to evaluate the intestinal release of probiotics. PMPB granules showed the ability to gradually release a large number of viable LGG cells in the model colon fluid. In vivo, the oral administration of PMPB granules in rats resulted in the successful release of probiotics in the colon environment. The biofilm-forming incubation method of immobilization on a complex polysaccharide matrix tested in this study has shown high efficacy and promising potential for the development of innovative biotechnologies.
Collapse
Affiliation(s)
- Irina Savitskaya
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (I.S.); (L.I.); (D.S.)
| | - Sirina Zhantlessova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (I.S.); (L.I.); (D.S.)
| | - Aida Kistaubayeva
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (I.S.); (L.I.); (D.S.)
| | - Ludmila Ignatova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (I.S.); (L.I.); (D.S.)
| | - Dina Shokatayeva
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (I.S.); (L.I.); (D.S.)
| | | | - Almagul Kushugulova
- Laboratory of Human Microbiome and Longevity, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan;
| | - Ilya Digel
- Institute for Bioengineering, Aachen University of Applied Sciences, Heinrich-Mußmann-Straße 1, D-52428 Jülich, Germany;
| |
Collapse
|
61
|
Liang M, Wang M, Zhou M, Nie S, Xu Y, Yang X, Yuan E, Ren J. Effect of walnut peptide‐ZnO nanocomposites on the colon adhesion behavior of Lactobacillus rhamnosus LRa05. FOOD FRONTIERS 2023; 4:1946-1957. [DOI: 10.1002/fft2.253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025] Open
Abstract
AbstractWhen the nanoparticles (NPs) in food contact materials are exposed, they may be ingested with the food matrix, resulting in unknown impacts. Here, the biological response of the nanocomposites of nano zinc oxide (nZnO) and walnut protein‐derived peptides (i.e., PW5, WN5, AE6, and WE7) on the Lactobacillus rhamnosus LRa05 growth and adhesion was studied. In an in vitro mouse intestinal adhesion model, we first spotted that the probiotics LRa05 primarily adhered to and colonized the colonic segment. nZnO effectively inhibited the growth and adhesion properties of LRa05 at high concentrations (≥ 1000 μg/mL). Fortunately, when compared to the individual nZnO, the nZnO‐walnut‐derived peptides nanocomposites significantly increased the growth of LRa05. It was found that the alterations in the adhesion ability of LRa05 after treatment with various substances (nZnO and nanocomposites of nZnO‐walnut peptides) were related to the auto‐aggregating property on the LRa05 surface. These results shed light on the effect of food matrices on the safety of nanomaterials in food, and they may have far‐reaching implications for the use of nanomaterials in the food industry.
Collapse
Affiliation(s)
- Ming Liang
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Min Wang
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Miao Zhou
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Shiying Nie
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Yongzhao Xu
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Xinquan Yang
- Kashi Guanghua Modern Agriculture Co. Kashi China
| | - Erdong Yuan
- School of Food Science and Engineering South China University of Technology Guangzhou China
| | - Jiaoyan Ren
- School of Food Science and Engineering South China University of Technology Guangzhou China
| |
Collapse
|
62
|
Lu S, Xu J, Zhao Z, Guo Y, Zhang H, Jurutka PW, Huang D, Cao C, Cheng S. Dietary Lactobacillus rhamnosus GG extracellular vesicles enhance antiprogrammed cell death 1 (anti-PD-1) immunotherapy efficacy against colorectal cancer. Food Funct 2023; 14:10314-10328. [PMID: 37916395 DOI: 10.1039/d3fo02018e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
There is a need to explore combination therapy to improve the efficacy of immunotherapy for colorectal cancer through food probiotics. In this study, extracellular vesicles (EV) derived from Lactobacillus rhamnosus GG (LGG-EV) were successfully isolated. Adjusting the culture temperature to 30 °C led to an elevated LGG-EV yield, and the addition of penicillin resulted in a decrease in particle size. In addition, LGG-EV have better gastrointestinal tract stability in a Ca2+ environment in vivo and in vitro. Oral administration of LGG-EV synergistically improved anti-PD-1 immunotherapy efficacy against colorectal cancer. Mechanistically, LGG-EV modulated intestinal immunity by increasing the CD8+ T/CD4+ T cell ratio in mesenteric lymph nodes and enhancing the ratio of MHC II+ DC cells, CD4+ T cells, and CD8+ T cells in tumor tissues. Meanwhile, the diversity of the gut microbiota and the abundance of beneficial bacteria, such as Lactobacillus, increased in the combined-treatment mice. In addition, there were significant changes in the levels of serum metabolites associated with the microbiota and anti-tumor effects, including uridine, which was elevated by the combination of anti-PD-1 and LGG-EV treatment. Our findings provide theoretical and mechanistic insights into the development of LGG-EV as postbiotics in combination with immune checkpoint inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Shun Lu
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Jing Xu
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Zihao Zhao
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Yuheng Guo
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Hanwen Zhang
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Peter W Jurutka
- School of Mathematical and Natural Sciences, Arizona State University, AZ 85306, USA
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Chongjiang Cao
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| | - Shujie Cheng
- Department of Food Nutrition and Safety, School of Engineering, China Pharmaceutical University, Nanjing 211198, P. R. China.
| |
Collapse
|
63
|
He Z, Wang T, Zhang S, Shi K, Wang F, Li Y, Lin C, Chen J. Evaluation of cholesterol transformation abilities and probiotic properties of Bacteroides dorei YGMCC0564. Front Microbiol 2023; 14:1279996. [PMID: 38029107 PMCID: PMC10666794 DOI: 10.3389/fmicb.2023.1279996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Hypercholesterolemia, a risk factor for cardiovascular disease (CVD), often requires therapeutic agents with varying degrees of side effects. This has created a need for safe and natural alternatives such as medications or functional foods that can improve lipid metabolism and reduce cholesterol levels. In recent years, Next-generation probiotics (NGPs) have recently emerged as a potential solution, offering distinct mechanisms compared to traditional probiotics. Among the NGPs, Bacteroides, a dominant bacterial genus in the human gut, has gained significant attention due to its prevalence, ability to break down plant polysaccharides, and production of short-chain fatty acids (SCFAs). Recent evidence has demonstrated that Bacteroides effectively reduces cholesterol levels, prevents obesity, and lowers the risk of CVD. However, research on Bacteroides is currently limited to a few species, leaving rooms for exploration of the beneficial functions of different species in this genus. In this study, we isolated 66 Bacteroides strains, including 9 distinct species, from healthy adults' fecal samples. By comparing their ability to assimilate cholesterol, we found that the transformation ability was not specific to any particular species. Notably, Bacteroides dorei YGMCC0564 revealed superior cholesterol-lowering capabilities and bile salt hydrolase (BSH) activity in vitro, surpassing that of Lactobacillus GG (LGG). YGMCC0564 exhibited favorable probiotic characteristics, including high survival rate in vitro simulation of gastrointestinal digestion, excellent adhesion ability, susceptibility to antibiotics, absence of hemolysis or virulence genes, and substantial production of SCFAs. The strain also demonstrated remarkable bile salt deconjugation activities and upregulation of the BT_416 gene associated with cholesterol, providing insights into a possible molecular mechanism underlying its cholesterol-reducing activity. These findings establish YGMCC0564 as a promising NPG candidate for improving cardiovascular health.
Collapse
Affiliation(s)
- Zhili He
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | - Tinghui Wang
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | | | - Kuojiang Shi
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | - Fan Wang
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | - Yanzhao Li
- Beijing Hotgen Biotechnology Inc., Beijing, China
| | - Chanqing Lin
- Beijing Hotgen Biotechnology Inc., Beijing, China
| | - Jianguo Chen
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| |
Collapse
|
64
|
Wang J, Wang L, Wang Q, Liu C, Zheng L. Lacticaseibacillus rhamnosus GG enhances fin regeneration under oxytetracycline exposure via activating Wnt signaling and modulating gut microbiota. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109155. [PMID: 37827248 DOI: 10.1016/j.fsi.2023.109155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Zebrafish possesses robust caudal fin regeneration which depends on multiple factors to maintain body integrity. However, it is uncertain whether the caudal fin regeneration is related to gut microbiota. Here, we investigated the effect of Lacticaseibacillus rhamnosus GG (LGG) on the regeneration of caudal fin under oxytetracycline (OTC) exposure. The results demonstrated that 1000 μg/L OTC exposure for 4 days decreased reactive oxygen species (ROS) production at 1 and 3 h post amputation (hpa), increased neutrophil recruitment at 6 hpa, enhanced the number of apoptotic cells at 1, 3, 6 and 12 hpa and inhibited Wnt signaling pathway at 48 hpa in wound site. Furthermore, OTC exposure caused dysbacteriosis by elevating level of Proteobacteria and decreasing the abundance of Firmicutes, particularly Lacticaseibacillus, thereby negatively impacting wound healing and repair. Additionally, the administration of 106 CFU/mL of LGG for 48 h could improve intestinal environment through increasing the colonization rate of LGG in OTC-treated larvae intestines. The regenerative process restored by LGG was accompanied with increased ROS production at 1, 3 and 6 hpa, inhibited neutrophil recruitment at 6 hpa, decreased the number of apoptotic cells at 1 hpa, and activated Wnt signaling pathway at 48 hpa in OTC-treated fish. LGG is a promising bacterium for restoring fin regeneration and provides new insights regarding the correlation among the gut microbiota and fin regeneration.
Collapse
Affiliation(s)
- Ju Wang
- Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lei Wang
- Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Qi Wang
- Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Changhong Liu
- Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Lei Zheng
- Engineering Research Center of Bio-Process, Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Intelligent Interconnected Systems Laboratory of Anhui Province, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
65
|
Splichalova A, Kindlova Z, Killer J, Neuzil Bunesova V, Vlkova E, Valaskova B, Pechar R, Polakova K, Splichal I. Commensal Bacteria Impact on Intestinal Toll-like Receptor Signaling in Salmonella-Challenged Gnotobiotic Piglets. Pathogens 2023; 12:1293. [PMID: 38003758 PMCID: PMC10675043 DOI: 10.3390/pathogens12111293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Gnotobiotic (GN) animals with simple and defined microbiota can help to elucidate host-pathogen interferences. Hysterectomy-derived germ-free (GF) minipigs were associated at 4 and 24 h post-hysterectomy with porcine commensal mucinolytic Bifidobacterium boum RP36 (RP36) strain or non-mucinolytic strain RP37 (RP37) or at 4 h post-hysterectomy with Lactobacillus amylovorus (LA). One-week-old GN minipigs were infected with Salmonella Typhimurium LT2 strain (LT2). We monitored histological changes in the ileum, mRNA expression of Toll-like receptors (TLRs) 2, 4, and 9 and their related molecules lipopolysaccharide-binding protein (LBP), coreceptors MD-2 and CD14, adaptor proteins MyD88 and TRIF, and receptor for advanced glycation end products (RAGE) in the ileum and colon. LT2 significantly induced expression of TLR2, TLR4, MyD88, LBP, MD-2, and CD14 in the ileum and TLR4, MyD88, TRIF, LBP, and CD14 in the colon. The LT2 infection also significantly increased plasmatic levels of inflammatory markers interleukin (IL)-6 and IL-12/23p40. The previous colonization with RP37 alleviated damage of the ileum caused by the Salmonella infection, and RP37 and LA downregulated plasmatic levels of IL-6. A defined oligo-microbiota composed of bacterial species with selected properties should probably be more effective in downregulating inflammatory response than single bacteria.
Collapse
Affiliation(s)
- Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Zdislava Kindlova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Jiri Killer
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Vera Neuzil Bunesova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
| | - Eva Vlkova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
| | - Barbora Valaskova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Radko Pechar
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, 165 00 Prague, Czech Republic; (J.K.); (V.N.B.); (E.V.); (R.P.)
- Department of Research, Food Research Institute Prague, 102 00 Prague, Czech Republic
| | - Katerina Polakova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic; (A.S.); (Z.K.); (B.V.); (K.P.)
| |
Collapse
|
66
|
Wei J, Li Z, Fan Y, Feng L, Zhong X, Li W, Guo T, Ning X, Li Z, Ou C. Lactobacillus rhamnosus GG aggravates vascular calcification in chronic kidney disease: A potential role for extracellular vesicles. Life Sci 2023; 331:122001. [PMID: 37625519 DOI: 10.1016/j.lfs.2023.122001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
AIMS Lactobacillus rhamnosus GG (LGG) is a probiotic with great promise in future clinical application, which can significantly promote bone formation. However, the effect of LGG on CKD-related vascular calcification is unclear. In this study, we aimed to investigate the effect of LGG on CKD-related vascular calcification. MATERIALS AND METHODS After 2 weeks of 5/6 nephrectomy, CKD rats received a special diet (4 % calcium and 1.8 % phosphate) combined with 1,25-dihydroxyvitamin D3 to induce vascular calcification. Meanwhile, CKD rats in the LGG group were gavaged orally with LGG (1 × 109 CFU bacteria/day). 16S RNA amplicon sequencing was performed to analyze the effect of LGG treatment on gut microbiota composition. Furthermore, differential ultracentrifugation was utilized to extract EVs. The effects of EVs on vascular calcification were evaluated in rat VSMCs, rat aortic rings, and CKD rat calcification models. In this study, vascular calcification was assessed by microcomputed tomography analysis, alizarin red staining, calcium content determination, and the expression of osteogenic transcription factors RUNX2 and BMP2. KEY FINDINGS LGG remarkably aggravated vascular calcification. LGG supplementation significantly altered gut microbiota composition in CKD rats, particularly increasing Lactobacillus. Interestingly, EVs presented a significant promoting effect on the development of calcification. Finally, mechanistic analysis proved that EVs aggravated vascular calcification through PI3K/AKT signaling. SIGNIFICANCE These results do not support the supplementation of LGG in CKD-associated vascular calcification patients. Our study presented a fresh perspective on LGG with potential risks and adverse effects. CKD patients should use specific probiotic strains cautiously.
Collapse
Affiliation(s)
- Jintao Wei
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Zehua Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Ying Fan
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xinglong Zhong
- Department of Cardiology, The Fourth Affiliated Hospital of Guangxi Medical University/Liuzhou Workers' Hospital, Liuzhou, PR China
| | - Weirun Li
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Tingting Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xiaodong Ning
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China.
| | - Caiwen Ou
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Dongguan 523018, PR China.
| |
Collapse
|
67
|
Zhang Y, Huang R, Jiang Y, Shen W, Pei H, Wang G, Pei P, Yang K. The role of bacteria and its derived biomaterials in cancer radiotherapy. Acta Pharm Sin B 2023; 13:4149-4171. [PMID: 37799393 PMCID: PMC10547917 DOI: 10.1016/j.apsb.2022.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
Bacteria-mediated anti-tumor therapy has received widespread attention due to its natural tumor-targeting ability and specific immune-activation characteristics. It has made significant progress in breaking the limitations of monotherapy and effectively eradicating tumors, especially when combined with traditional therapy, such as radiotherapy. According to their different biological characteristics, bacteria and their derivatives can not only improve the sensitivity of tumor radiotherapy but also protect normal tissues. Moreover, genetically engineered bacteria and bacteria-based biomaterials have further expanded the scope of their applications in radiotherapy. In this review, we have summarized relevant researches on the application of bacteria and its derivatives in radiotherapy in recent years, expounding that the bacteria, bacterial derivatives and bacteria-based biomaterials can not only directly enhance radiotherapy but also improve the anti-tumor effect by improving the tumor microenvironment (TME) and immune effects. Furthermore, some probiotics can also protect normal tissues and organs such as intestines from radiation via anti-inflammatory, anti-oxidation and apoptosis inhibition. In conclusion, the prospect of bacteria in radiotherapy will be very extensive, but its biological safety and mechanism need to be further evaluated and studied.
Collapse
Affiliation(s)
- Yu Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ruizhe Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yunchun Jiang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wenhao Shen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Pei Pei
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
68
|
Fu HY, Yu HD, Bai YP, Yue LF, Wang HM, Li LL. Effect and safety of probiotics for treating urticaria: A systematic review and meta-analysis. J Cosmet Dermatol 2023; 22:2663-2670. [PMID: 37221968 DOI: 10.1111/jocd.15782] [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: 12/29/2022] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND To assess the effect and safety of probiotics for treating urticaria. METHODS Randomized controlled trial (RCT) papers on the probiotics treatment published before May 2019 were retrieved from various databases like PubMed, EMbase, MEDLINE (Ovid), SCI-Hub, Springer, ClinicalKey, VIP, and CNKI. The treatment plan that we include are oral administration of single probiotic, multiple probiotics, and the combination of probiotics and antihistamines. Meta-analysis of the data was performed by RevMan 5.3 software. RESULTS A total of nine RCT papers were included: four papers for oral administration of single probiotic, three papers for oral administration of multiple probiotics, and two papers for oral administration of a probiotic combined with antihistamines. The results of meta-analysis showed that the therapeutic effect of the probiotic group was significantly higher than the control group (placebo or antihistamines) (RR = 1.09, 95% CI: 1.03-1.16, p = 0.006). And compared with the placebo group, the therapeutic effect of single probiotic group was significantly improved (RR = 1.11, 95% CI: 1.01-1.21, p = 0.03). Regarding therapeutic effect, there was no statistically significant difference between the multiple probiotics group and placebo group (RR = 1.00, 95% CI: 0.94 ~ 1.07, p = 0.91); the therapeutic effect of single probiotic combined antihistamine group was significantly higher than the antihistamine group (RR = 1.13, 95% CI: 1.07-1.19, p < 0.0001). Regarding the incidence of adverse reactions, there was no significant difference between the probiotic group and the control group (p = 0.46). CONCLUSION The treatment plan of oral administration of probiotics has significant therapeutic effects on urticaria, but the therapeutic effects of the administration of multiple probiotics and the safety of probiotic therapy are still not yet obvious. Some large-scale, multi-centered RCT studies are needed in the future for clarification.
Collapse
Affiliation(s)
- Hong-Yu Fu
- Department of Emergency, The Dongcheng District First People's Hospital of Beijing Municipality, Beijing city, China
| | - Hong-da Yu
- Department of Dermatology & STD, Beijing Dongzhimen Hospital, Beijing city, China
| | - Yan-Ping Bai
- Department of Dermatology & STD, Beijing Dongzhimen Hospital, Beijing city, China
| | - Li-Feng Yue
- Department of Encephalopathy, Beijing Dongzhimen Hospital, Beijing city, China
| | - Hong-Mei Wang
- Department of Emergency, The Dongcheng District First People's Hospital of Beijing Municipality, Beijing city, China
| | - Ling-Ling Li
- Department of Dermatology & STD, Beijing Dongzhimen Hospital, Beijing city, China
| |
Collapse
|
69
|
Cheng L, Shi J, Peng H, Tong R, Hu Y, Yu D. Probiotics and liver fibrosis: An evidence-based review of the latest research. J Funct Foods 2023; 109:105773. [DOI: 10.1016/j.jff.2023.105773] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
70
|
Poto R, Laniro G, de Paulis A, Spadaro G, Marone G, Gasbarrini A, Varricchi G. Is there a role for microbiome-based approach in common variable immunodeficiency? Clin Exp Med 2023; 23:1981-1998. [PMID: 36737487 PMCID: PMC9897624 DOI: 10.1007/s10238-023-01006-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023]
Abstract
Common variable immunodeficiency (CVID) is a primary immunodeficiency characterized by low levels of serum immunoglobulins and increased susceptibility to infections, autoimmune disorders and cancer. CVID embraces a plethora of heterogeneous manifestations linked to complex immune dysregulation. While CVID is thought to be due to genetic defects, the exact cause of this immune disorder is unknown in the large majority of cases. Compelling evidences support a linkage between the gut microbiome and the CVID pathogenesis, therefore a potential for microbiome-based treatments to be a therapeutic pathway for this disorder. Here we discuss the potential of treating CVID patients by developing a gut microbiome-based personalized approach, including diet, prebiotics, probiotics, postbiotics and fecal microbiota transplantation. We also highlight the need for a better understanding of microbiota-host interactions in CVID patients to prime the development of improved preventive strategies and specific therapeutic targets.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità (ISS), Rome, Italy
| | - Gianluca Laniro
- Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of Rome, Rome, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy
- World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131, Naples, Italy
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Catholic University of Rome, Rome, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131, Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131, Naples, Italy.
- World Allergy Organization (WAO), Center of Excellence, 80131, Naples, Italy.
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131, Naples, Italy.
| |
Collapse
|
71
|
Chen YW, Lee ML, Chiang CY, Fu E. Effects of systemic Bifidobacterium longum and Lactobacillus rhamnosus probiotics on the ligature-induced periodontitis in rat. J Dent Sci 2023; 18:1477-1485. [PMID: 37799895 PMCID: PMC10548012 DOI: 10.1016/j.jds.2023.04.013] [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: 03/06/2023] [Revised: 04/14/2023] [Indexed: 10/07/2023] Open
Abstract
Background/purpose Probiotics might be beneficial in preventing periodontitis. Effects of Bifidobacterium and Lactobacillus on periodontitis were examined using the ligature-induced rat model. Materials and methods Thirty-five male Sprague-Dawley rats were divided into control, ligation, Bifidobacterium longum (BL986), Lactobacillus rhamnosus (LRH09), and combination groups. Periodontitis was induced in maxillary second molars. From the day before ligation, phosphate-buffered saline (for control and ligation groups) or probiotics (2 × 109 CFU/g for probiotic groups) were fed daily. On day 8, gingival mRNA expressions for interleukin (IL)-1β, IL-6, tissue necrosis factor (TNF)-α, IL-10, and NF-κB were determined via qPCR. Micro-computed tomography (μCT) and histomorphometry were employed to examine periodontal destruction. Results Compared to the ligation group, mRNA of IL-1β, TNF-α, IL-6, and NF-κB in probiotic groups were significantly decreased, but IL-10 was increased. Besides, the IL-10 was more significant in the combination group than in single-use group. Through μCT, the cementoenamel junction (CEJ)-to-bone distance and trabecular separation in combination group were less than that in ligation group, although the bone volume fraction and trabecular number/thickness showed an increase in three probiotic groups. Histopathologically, the combination group had significantly smaller gingival inflammatory cell-infiltrated area and CEJ-to-epithelium distance than the ligation group and the group with BL986 or LRH09. Additionally, the CEJ-to-bone distance was significantly smaller in the combination group than in the ligation and BL986 groups. Conclusion Systemic combination of BL986 and LRH09 had a synergistic effect on enhancing IL-10 and ameliorating the induced experimental periodontitis, although the single-use still presented partially alleviative effects.
Collapse
Affiliation(s)
- Ying-Wu Chen
- Periodontics Division, Department of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Lun Lee
- Institute of Dental Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Yang Chiang
- Periodontics Division, Department of Dentistry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Institute of Dental Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Earl Fu
- Department of Dentistry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Xindian, New Taipei City, Taiwan
| |
Collapse
|
72
|
Zhong Z, Liu G, Tang Z, Xiang S, Yang L, Huang L, He Y, Fan T, Liu S, Zheng X, Zhang T, Qi Y, Huang J, Zhang Y. Efficient plant genome engineering using a probiotic sourced CRISPR-Cas9 system. Nat Commun 2023; 14:6102. [PMID: 37773156 PMCID: PMC10541446 DOI: 10.1038/s41467-023-41802-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023] Open
Abstract
Among CRISPR-Cas genome editing systems, Streptococcus pyogenes Cas9 (SpCas9), sourced from a human pathogen, is the most widely used. Here, through in silico data mining, we have established an efficient plant genome engineering system using CRISPR-Cas9 from probiotic Lactobacillus rhamnosus. We have confirmed the predicted 5'-NGAAA-3' PAM via a bacterial PAM depletion assay and showcased its exceptional editing efficiency in rice, wheat, tomato, and Larix cells, surpassing LbCas12a, SpCas9-NG, and SpRY when targeting the identical sequences. In stable rice lines, LrCas9 facilitates multiplexed gene knockout through coding sequence editing and achieves gene knockdown via targeted promoter deletion, demonstrating high specificity. We have also developed LrCas9-derived cytosine and adenine base editors, expanding base editing capabilities. Finally, by harnessing LrCas9's A/T-rich PAM targeting preference, we have created efficient CRISPR interference and activation systems in plants. Together, our work establishes CRISPR-LrCas9 as an efficient and user-friendly genome engineering tool for diverse applications in crops and beyond.
Collapse
Affiliation(s)
- Zhaohui Zhong
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, School of Life Sciences, Southwest University, 400715, Chongqing, China
| | - Guanqing Liu
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, 225012, Yangzhou, China
- Key Laboratory of Plant Functional Genomics of the Ministry of Education/Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, 225012, Yangzhou, China
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, 225012, Yangzhou, China
| | - Zhongjie Tang
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Shuyue Xiang
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Liang Yang
- Horticulture Research Institute, Sichuan Academy of Agricultural Sciences, Sichuan, China
- Vegetable Germplasm Innovation and Variety Improvement Key Laboratory of Sichuan Province, 610066, Chengdu, China
| | - Lan Huang
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Yao He
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Tingting Fan
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Shishi Liu
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
| | - Xuelian Zheng
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, School of Life Sciences, Southwest University, 400715, Chongqing, China
| | - Tao Zhang
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, 225012, Yangzhou, China
- Key Laboratory of Plant Functional Genomics of the Ministry of Education/Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, 225012, Yangzhou, China
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, 225012, Yangzhou, China
| | - Yiping Qi
- Department of Plant Science and Landscape Architecture, University of Maryland, College Park, MD, 20742, USA.
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD, 20850, USA.
| | - Jian Huang
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China.
| | - Yong Zhang
- Department of Biotechnology, School of Life Sciences and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, 610054, Chengdu, China.
- Chongqing Key Laboratory of Plant Resource Conservation and Germplasm Innovation, Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, School of Life Sciences, Southwest University, 400715, Chongqing, China.
| |
Collapse
|
73
|
Zhao Y, Kang Y, Zhao Z, Yang G, Gao Y, Gao L, Wang C, Li S. Lacticaseibacillus rhamnosus TF318 prevents depressive behavior in rats by inhibiting HPA-axis hyperactivity and upregulating BDNF expression. Neurosci Lett 2023; 814:137460. [PMID: 37619699 DOI: 10.1016/j.neulet.2023.137460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Growing evidence suggests that probiotics can ameliorate depression by regulating the microbiota-gut-brain axis. However, the mechanism of action of probiotics in depressive disorders remains incompletely understood. This study aimed to investigate the effect of Lacticaseibacillus rhamnosus TF318 in a corticosterone (CORT)-induced rat model of depression. The sucrose preference test (SPT) and Morris water maze (MWM) test showed that oral administration of L. rhamnosus TF318 for 21 d significantly prevented depressive behaviors. Administration of L. rhamnosus TF318 resulted in lower hippocampal levels of adrenocorticotropic hormone and corticotropin-releasing factor and serum levels of CORT and restoration of hippocampal levels of 5-hydroxytryptamine, dopamine, and norepinephrine. A marked increase was observed in the hippocampal concentration of brain-derived neurotrophic factor (BDNF), a change that may have involved the cyclic adenosine monophosphate (cAMP)/cAMP response element-binding (CREB)/BDNF signaling pathway. Treatment with L. rhamnosus TF318 corrected CORT-induced abnormalities in the gut microbiota, significantly increasing the relative abundance of Firmicutes. In conclusion, supplementation with L. rhamnosus TF318 prevented CORT-induced depressive behaviors by upregulating BDNF expression and modulating gut microbiota, suggesting that this strain has the potential as a novel probiotic with antidepressant effects.
Collapse
Affiliation(s)
- Yujuan Zhao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - You Kang
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Zijian Zhao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Ge Yang
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Yansong Gao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Lei Gao
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Chao Wang
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China
| | - Shengyu Li
- Institute of Agricultural Products Processing Technology, Jilin Academy of Agricultural Sciences/National R&D Center for Milk Processing, Changchun 130033, PR China.
| |
Collapse
|
74
|
Asensio-Grau A, Calvo-Lerma J, Ferriz-Jordán M, García-Hernández J, Heredia A, Andrés A. Effect of Lactobacillaceae Probiotics on Colonic Microbiota and Metabolite Production in Cystic Fibrosis: A Comparative In Vitro Study. Nutrients 2023; 15:3846. [PMID: 37686878 PMCID: PMC10490339 DOI: 10.3390/nu15173846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Cystic Fibrosis-related gut dysbiosis (CFRGD) has become a recognised complication in children with this condition, and current evidence remains insufficient to guide the selection of probiotic strains for supplementation treatments. The aim of this study was to characterise the effect of three probiotic strains on CFRGD by means of a dynamic in vitro simulation of the colonic fermentation (SHIME®). The configuration of the system included three bioreactors colonised with the faecal inoculum of a child with cystic fibrosis. For 20 days, each bioreactor was supplied daily with either Lacticaseibacillus rhamnosus GG (ATCC 53103 TM), Limosilactobacillus reuteri (DSM 17938) or Lactiplantibacillus plantarum (DSM 22266). The baseline microbiota was characterised by a high abundance of Prevotella, Faecalibacterium and Acidaminococcus genera. After 20 days of supplementation, L. rhamnosus and L. plantarum reduced Prevotella significantly, and the three strains led to increased Faecalibacterium and Bifidobacterium and decreased Acidaminococcus, with some of these changes being maintained 10 days after ceasing supplementation. The metabolic activity remained unaltered in terms of short-chain fatty acids, but branched-chain fatty acids showed a significant decrease, especially with L. plantarum. Additionally, ammonia decreased at 20 days of supplementation, and lactate continuously increased with the three strains. The effects on colonic microbiota of L. rhamnosus, L. reuteri or L. plantarum were established, including increased beneficial bacteria, such as Faecalibacterium, and beneficial metabolites such as lactate; and on the other hand, a reduction in pathogenic genera, including Prevotella or Acidaminococcus and branched-chain fatty acids, overall supported their use as probiotics in the context of CFRGD.
Collapse
Affiliation(s)
- Andrea Asensio-Grau
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Joaquim Calvo-Lerma
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Miguel Ferriz-Jordán
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
| | - Jorge García-Hernández
- Advanced Food Microbiology Centre (CAMA), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain
| | - Ana Heredia
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| | - Ana Andrés
- Institute of Food Engineering (IIA-FoodUPV), Polytechnic University of Valencia, Camino de Vera s/n, 46022 València, Spain; (A.A.-G.); (M.F.-J.); (A.A.)
- Joint Research Unit NutriCura, Avda. Fernando Abril Martorell 106, 46026 València, Spain
| |
Collapse
|
75
|
Hou Y, Zheng S, Zou F, Wang D, Da H, Zhou Y, Fan X, Liu J, Zhao H, He J, Li H, Sun X, Liu Y. Lactobacillus rhamnosus 76 alleviates airway inflammation in ovalbumin-allergic mice and improves mucus secretion by down-regulating STAT6/SPDEF pathway. Immunobiology 2023; 228:152712. [PMID: 37515878 DOI: 10.1016/j.imbio.2023.152712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 07/31/2023]
Abstract
Previous studies have reported a correlation between the dysregulation of intestinal microbiota and the occurrence of asthma. This study aimed to investigate the effect of probiotic Lactobacillus rhamnosus 76 (LR76) on ovalbumin (OVA)-allergic mice and the mechanism of LR76 affecting mucus secretion in asthma. OVA-allergic mice were supplemented with LR76, and 16HBE cells induced by interleukin-13 (IL-13) were treated with LR76 supernatant (LR76-s) to observe the effect of LR76. In OVA-sensitized mice, LR76 alleviated the inflammatory cell infiltration in lung tissue and reduced the inflammatory cell counts of BALF. The expression level of mRNA, including Il4, Il5, Il13, Il25, Tgfb1, Il10, and Ifng, was decreased in the lung tissue of mice in the LR76 group compared with the OVA group. MUC5AC expression was down-regulated, while SCGB1A1 was up-regulated in the lung tissue of OVA-allergic mice after being supplemented with LR76 and in 16HBE cells induced by IL-13 after incubating with LR76-s. LR76 and LR76-s down-regulated the expression of proteins, including STAT6, p-STAT6, and SPDEF, and mRNA of STAT6 and SPDEF. In conclusion, LR76 alleviated airway inflammation and Th2 response in OVA-allergic mice and improved the mucus secretion of mouse lung tissue and 16HBE cells in the asthma model by down-regulating STAT6/SPDEF pathway.
Collapse
Affiliation(s)
- Yangfan Hou
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Shuping Zheng
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Fan Zou
- Department of Respiratory and Critical Care Medicine, Affliated Hospital of Zunyi Medical University, Zunyi 563001, Guizhou Province, PR China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Hongju Da
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Yong Zhou
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Xinping Fan
- Department of Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Jianghao Liu
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Hongyan Zhao
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Jin He
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Hongxin Li
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Xiuzhen Sun
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, PR China.
| |
Collapse
|
76
|
Mego M, Danis R, Chovanec J, Jurisova S, Bystricky B, Porsok S, Konkolovsky P, Vaclav V, Wagnerova M, Streško M, Brezinova B, Rečková M, Sutekova D, Pazderova N, Novisedlakova M, Zomborska E, Ciernikova S, Svetlovska D, Drgona L. Randomized double-blind, placebo-controlled multicenter phase III study of prevention of irinotecan-induced diarrhea by a probiotic mixture containing Bifidobacterium BB-12 ®Lactobacillus rhamnosus LGG ® in colorectal cancer patients. Front Oncol 2023; 13:1168654. [PMID: 37601667 PMCID: PMC10438450 DOI: 10.3389/fonc.2023.1168654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Background The incidence of irinotecan-induced diarrhea varies between 60-90%, by which the incidence of severe diarrhea is 20-40%. The objective of this phase III trial was to determine the effectiveness of the probiotic mixture containing Bifidobacterium, BB-12® and Lactobacillus rhamnosus, LGG® in the prophylaxis of irinotecan-induced diarrhea in metastatic colorectal cancer patients due to a reduction in the activity of intestinal beta-D-glucuronidase. Methods From March 2016 to May 2022, a total of 242 patients with colorectal cancer starting a new line of irinotecan-based therapy were registered to the study in 11 cancer centers in Slovakia. Patients were randomized in a ratio 1:1 to probiotic formula vs. placebo that was administered for 6 weeks. Each capsule of Probio-Tec® BG-Vcap-6.5 contained 2.7x109 colony-forming units (CFU) of 2 lyophilized probiotic strains Bifidobacterium, BB-12® (50%) and Lactobacillus rhamnosus GG, LGG® (50%). Results Administration of probiotics compared to placebo was not associated with a significant reduction of grade 3/4 diarrhea (placebo arm 11.8% vs. probiotic arm 7.9%, p=0.38). Neither the overall incidence of diarrhea (46.2% vs. 41.2%, p=0.51) nor the incidence of enterocolitis (3.4% vs. 0.9%, p=0.37) was different in the placebo vs. probiotic arm. Subgroup analysis revealed that patients with colostomy had higher incidence of any diarrhea and grade 3/4 diarrhea in the placebo arm compared to the probiotic arm (48.5% vs. 22.2%, p=0.06 and 15.2% vs. 0%, p=0.06, respectively). Moreover, patients on probiotic arm had significantly better diarrhea-free survival (HR = 0.41, 95%CI 0.18 - 0.95, p=0.05) and needed less loperamide (p=0.01) compared to patients on placebo arm. We did not observe any infection caused by probiotic strains used in this study. Conclusion This study failed to achieve its primary endpoint, and results suggest a lack of benefit of administered probiotic formula for the prevention of irinotecan-induced diarrhea. However, subgroup analysis suggests a possible benefit in patients with colostomy.
Collapse
Affiliation(s)
- Michal Mego
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Radoslav Danis
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Jozef Chovanec
- Department of Oncology, St. Jacob Hospital, Bardejov, Slovakia
| | - Silvia Jurisova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Stefan Porsok
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Vladimir Vaclav
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Maria Wagnerova
- Department of Oncology, East Slovakia Comprehensive Cancer Center, Kosice, Slovakia
| | - Marian Streško
- Department of Oncology, Faculty Hospital, Trnava, Trebisov, Slovakia
| | | | - Mária Rečková
- Department of Oncology, Regional Cancer Center, Poprad, Slovakia
| | - Dagmar Sutekova
- Department of Oncology, University Hospital Martin, Martin, Slovakia
| | - Natalia Pazderova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Mária Novisedlakova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Eva Zomborska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Sona Ciernikova
- Biomedical Center, Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniela Svetlovska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Lubos Drgona
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
77
|
Li X, Hu B, Zheng J, Pan Z, Cai Y, Zhao M, Jin X, Li ZQ. Probiotics Alleviate Chemotherapy-Associated Intestinal Mucosal Injury via the TLR4-NFκB Signaling Pathway. Drug Des Devel Ther 2023; 17:2183-2192. [PMID: 37521036 PMCID: PMC10386857 DOI: 10.2147/dddt.s403087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Temozolomide (TMZ) induces intestinal mucosa injury that cannot be fully counteracted by supportive treatment. Probiotics regulate gut microbial composition and the host immune system and may alleviate this side effect. We aimed to investigate the potential and mechanism of Lactobacillus rhamnosus GG (LGG) in relieving intestinal mucosal injury induced by TMZ. Methods Glioblastoma mice were divided into four groups: CON (control), LGG (109 CFU/mL, treated for 7 days), TMZ (50 mg/kg·d, treated for 5 days), LGG+TMZ (LGG for 7 days and TMZ subsequently for 5 days). Body weight, food intake, and fecal pH were recorded. Intestinal tissue samples were collected 1 day after the end of TMZ treatment. Degree of damage to intestine, expression of IL1β, IL6, TNFα, and IL10 in jejunum were determined. Levels of tight-junction proteins (ZO1, occludin), TLR4, IKKβ, IκBα, and P65 with their phosphorylation in jejunum were measured. Results Decreases in body weight, food intake, spleen index in the TMZ group were mitigated in the LGG+TMZ group, and the degree of intestinal shortening and damage to jejunum villus were also alleviated. The expression of tight-junction proteins in the LGG+TMZ group was significantly greater than that in the TMZ group. IκBα in intestinal tissue significantly decreased in the TMZ group, phos-IKKβ and phos-P65 increased compared to the CON group, and LGG reversed such changes in IκBα and phos-P65 in the LGG+TMZ group. Intestinal inflammatory cytokines were significantly increased in the TMZ group, but lower in the LGG+TMZ group. Moreover, expression of TLR4 in LGG group was significantly lower than that in the CON group. LGG inhibited the rise of TLR4 after TMZ in the LGG+TMZ group compared to the TMZ group. Conclusion LGG inhibits the activation of the TLR4-NFκB pathway and alleviates intestinal mucosal inflammation induced by TMZ, thereby protect the jejunum villi and mucosal physical barrier.
Collapse
Affiliation(s)
- Xiaochong Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Bowen Hu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Jiachen Zheng
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
- The Second Clinical School, Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Zhiyong Pan
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Yuxiang Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Mingjuan Zhao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Xiaoqing Jin
- Emergency Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| | - Zhi-Qiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People’s Republic of China
| |
Collapse
|
78
|
Stolfi C, Pacifico T, Monteleone G, Laudisi F. Impact of Western Diet and Ultra-Processed Food on the Intestinal Mucus Barrier. Biomedicines 2023; 11:2015. [PMID: 37509654 PMCID: PMC10377275 DOI: 10.3390/biomedicines11072015] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/19/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The intestinal epithelial barrier plays a key role in the absorption of nutrients and water, in the regulation of the interactions between luminal contents and the underlying immune cells, and in the defense against enteric pathogens. Additionally, the intestinal mucus layer provides further protection due to mucin secretion and maturation by goblet cells, thus representing a crucial player in maintaining intestinal homeostasis. However, environmental factors, such as dietary products, can disrupt this equilibrium, leading to the development of inflammatory intestinal disorders. In particular, ultra-processed food, which is broadly present in the Western diet and includes dietary components containing food additives and/or undergoing multiple industrial processes (such as dry heating cooking), was shown to negatively impact intestinal health. In this review, we summarize and discuss current knowledge on the impact of a Western diet and, in particular, ultra-processed food on the mucus barrier and goblet cell function, as well as potential therapeutic approaches to maintain and restore the mucus layer under pathological conditions.
Collapse
Affiliation(s)
- Carmine Stolfi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy
| |
Collapse
|
79
|
Alcántara C, Perez M, Huedo P, Altadill T, Espadaler-Mazo J, Arqués JL, Zúñiga M, Monedero V. Study of the biosynthesis and functionality of polyphosphate in Bifidobacterium longum KABP042. Sci Rep 2023; 13:11076. [PMID: 37422465 PMCID: PMC10329679 DOI: 10.1038/s41598-023-38082-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/03/2023] [Indexed: 07/10/2023] Open
Abstract
Polyphosphate (poly-P) biosynthesis in bacteria has been linked to many physiological processes and has been characterized as an interesting functional molecule involved in intestinal homeostasis. We determined the capacity for poly-P production of 18 probiotic strains mainly belonging to Bifidobacterium and former Lactobacillus genera, showing that poly-P synthesis varied widely between strains and is dependent on the availability of phosphate and the growth phase. Bifidobacteria were especially capable of poly-P synthesis and poly-P kinase (ppk) genes were identified in their genomes together with a repertoire of genes involved in phosphate transport and metabolism. In Bifidobacterium longum KABP042, the strain we found with highest poly-P production, variations in ppk expression were linked to growth conditions and presence of phosphate in the medium. Moreover, the strain produced poly-P in presence of breast milk and lacto-N-tetraose increased the amount of poly-P synthesized. Compared to KABP042 supernatants low in poly-P, exposure of Caco-2 cells to KABP042 supernatants rich in poly-P resulted in decreased epithelial permeability and increased barrier resistance, induction of epithelial protecting factors such as HSP27 and enhanced expression of tight junction protein genes. These results highlight the role of bifidobacteria-derived poly-P as a strain-dependent functional factor acting on epithelial integrity.
Collapse
Affiliation(s)
- Cristina Alcántara
- Laboratorio de Bacterias Lácticas y Probióticos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), 46980, Paterna, Valencia, Spain
| | - Marta Perez
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
| | - Pol Huedo
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
| | - Tatiana Altadill
- R&D Department, AB-Biotics S.A. (Part of Kaneka Corporation), Barcelona, Spain
- Basic Sciences Department, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Juan Luis Arqués
- Departamento de Tecnología de Alimentos, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid, Spain
| | - Manuel Zúñiga
- Laboratorio de Bacterias Lácticas y Probióticos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), 46980, Paterna, Valencia, Spain
| | - Vicente Monedero
- Laboratorio de Bacterias Lácticas y Probióticos, Instituto de Agroquímica y Tecnología de Alimentos (IATA-CSIC), 46980, Paterna, Valencia, Spain.
| |
Collapse
|
80
|
Huang C, Hao W, Wang X, Zhou R, Lin Q. Probiotics for the treatment of ulcerative colitis: a review of experimental research from 2018 to 2022. Front Microbiol 2023; 14:1211271. [PMID: 37485519 PMCID: PMC10358780 DOI: 10.3389/fmicb.2023.1211271] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Ulcerative colitis (UC) has become a worldwide public health problem, and the prevalence of the disease among children has been increasing. The pathogenesis of UC has not been elucidated, but dysbiosis of the gut microbiota is considered the main cause of chronic intestinal inflammation. This review focuses on the therapeutic effects of probiotics on UC and the potential mechanisms involved. In animal studies, probiotics have been shown to alleviate symptoms of UC, including weight loss, diarrhea, blood in the stool, and a shortened colon length, while also restoring intestinal microecological homeostasis, improving gut barrier function, modulating the intestinal immune response, and attenuating intestinal inflammation, thereby providing theoretical support for the development of probiotic-based microbial products as an adjunctive therapy for UC. However, the efficacy of probiotics is influenced by factors such as the bacterial strain, dose, and form. Hence, the mechanisms of action need to be investigated further. Relevant clinical trials are currently lacking, so the extension of animal experimental findings to clinical application requires a longer period of consideration for validation.
Collapse
Affiliation(s)
- Cuilan Huang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Wujuan Hao
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Xuyang Wang
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| | - Renmin Zhou
- Department of Digestive, Affiliated Children’s Hospital of Jiangnan University, Wuxi, China
| | - Qiong Lin
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi Children’s Hospital, Wuxi, China
| |
Collapse
|
81
|
Qian X, Tian P, Lin G, Xu X, Wang G, Zhang H, Chen W. Detection of colonization capacity of probiotic Bifidobacterium breve CCFM1025 in the human gut. Future Microbiol 2023; 18:595-606. [PMID: 37314317 DOI: 10.2217/fmb-2022-0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Abstract
Aim: To detect the gut colonization capacity of Bifidobacterium breve CCFM1025 with clinical antidepressant-like effects. Materials & methods: A unique gene sequence of B. breve CCFM1025 was discovered based on the genome analysis of 104 B. breve strains and a strain-specific primer (1025T5) was designed. In vitro and in vivo samples were used to validate the specificity and quantitative capability of this primer in the PCR system. Results: Quantitative PCR using strain-specific primers enabled absolute quantification of CCFM1025 in fecal samples within 104-1010 cells/g (R2 >0.99). CCFM1025 remained highly detectable in volunteer feces 14 days after cessation of administration, demonstrating its favorable colonization characteristics. Conclusion: CCFM1025 can colonize the healthy human gut.
Collapse
Affiliation(s)
- Xin Qian
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Peijun Tian
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Guopeng Lin
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xinglong Xu
- Jingjiang Chinese Medicine Hospital, Taizhou, Jiangsu, 214500, China
| | - Gang Wang
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science & Resources, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- School of Food Science & Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
82
|
Liu L, Narrowe AB, Firrman JA, Mahalak KK, Bobokalonov JT, Lemons JMS, Bittinger K, Daniel S, Tanes C, Mattei L, Friedman ES, Soares JW, Kobori M, Zeng WB, Tomasula PM. Lacticaseibacillus rhamnosus Strain GG (LGG) Regulate Gut Microbial Metabolites, an In Vitro Study Using Three Mature Human Gut Microbial Cultures in a Simulator of Human Intestinal Microbial Ecosystem (SHIME). Foods 2023; 12:2105. [PMID: 37297350 PMCID: PMC10252382 DOI: 10.3390/foods12112105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/13/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
In the present research, we investigated changes in the gut metabolome that occurred in response to the administration of the Laticaseibacillus rhamnosus strain GG (LGG). The probiotics were added to the ascending colon region of mature microbial communities established in a human intestinal microbial ecosystem simulator. Shotgun metagenomic sequencing and metabolome analysis suggested that the changes in microbial community composition corresponded with changes to metabolic output, and we can infer linkages between some metabolites and microorganisms. The in vitro method permits a spatially-resolved view of metabolic transformations under human physiological conditions. By this method, we found that tryptophan and tyrosine were mainly produced in the ascending colon region, while their derivatives were detected in the transverse and descending regions, revealing sequential amino acid metabolic pathways along with the colonic tract. The addition of LGG appeared to promote the production of indole propionic acid, which is positively associated with human health. Furthermore, the microbial community responsible for the production of indole propionic acid may be broader than is currently known.
Collapse
Affiliation(s)
- LinShu Liu
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Adrienne B. Narrowe
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Jenni A. Firrman
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Karley K. Mahalak
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Jamshed T. Bobokalonov
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
- V.I. Nikitin Chemistry Institute of Tajikistan Academy of Sciences, Dushanbe 734063, Tajikistan
| | - Johanna M. S. Lemons
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Scott Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lisa Mattei
- Division of Gastroenterology, Hepatology, and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Elliot S. Friedman
- Division of Gastroenterology & Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jason W. Soares
- Bioprocessing and Bioengineering Group, US Army Combat Capabilities Development Command Soldier Center (CCDC-SC), Natick, MA 01760, USA
| | - Masuko Kobori
- Food Research Institute, National Agriculture and Food Research Organization, Tsukuba 305-8642, Ibaraki, Japan
| | - Wei-Bin Zeng
- Department of Mathematics, University of Louisville, Louisville, KY 40292, USA
| | - Peggy M. Tomasula
- Dairy and Functional Foods Research Unit, Eastern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Wyndmoor, PA 19038, USA
| |
Collapse
|
83
|
Pagnini C, Di Paolo MC, Urgesi R, Pallotta L, Fanello G, Graziani MG, Delle Fave G. Safety and Potential Role of Lactobacillus rhamnosus GG Administration as Monotherapy in Ulcerative Colitis Patients with Mild-Moderate Clinical Activity. Microorganisms 2023; 11:1381. [PMID: 37374884 DOI: 10.3390/microorganisms11061381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Probiotics are microorganisms that confer benefits to the host, and, for this reason, they have been proposed in several pathologic states. Specifically, probiotic bacteria have been investigated as a therapeutic option in ulcerative colitis (UC) patients, but clinical results are dishomogeneous. In particular, many probiotic species with different therapeutic schemes have been proposed, but no study has investigated probiotics in monotherapy in adequate trials for the induction of remission. Lactobacillus rhamnosus GG (LGG) is the more intensively studied probiotic and it has ideal characteristics for utilization in UC patients. The aim of the present study is to investigate the clinical efficacy and safety of LGG administration in an open trial, delivered in monotherapy at two different doses, in UC patients with mild-moderate disease. The UC patients with mild-moderate disease activity (Partial Mayo score ≥ 2) despite treatment with oral mesalamine were included. The patients stopped oral mesalamine and were followed up for one month, then were randomized to receive LGG supplement at dose of 1.2 or 2.4 × 1010 CFU/day for one month. At the end of the study, the clinical activity was evaluated and compared to that at the study entrance (efficacy). Adverse events were recorded (safety). The primary end-point was clinical improvement (reduction in the Partial Mayo score) and no serious adverse events, while the secondary end-points were the evaluation of different efficacies and safeties between the two doses of LGG. The patients with disease flares dropped out of the study and went back to standard therapy. The efficacy data were analyzed in an intention-to-treat (ITT) and per-protocol (PP) analysis. Out of the 76 patients included in the study, 75 started the probiotic therapy (n = 38 and 37 per group). In the ITT analysis, 32/76 (42%) responded to treatment, 21/76 (28%) remained stable, and 23/76 (30%) had a worsening of their clinical condition; 55 (72%) completed the treatment and were analyzed in a PP analysis: 32/55 (58%) had a clinical response, 21 (38%) remained stable, and 2 (4%) had a light worsening of their clinical condition (p < 0.0001). Overall, 37% of the patients had a disease remission. No severe adverse event was recorded, and only one patient stopped therapy due to obstinate constipation. No difference in the clinical efficacy and safety has been recorded between groups treated with different doses of LGG. The present prospective clinical trial demonstrates, for the first time, that LGG in monotherapy is safe and effective for the induction of remission in UC patients with mild-moderate disease activity (ClinicalTrials.gov identifier: NCT04102852).
Collapse
Affiliation(s)
- Cristiano Pagnini
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Carla Di Paolo
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Riccardo Urgesi
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Lorella Pallotta
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Fanello
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Maria Giovanna Graziani
- Department of Gastroenterology and Digestive Endoscopy, S. Giovanni Addolorata Hospital, Via dell'Amba Aradam 9, 00184 Rome, Italy
| | - Gianfranco Delle Fave
- Department of Gastroenterology, "Sapienza" University of Rome, 00185 Rome, Italy
- Onlus "S. Andrea", 00199 Rome, Italy
| |
Collapse
|
84
|
Lopez-Escalera S, Lund ML, Hermes GDA, Choi BSY, Sakamoto K, Wellejus A. In Vitro Screening for Probiotic Properties of Lactobacillus and Bifidobacterium Strains in Assays Relevant for Non-Alcoholic Fatty Liver Disease Prevention. Nutrients 2023; 15:nu15102361. [PMID: 37242245 DOI: 10.3390/nu15102361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial metabolic disorder that poses health challenges worldwide and is expected to continue to rise dramatically. NAFLD is associated with metabolic syndrome, type 2 diabetes mellitus, and impaired gut health. Increased gut permeability, caused by disturbance of tight junction proteins, allows passage of damaging microbial components that, upon reaching the liver, have been proposed to trigger the release of inflammatory cytokines and generate cellular stress. A growing body of research has suggested the utilization of targeted probiotic supplements as a preventive therapy to improve gut barrier function and tight junctions. Furthermore, specific microbial interactions and metabolites induce the secretion of hormones such as GLP-1, resulting in beneficial effects on liver health. To increase the likelihood of finding beneficial probiotic strains, we set up a novel screening platform consisting of multiple in vitro and ex vivo assays for the screening of 42 bacterial strains. Analysis of transepithelial electrical resistance response via co-incubation of the 42 bacterial strains with human colonic cells (Caco-2) revealed improved barrier integrity. Then, strain-individual metabolome profiling was performed revealing species-specific clusters. GLP-1 secretion assay with intestinal secretin tumor cell line (STC-1) found at least seven of the strains tested capable of enhancing GLP-1 secretion in vitro. Gene expression profiling in human biopsy-derived intestinal organoids was performed using next generation sequencing transcriptomics post bacterial co-incubation. Here, different degrees of immunomodulation by the increase in certain cytokine and chemokine transcripts were found. Treatment of mouse primary hepatocytes with selected highly produced bacterial metabolites revealed that indole metabolites robustly inhibited de novo lipogenesis. Collectively, through our comprehensive bacterial screening pipeline, not previously ascribed strains from both Lactobacillus and Bifidobacterium genera were proposed as potential probiotics based on their ability to increase epithelial barrier integrity and immunity, promote GLP-1 secretion, and produce metabolites relevant to liver health.
Collapse
Affiliation(s)
- Silvia Lopez-Escalera
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
- Fakultät für Biowissenschaften, Friedrich-Schiller Universität Jena, Bachstraβe 18K, 07743 Jena, Germany
| | - Mari L Lund
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Gerben D A Hermes
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| | - Béatrice S-Y Choi
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Anja Wellejus
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970 Hørsholm, Denmark
| |
Collapse
|
85
|
Vinderola G, Cotter PD, Freitas M, Gueimonde M, Holscher HD, Ruas-Madiedo P, Salminen S, Swanson KS, Sanders ME, Cifelli CJ. Fermented foods: a perspective on their role in delivering biotics. Front Microbiol 2023; 14:1196239. [PMID: 37250040 PMCID: PMC10213265 DOI: 10.3389/fmicb.2023.1196239] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Fermented foods are often erroneously equated with probiotics. Although they might act as delivery vehicles for probiotics, or other 'biotic' substances, including prebiotics, synbiotics, and postbiotics, stringent criteria must be met for a fermented food to be considered a 'biotic'. Those criteria include documented health benefit, sufficient product characterization (for probiotics to the strain level) and testing. Similar to other functional ingredients, the health benefits must go beyond that of the product's nutritional components and food matrix. Therefore, the 'fermented food' and 'probiotic' terms may not be used interchangeably. This concept would apply to the other biotics as well. In this context, the capacity of fermented foods to deliver one, several, or all biotics defined so far will depend on the microbiological and chemical level of characterization, the reproducibility of the technological process used to produce the fermented foods, the evidence for health benefits conferred by the biotics, as well as the type and amount of testing carried out to show the probiotic, prebiotic, synbiotic, and postbiotic capacity of that fermented food.
Collapse
Affiliation(s)
- Gabriel Vinderola
- Instituto de Lactología Industrial (CONICET-UNL), Faculty of Chemical Engineering, National University of Litoral, Santa Fe, Argentina
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark and APC Microbiome Ireland, Cork, Ireland
| | - Miguel Freitas
- Health and Scientific Affairs, Danone North America, White Plains, NY, United States
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias—Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Asturias, Spain
| | - Hannah D. Holscher
- Department of Food Science and Human Nutrition, Division of Nutritional Sciences, 260 Edward R. Madigan Laboratory, University of Illinois, Urbana, IL, United States
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias—Consejo Superior de Investigaciones Científicas (IPLA-CSIC), Villaviciosa, Asturias, Spain
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| | - Kelly S. Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, United States
| | | |
Collapse
|
86
|
Furone F, Bellomo C, Carpinelli M, Nicoletti M, Hewa-Munasinghege FN, Mordaa M, Mandile R, Barone MV, Nanayakkara M. The protective role of Lactobacillus rhamnosus GG postbiotic on the alteration of autophagy and inflammation pathways induced by gliadin in intestinal models. Front Med (Lausanne) 2023; 10:1085578. [PMID: 37215707 PMCID: PMC10192745 DOI: 10.3389/fmed.2023.1085578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/21/2023] [Indexed: 05/24/2023] Open
Abstract
Celiac disease (CD) is an autoimmune enteropathy caused by an abnormal immune response to gliadin peptides in genetically predisposed individuals. For people with CD, the only available therapy thus far is the lifelong necessity for a gluten-free diet (GFD). Innovative therapies include probiotics and postbiotics as dietary supplements, both of which may benefit the host. Therefore, the present study aimed to investigate the possible beneficial effects of the postbiotic Lactobacillus rhamnosus GG (LGG) in preventing the effects induced by indigested gliadin peptides on the intestinal epithelium. In this study, these effects on the mTOR pathway, autophagic function, and inflammation have been evaluated. Furthermore, in this study, we stimulated the Caco-2 cells with the undigested gliadin peptide (P31-43) and with the crude gliadin peptic-tryptic peptides (PTG) and pretreated the samples with LGG postbiotics (ATCC 53103) (1 × 108). In this study, the effects induced by gliadin before and after pretreatment have also been investigated. The phosphorylation levels of mTOR, p70S6K, and p4EBP-1 were increased after treatment with PTG and P31-43, indicating that the intestinal epithelial cells responded to the gliadin peptides by activating the mTOR pathway. Moreover, in this study, an increase in the phosphorylation of NF-κβ was observed. Pretreatment with LGG postbiotic prevented both the activation of the mTOR pathway and the NF-κβ phosphorylation. In addition, P31-43 reduced LC3II staining, and the postbiotic treatment was able to prevent this reduction. Subsequently, to evaluate the inflammation in a more complex intestinal model, the intestinal organoids derived from celiac disease patient biopsies (GCD-CD) and controls (CTR) were cultured. Stimulation with peptide 31-43 in the CD intestinal organoids induced NF-κβ activation, and pretreatment with LGG postbiotic could prevent it. These data showed that the LGG postbiotic can prevent the P31-43-mediated increase in inflammation in both Caco-2 cells and in intestinal organoids derived from CD patients.
Collapse
Affiliation(s)
- Francesca Furone
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| | - Claudia Bellomo
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| | - Martina Carpinelli
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| | - Martina Nicoletti
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| | | | - Majed Mordaa
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| | - Roberta Mandile
- Department of Translational Medical Sciences, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Vittoria Barone
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, Naples, Italy
| | - Merlin Nanayakkara
- Department of Translational Medical Science (Section of Paediatrics), University of Naples Federico II, Naples, Italy
| |
Collapse
|
87
|
Keathley J, White J, Reid G. The Impact of Nutrition, Physical Activity, Beneficial Microbes, and Fecal Microbiota Transplant for Improving Health. Life (Basel) 2023; 13:life13051124. [PMID: 37240769 DOI: 10.3390/life13051124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
The recognition that microbes are integral to human life has led to studies on how to manipulate them in favor of health outcomes. To date, there has been no conjoint recommendation for the intake of dietary compounds that can complement the ingested organisms in terms of promoting an improved health outcome. The aim of this review is to discuss how beneficial microbes in the form of probiotics, fermented foods, and donor feces are being used to manage health. In addition, we explore the rationale for selecting beneficial microbial strains and aligning diets to accommodate their propagation in the gut. A pilot clinical trial design is presented to examine the effects of probiotics and exercise in patients with phenylketonuria (PKU); it is the most common inborn error of amino acid metabolism, and it is a complication that requires lifelong dietary intervention. The example design is provided to illustrate the importance of using omics technology to see if the intervention elevates neuroactive biogenic amines in the plasma; increases the abundance of Eubacterium rectale, Coprococcus eutactus, Akkermansia muciniphila, or Butyricicoccus; and increases Escherichia/Shigella in the gut, all as markers of improved health. By emphasizing the combined importance of diet, microbial supplements, and the gut microbiome, we hope that future studies will better align these components, not only to improve outcomes, but also to enhance our understanding of the mechanisms.
Collapse
Affiliation(s)
- Justine Keathley
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Jessica White
- Department of Food and Nutritional Sciences, Brescia College, 1285 Western Road, London, ON N6G 1H2, Canada
| | - Gregor Reid
- Departments of Microbiology & Immunology and Surgery, The University of Western Ontario, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2, Canada
| |
Collapse
|
88
|
Al-Ishaq RK, Samuel SM, Büsselberg D. The Influence of Gut Microbial Species on Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24098118. [PMID: 37175825 PMCID: PMC10179351 DOI: 10.3390/ijms24098118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with an alarming incidence rate and a considerable burden on the patient's life and health care providers. An increase in blood glucose level and insulin resistance characterizes it. Internal and external factors such as urbanization, obesity, and genetic mutations could increase the risk of DM. Microbes in the gut influence overall health through immunity and nutrition. Recently, more studies have been conducted to evaluate and estimate the role of the gut microbiome in diabetes development, progression, and management. This review summarizes the current knowledge addressing three main bacterial species: Bifidobacterium adolescentis, Bifidobacterium bifidum, and Lactobacillus rhamnosus and their influence on diabetes and its underlying molecular mechanisms. Most studies illustrate that using those bacterial species positively reduces blood glucose levels and activates inflammatory markers. Additionally, we reported the relationship between those bacterial species and metformin, one of the commonly used antidiabetic drugs. Overall, more research is needed to understand the influence of the gut microbiome on the development of diabetes. Furthermore, more efforts are required to standardize the model used, concentration ranges, and interpretation tools to advance the field further.
Collapse
Affiliation(s)
- Raghad Khalid Al-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| |
Collapse
|
89
|
Yu Q, Qian J, Guo Y, Qian H, Yao W, Cheng Y. Applicable Strains, Processing Techniques and Health Benefits of Fermented Oat Beverages: A Review. Foods 2023; 12:1708. [PMID: 37107502 PMCID: PMC10137769 DOI: 10.3390/foods12081708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Based on the high nutrients of oat and the demand of health-conscious consumers for value-added and functional foods, fermented oat beverages have great market prospects. This review summarizes the applicable strains, processing techniques and health benefits of fermented oat beverages. Firstly, the fermentation characteristics and conditions of the applicable strains are systematically described. Secondly, the advantages of pre-treatment processes such as enzymatic hydrolysis, germination, milling and drying are summarized. Furthermore, fermented oat beverages can increase the nutrient content and reduce the content of anti-nutritional factors, thereby reducing some risk factors related to many diseases such as diabetes, high cholesterol and high blood pressure. This paper discusses the current research status of fermented oat beverages, which has academic significance for researchers interested in the application potential of oat. Future studies on fermenting oat beverages can focus on the development of special compound fermentation agents and the richness of their taste.
Collapse
Affiliation(s)
- Qian Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Jiaqin Qian
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yahui Guo
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - He Qian
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Weirong Yao
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| |
Collapse
|
90
|
Ruiz de la Bastida A, Peirotén Á, Langa S, Rodríguez-Mínguez E, Curiel JA, Arqués JL, Landete JM. Fermented soy beverages as vehicle of probiotic lactobacilli strains and source of bioactive isoflavones: A potential double functional effect. Heliyon 2023; 9:e14991. [PMID: 37095934 PMCID: PMC10121624 DOI: 10.1016/j.heliyon.2023.e14991] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 02/14/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
Soy beverages can be a source of bioactive isoflavones, with potential human health benefits. In this work, the suitability of three Lacticaseibacillus and three Bifidobacterium probiotic strains as functional starters for soy beverage fermentation were evaluated, alongside with the effect of refrigerated storage on the viability of the strains and the isoflavone composition of the fermented beverages. The three bifidobacteria strains suffered a decrease in their viability during refrigeration and only Bifidobacterium breve INIA P734 produced high concentrations of bioactive isoflavones. Meanwhile, L. rhamnosus GG and L. rhamnosus INIA P344 produced high levels of aglycones and, with L. paracasei INIA P272, maintained their viability during the refrigeration period, constituting promising starters to obtain functional soy beverages that could gather the benefits of the bioactive isoflavone aglycones and the probiotic strains. Moreover, the three lactobacilli caused an increase in the antioxidant capacity of the fermented beverages, which was maintained over the refrigerated storage.
Collapse
|
91
|
Bietto F, Scardaci R, Brovia M, Kokalari I, Barbero F, Fenoglio I, Pessione E. Food-grade titanium dioxide can affect microbiota physiology, adhesion capability, and interbacterial interactions: A study onL. rhamnosus and E. faecium. Food Chem Toxicol 2023; 176:113760. [PMID: 37028743 DOI: 10.1016/j.fct.2023.113760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Food-grade titanium dioxide (TiO2-FG) is a widespread metal oxide used in the food industries. Recently, the European Food Safety Authority concluded that TiO2-FG cannot be considered safe for consumption due to its genotoxicity; however, its effect on the gut microbiota has not yet been completely unraveled. We studied the effects of TiO2-FG (0.125 mg/mL) on Lactobacillus rhamnosus GG (LGG) and Enterococcus faecium NCIMB10415 (Ent), in particular some physiological and phenotypic traits (growth kinetics, bile salts, and ampicillin resistance) and their interactions with the host (auto-aggregation, biofilm formation, and adhesion on Caco-2/TC7 monolayers) and other gut microorganisms (antimicrobial activity towards pathogens). The results obtained revealed that TiO2-FG alters both LGG and Ent growth and lowers bile resistance (62 and 34.5%, respectively) and adhesion on Caco-2/TC7 monolayers (34.8 and 14.16%, respectively). The other outcomes were strictly species-specific: Ent showed a lower ampicillin sensitivity (14.48%) and auto-aggregation (38.1%), while LGG showed a reduced biofilm formation (37%) and antimicrobial activity towards Staphylococcus aureus (35.73%). Overall, these results suggest an adverse effect of TiO2-FG on both the endogenous and exogenously administered probiotics, contributing to the argument against using TiO2-FG as a food additive.
Collapse
Affiliation(s)
- F Bietto
- Laboratory of Microbial Biochemistry and Proteomics, Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy.
| | - R Scardaci
- Laboratory of Microbial Biochemistry and Proteomics, Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy.
| | - M Brovia
- Laboratory of Microbial Biochemistry and Proteomics, Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy.
| | - I Kokalari
- Department of Chemistry, University of Turin, via P. Giuria 7, 10125, Torino, Italy.
| | - F Barbero
- Department of Chemistry, University of Turin, via P. Giuria 7, 10125, Torino, Italy.
| | - I Fenoglio
- Department of Chemistry, University of Turin, via P. Giuria 7, 10125, Torino, Italy.
| | - E Pessione
- Laboratory of Microbial Biochemistry and Proteomics, Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina 13, 10123, Torino, Italy.
| |
Collapse
|
92
|
Kistaubayeva A, Abdulzhanova M, Zhantlessova S, Savitskaya I, Karpenyuk T, Goncharova A, Sinyavskiy Y. The Effect of Encapsulating a Prebiotic-Based Biopolymer Delivery System for Enhanced Probiotic Survival. Polymers (Basel) 2023; 15:polym15071752. [PMID: 37050363 PMCID: PMC10097185 DOI: 10.3390/polym15071752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 04/03/2023] Open
Abstract
Orally delivered probiotics must survive transit through harsh environments during gastrointestinal (GI) digestion and be delivered and released into the target site. The aim of this work was to evaluate the survivability and delivery of gel-encapsulated Lactobacillus rhamnosus GG (LGG) to the colon. New hybrid symbiotic beads alginate/prebiotic pullulan/probiotic LGG were obtained by the extrusion method. The average size of the developed beads was 3401 µm (wet), 921 µm (dry) and the bacterial titer was 109 CFU/g. The morphology of the beads was studied by a scanning electron microscope, demonstrating the structure of the bacterial cellulose shell and loading with probiotics. For the first time, we propose adding an enzymatic extract of feces to an artificial colon fluid, which mimics the total hydrolytic activity of the intestinal microbiota. The beads can be digested by fecalase with cellulase activity, indicating intestinal release. The encapsulation of LGG significantly enhanced their viability under simulated GI conditions. However, the beads, in combination with the prebiotic, provided greater protection of bacteria, enhancing their survival and even increasing cell numbers in the capsules. These data suggest the promising prospects of coencapsulation as an innovative delivery method based on the inclusion of probiotic bacteria in a symbiotic matrix.
Collapse
Affiliation(s)
- Aida Kistaubayeva
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Malika Abdulzhanova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Sirina Zhantlessova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Irina Savitskaya
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Tatyana Karpenyuk
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Alla Goncharova
- Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | | |
Collapse
|
93
|
Eliuz Tipici B, Coskunpinar E, Altunkanat D, Cagatay P, Omer B, Palanduz S, Satman I, Aral F. Lactobacillus GG is associated with mucin genes expressions in type 2 diabetes mellitus: a randomized, placebo-controlled trial. Eur J Nutr 2023:10.1007/s00394-023-03139-3. [PMID: 36997822 DOI: 10.1007/s00394-023-03139-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 03/21/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE Recent studies indicate that dysbiosis of gut microbiota and low-grade inflammation are important pathogenic determinants of type two diabetes mellitus (T2DM). The aim of this study is to investigate the effects of Lactobacillus GG on glycemic control, lipid profile, inflammatory parameters, and some gene expression levels in individuals with T2DM. METHODS In a randomized, placebo-controlled trial, 34 women, aged 30-60 years with T2DM consumed daily probiotics or placebo for 8 weeks. The probiotic group consumed 10 × 109 Cfu/day Lactobacillus rhamnosus GG ATCC 53,103 (LGG), approved by the TR Ministry of Food, Agriculture, and Livestock. Anthropometric measurements, food diary, fasting blood, and fecal samples were taken at baseline and post-treatment. RESULTS Fasting blood glucose was significantly decreased in probiotic (p = 0.049) and placebo (p = 0.028), but there was no difference between the groups. In the probiotic group, no significant difference was observed in HbA1c, fructosamine, lipid profile, and inflammatory variables compared to baseline. In this group, with LGG supplementation, mucin 2 and 3A (MUC2 and MUC3A) gene expressions increased more than ninefolds (p = 0.046 and p = 0.008, respectively) at post-treatment. Meanwhile, there was no significant change in any of the gene expressions in the placebo group. There was no significant difference in energy, protein, dietary fiber, and cholesterol intakes between placebo and probiotic groups during the study. However, daily fat intake (p = 0.003), body weight (p = 0.014), and body fat (p = 0.015) in the probiotic group were significantly decreased. CONCLUSION In this study, the effects of a single probiotic strain were investigated for 8 weeks. At the end of the study, although there was no finding that clearly reflected on the glycemic parameters of T2DM, its beneficial effects on the expression of mucin genes, which are responsible for weight loss and protection of intestinal barrier functions, cannot be denied. Further studies are needed to reveal the importance of these findings. CLINICAL TRIAL REGISTRATION ID: NCT05066152, October 4, 2021 retrospectively registered in ClinicalTrials.gov PRS web site.
Collapse
Affiliation(s)
- Beyza Eliuz Tipici
- Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Ender Coskunpinar
- Department of Medical Biology, School of Medicine, University of Health Sciences, Istanbul, Turkey
- Division of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Derya Altunkanat
- Division of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Penbe Cagatay
- Department of Biostatistics, Faculty of Medicine, Istanbul Arel University, Istanbul, Turkey
| | - Beyhan Omer
- Department of Biochemistry, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sukru Palanduz
- Division of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ilhan Satman
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ferihan Aral
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
94
|
Van den Abbeele P, Goggans M, Deyaert S, Baudot A, Van de Vliet M, Calatayud Arroyo M, Lelah M. Lacticaseibacillus rhamnosus ATCC 53103 and Limosilactobacillus reuteri ATCC 53608 Synergistically Boost Butyrate Levels upon Tributyrin Administration Ex Vivo. Int J Mol Sci 2023; 24:5859. [PMID: 36982942 PMCID: PMC10054277 DOI: 10.3390/ijms24065859] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Modulation of the gut microbiota is a trending strategy to improve health. While butyrate has been identified as a key health-related microbial metabolite, managing its supply to the host remains challenging. Therefore, this study investigated the potential to manage butyrate supply via tributyrin oil supplementation (TB; glycerol with three butyrate molecules) using the ex vivo SIFR® (Systemic Intestinal Fermentation Research) technology, a highly reproducible, in vivo predictive gut model that accurately preserves in vivo-derived microbiota and enables addressing interpersonal differences. Dosing 1 g TB/L significantly increased butyrate with 4.1 (±0.3) mM, corresponding with 83 ± 6% of the theoretical butyrate content of TB. Interestingly, co-administration of Limosilactobacillus reuteri ATCC 53608 (REU) and Lacticaseibacillus rhamnosus ATCC 53103 (LGG) markedly enhanced butyrate to levels that exceeded the theoretical butyrate content of TB (138 ± 11% for REU; 126 ± 8% for LGG). Both TB + REU and TB + LGG stimulated Coprococcus catus, a lactate-utilizing, butyrate-producing species. The stimulation of C. catus with TB + REU was remarkably consistent across the six human adults tested. It is hypothesized that LGG and REU ferment the glycerol backbone of TB to produce lactate, a precursor of butyrate. TB + REU also significantly stimulated the butyrate-producing Eubacterium rectale and Gemmiger formicilis and promoted microbial diversity. The more potent effects of REU could be due to its ability to convert glycerol to reuterin, an antimicrobial compound. Overall, both the direct butyrate release from TB and the additional butyrate production via REU/LGG-mediated cross-feeding were highly consistent. This contrasts with the large interpersonal differences in butyrate production that are often observed upon prebiotic treatment. Combining TB with LGG and especially REU is thus a promising strategy to consistently supply butyrate to the host, potentially resulting in more predictable health benefits.
Collapse
Affiliation(s)
| | - Mallory Goggans
- NutriScience Innovations, 130C Old Gate Lane, Milford, CT 06460, USA
| | - Stef Deyaert
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | - Aurélien Baudot
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
| | - Michiel Van de Vliet
- Cryptobiotix SA, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium
- Laboratory of Microbiology, Ghent University, Karel Lodewijk Ledeganckstraat 35, 9000 Ghent, Belgium
| | - Marta Calatayud Arroyo
- Institute of Agrochemistry and Food Technology (IATA), Spanish Research Council (CSIC), Carrer del Catedràtic Agustín Escardino Benlloch, 7, 46980 Valencia, Spain
| | - Michael Lelah
- NutriScience Innovations, 130C Old Gate Lane, Milford, CT 06460, USA
| |
Collapse
|
95
|
Salemi R, Vivarelli S, Ricci D, Scillato M, Santagati M, Gattuso G, Falzone L, Libra M. Lactobacillus rhamnosus GG cell-free supernatant as a novel anti-cancer adjuvant. J Transl Med 2023; 21:195. [PMID: 36918929 PMCID: PMC10015962 DOI: 10.1186/s12967-023-04036-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/04/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Gut microbiota modulation has been demonstrated to be effective in protecting patients against detrimental effects of anti-cancer therapies, as well as to improve the efficacy of certain anti-cancer treatments. Among the most characterized probiotics, Lactobacillus rhamnosus GG (LGG) is currently utilized in clinics to alleviate diarrhea, mucositis or intestinal damage which might be associated with several triggers, including Clostridium difficile infections, inflammatory gut diseases, antibiotic consumption, chemotherapy or radiation therapy. Here, we investigate whether LGG cell-free supernatant (LGG-SN) might exert anti-proliferative activity toward colon cancer and metastatic melanoma cells. Moreover, we assess the potential adjuvant effect of LGG-SN in combination with anti-cancer drugs. METHODS LGG-SN alone or in combination with either 5-Fuorouracil and Irinotecan was used to treat human colon and human melanoma cancer cell lines. Dimethylimidazol-diphenyl tetrazolium bromide assay was employed to detect cellular viability. Trypan blue staining, anti-cleaved caspase-3 and anti-total versus anti-cleaved PARP western blots, and annexin V/propidium iodide flow cytometry analyses were used to assess cell death. Flow cytometry measurement of cellular DNA content (with propidium iodide staining) together with qPCR analysis of cyclins expression were used to assess cell cycle. RESULTS We demonstrate that LGG-SN is able to selectively reduce the viability of cancer cells in a concentration-dependent way. While LGG-SN does not exert any anti-proliferative activity on control fibroblasts. In cancer cells, the reduction in viability is not associated with apoptosis induction, but with a mitotic arrest in the G2/M phase of cell cycle. Additionally, LGG-SN sensitizes cancer cells to both 5-Fluorouracil and Irinotecan, thereby showing a positive synergistic action. CONCLUSION Overall, our results suggest that LGG-SN may contain one or more bioactive molecules with anti-cancer activity which sensitize cancer cells to chemotherapeutic drugs. Thus, LGG could be proposed as an ideal candidate for ground-breaking integrated approaches to be employed in oncology, to reduce chemotherapy-related side effects and overcome resistance or relapse issues, thus ameliorating the therapeutic response in cancer patients.
Collapse
Affiliation(s)
- Rossella Salemi
- Department of Biomedical and Biotechnological Sciences, Section of General Pathology, Clinics and Oncology, University of Catania, Catania, Italy
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences, Section of General Pathology, Clinics and Oncology, University of Catania, Catania, Italy.,Department of Biomedical and Dental Sciences, Morphological and Functional Imaging, Section of Occupational Medicine, University of Messina, Messina, Italy
| | - Daria Ricci
- Department of Biomedical and Biotechnological Sciences, Section of General Pathology, Clinics and Oncology, University of Catania, Catania, Italy
| | - Marina Scillato
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences, Section of Microbiology, University of Catania, Catania, Italy
| | - Giuseppe Gattuso
- Department of Biomedical and Biotechnological Sciences, Section of General Pathology, Clinics and Oncology, University of Catania, Catania, Italy
| | - Luca Falzone
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Section of General Pathology, Clinics and Oncology, University of Catania, Catania, Italy. .,Research Center for Prevention, Diagnosis, and Treatment of Cancer, University of Catania, Catania, Italy.
| |
Collapse
|
96
|
Kell M, Lee ZC, Hernandez M, Crader M, Norwood J. A Case Report of Bacteremia Due to a Symptomatic and Rare Lactobacillus Rhamnosus Infected Splenic Hematoma and the Ultimate Treatment Model. Cureus 2023; 15:e36128. [PMID: 37065349 PMCID: PMC10100192 DOI: 10.7759/cureus.36128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 03/16/2023] Open
Abstract
We present the case of a 76-year-old male who presented to our hospital with a rare infection of Lactobacillus rhamnosus. The patient had a suspected urinary tract infection (UTI) secondary to a chronic indwelling catheter; however, when symptoms did not improve on standard therapy, blood cultures revealed the growth of L. rhamnosus. The patient was found via imaging to have a concurrent infectious splenic hematoma, and aspiration confirmed the presence of L. rhamnosus. The patient resided in an area nursing home and was a poor historian; however, it is conceivable that this infection was acquired via diet or from normal gut flora as the patient did not present on probiotic supplementation. In this case report, we present both pharmaceutical and interventional treatment strategies as well as a timeline of treatment for this rarely-seen infection.
Collapse
|
97
|
Preparation and Evaluation of a Dosage Form for Individualized Administration of Lyophilized Probiotics. Pharmaceutics 2023; 15:pharmaceutics15030910. [PMID: 36986771 PMCID: PMC10053861 DOI: 10.3390/pharmaceutics15030910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Probiotics have been used in human and veterinary medicine to increase resistance to pathogens and provide protection against external impacts for many years. Pathogens are often transmitted to humans through animal product consumption. Therefore, it is assumed that probiotics protecting animals may also protect the humans who consume them. Many tested strains of probiotic bacteria can be used for individualized therapy. The recently isolated Lactobacillus plantarum R2 Biocenol™ has proven to be preferential in aquaculture, and potential benefits in humans are expected. A simple oral dosage form should be developed to test this hypothesis by a suitable preparation method, i.e., lyophilization, allowing the bacteria to survive longer. Lyophilizates were formed from silicates (Neusilin® NS2N; US2), cellulose derivates (Avicel® PH-101), and saccharides (inulin; saccharose; modified starch® 1500). They were evaluated for their physicochemical properties (pH leachate, moisture content, water absorption, wetting time, DSC tests, densities, and flow properties); their bacterial viability was determined in conditions including relevant studies over 6 months at 4 °C and scanned under an electron microscope. Lyophilizate composed of Neusilin® NS2N and saccharose appeared to be the most advantageous in terms of viability without any significant decrease. Its physicochemical properties are also suitable for capsule encapsulation, subsequent clinical evaluation, and individualized therapy.
Collapse
|
98
|
Calvigioni M, Bertolini A, Codini S, Mazzantini D, Panattoni A, Massimino M, Celandroni F, Zucchi R, Saba A, Ghelardi E. HPLC-MS-MS quantification of short-chain fatty acids actively secreted by probiotic strains. Front Microbiol 2023; 14:1124144. [PMID: 36937254 PMCID: PMC10020375 DOI: 10.3389/fmicb.2023.1124144] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Short-chain fatty acids (SCFAs) are the main by-products of microbial fermentations occurring in the human intestine and are directly involved in the host's physiological balance. As impaired gut concentrations of acetic, propionic, and butyric acids are often associated with systemic disorders, the administration of SCFA-producing microorganisms has been suggested as attractive approach to solve symptoms related to SCFA deficiency. Methods In this research, nine probiotic strains (Bacillus clausii NR, OC, SIN, and T, Bacillus coagulans ATCC 7050, Bifidobacterium breve DSM 16604, Limosilactobacillus reuteri DSM 17938, Lacticaseibacillus rhamnosus ATCC 53103, and Saccharomyces boulardii CNCM I-745) commonly included in commercial formulations were tested for their ability to secrete SCFAs by using an improved protocol in high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS-MS). Results The developed method was highly sensitive and specific, showing excellent limits of detection and quantification of secreted SCFAs. All tested microorganisms were shown to secrete acetic acid, with only B. clausii and S. boulardii additionally able to produce propionic and butyric acids. Quantitative differences in the secretion of SCFAs were also evidenced. Discussion The experimental approach described in this study may contribute to the characterization of probiotics as SCFA-producing organisms, a crucial stage toward their application to improve SCFA deficiency.
Collapse
Affiliation(s)
- Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Andrea Bertolini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Simone Codini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Adelaide Panattoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mariacristina Massimino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Riccardo Zucchi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Saba
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Research Center Nutraceuticals and Food for Health–Nutrafood, University of Pisa, Pisa, Italy
| |
Collapse
|
99
|
Niu Z, Zou M, Bei T, Zhang N, Li D, Wang M, Li C, Tian H. Effect of fructooligosaccharides on the colonization of Lactobacillus rhamnosus AS 1.2466T in the gut of mice. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.07.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
100
|
Zangl I, Beyer R, Gattesco A, Labuda R, Pap IJ, Strauss J, Schüller C. Limosilactobacillus fermentum Limits Candida glabrata Growth by Ergosterol Depletion. Microbiol Spectr 2023; 11:e0332622. [PMID: 36802215 PMCID: PMC10100998 DOI: 10.1128/spectrum.03326-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/27/2023] [Indexed: 02/23/2023] Open
Abstract
Candida glabrata is a human-associated opportunistic fungal pathogen. It shares its niche with Lactobacillus spp. in the gastrointestinal and vaginal tract. In fact, Lactobacillus species are thought to competitively prevent Candida overgrowth. We investigated the molecular aspects of this antifungal effect by analyzing the interaction of C. glabrata strains with Limosilactobacillus fermentum. From a collection of clinical C. glabrata isolates, we identified strains with different sensitivities to L. fermentum in coculture. We analyzed the variation of their expression pattern to isolate the specific response to L. fermentum. C. glabrata-L. fermentum coculture induced genes associated with ergosterol biosynthesis, weak acid stress, and drug/chemical stress. L. fermentum coculture depleted C. glabrata ergosterol. The reduction of ergosterol was dependent on the Lactobacillus species, even in coculture with different Candida species. We found a similar ergosterol-depleting effect with other lactobacillus strains (Lactobacillus crispatus and Lactobacillus rhamosus) on Candida albicans, Candida tropicalis, and Candida krusei. The addition of ergosterol improved C. glabrata growth in the coculture. Blocking ergosterol synthesis with fluconazole increased the susceptibility against L. fermentum, which was again mitigated by the addition of ergosterol. In accordance, a C. glabrata Δerg11 mutant, defective in ergosterol biosynthesis, was highly sensitive to L. fermentum. In conclusion, our analysis indicates an unexpected direct function of ergosterol for C. glabrata proliferation in coculture with L. fermentum. IMPORTANCE The yeast Candida glabrata, an opportunistic fungal pathogen, and the bacterium Limosilactobacillus fermentum both inhabit the human gastrointestinal and vaginal tract. Lactobacillus species, belonging to the healthy human microbiome, are thought to prevent C. glabrata infections. We investigated the antifungal effect of Limosilactobacillus fermentum on C. glabrata strains quantitively in vitro. The interaction between C. glabrata and L. fermentum evokes an upregulation of genes required for the synthesis of ergosterol, a sterol constituent of the fungal plasma membrane. We found a dramatic reduction of ergosterol in C. glabrata when it was exposed to L. fermentum. This effect extended to other Candida species and other Lactobacillus species. Furthermore, fungal growth was efficiently suppressed by a combination of L. fermentum and fluconazole, an antifungal drug which inhibits ergosterol synthesis. Thus, fungal ergosterol is a key metabolite for the suppression of C. glabrata by L. fermentum.
Collapse
Affiliation(s)
- Isabella Zangl
- University of Natural Resources and Life Sciences, Vienna, Institute of Microbial Genetics, Tulln, Austria
| | - Reinhard Beyer
- University of Natural Resources and Life Sciences, Vienna, Institute of Microbial Genetics, Tulln, Austria
| | - Arianna Gattesco
- University of Natural Resources and Life Sciences, Vienna, Institute of Microbial Genetics, Tulln, Austria
| | - Roman Labuda
- Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine Vienna, Vienna, Austria
- Bioactive Microbial Metabolites, University of Natural Resources and Life Sciences, Vienna (BOKU), Institute of Microbial Genetics, Tulln, Austria
| | - Ildiko-Julia Pap
- University Hospital of St. Pölten, Institute for Hygiene and Microbiology, St. Pölten, Austria
| | - Joseph Strauss
- University of Natural Resources and Life Sciences, Vienna, Institute of Microbial Genetics, Tulln, Austria
- Bioactive Microbial Metabolites, University of Natural Resources and Life Sciences, Vienna (BOKU), Institute of Microbial Genetics, Tulln, Austria
| | - Christoph Schüller
- University of Natural Resources and Life Sciences, Vienna, Institute of Microbial Genetics, Tulln, Austria
- Core Facility Bioactive Molecules: Screening and Analysis, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|