1
|
Xu Y, Gao Z, Liu J, Yang Q, Xu S. Role of gut microbiome in suppression of cancers. Gut Microbes 2025; 17:2495183. [PMID: 40254597 PMCID: PMC12013426 DOI: 10.1080/19490976.2025.2495183] [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: 12/14/2024] [Revised: 03/23/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025] Open
Abstract
The pathogenesis of cancer is closely related to the disruption of homeostasis in the human body. The gut microbiome plays crucial roles in maintaining the homeostasis of its host throughout lifespan. In recent years, a large number of studies have shown that dysbiosis of the gut microbiome is involved in the entire process of cancer initiation, development, and prognosis by influencing the host immune system and metabolism. Some specific intestinal bacteria promote the occurrence and development of cancers under certain conditions. Conversely, some other specific intestinal bacteria suppress the oncogenesis and progression of cancers, including inhibiting the occurrence of cancers, delaying the progression of cancers and boosting the therapeutic effect on cancers. The promoting effects of the gut microbiome on cancers have been comprehensively discussed in the previous review. This article will review the latest advances in the roles and mechanisms of gut microbiome in cancer suppression, providing a new perspective for developing strategies of cancer prevention and treatment.
Collapse
Affiliation(s)
- Yao Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| | - Zhaoyu Gao
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| | - Jiaying Liu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qianqian Yang
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Shunjiang Xu
- Central Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, P. R. China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, P. R. China
- Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, P. R. China
| |
Collapse
|
2
|
Côco LZ, de Souza Belisário E, Vasquez EC, Pereira TMC, Aires R, Campagnaro BP. Probiotics: a promising future in the treatment of ulcerative colitis? Pharmacol Rep 2025; 77:645-657. [PMID: 40214948 DOI: 10.1007/s43440-025-00724-7] [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: 01/29/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 05/13/2025]
Abstract
Ulcerative colitis is an idiopathic and chronic inflammatory bowel disease, characterized by inflammation of the mucosa of the colon and rectum. Clinical manifestations commonly include abdominal pain, diarrhea (with or without hematochezia), and weight loss. The pathogenesis of ulcerative colitis is multifactorial, involving a combination of genetic predispositions and lifestyle factors. High consumption of processed food, sedentary habits, alcohol intake, and stress are among the lifestyle factors implicated in disease onset and progression. Current treatment strategies focus on managing symptoms and inducing remission, however, the chronic nature of the disease, along with the adverse effects of conventional therapies, often compromises patient's quality of life. Therefore, exploring alternative therapies that can prolong remission and reduce symptom burden is important. Experimental evidence suggests that probiotics may extend remission duration in ulcerative colitis. Moreover, probiotics exhibit efficacy in amelioration clinical symptoms by reducing inflammation markers, preserving, and restoring intestinal epithelial. This review explores the advantages of the administration of probiotics in the treatment of ulcerative colitis, elucidating their mechanism of action.
Collapse
Affiliation(s)
- Larissa Zambom Côco
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Eduarda de Souza Belisário
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Elisardo Corral Vasquez
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Thiago Melo Costa Pereira
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Rafaela Aires
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Bianca Prandi Campagnaro
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil.
| |
Collapse
|
3
|
Inokuma K, Sasaki D, Shintani T, Inoue J, Oyama K, Noda Y, Maeda T, Yamada R, Matsuki Y, Kodama Y, Kondo A. Combination of probiotics enhancing butyrogenesis in colonic microbiota model of patients with ulcerative colitis. Appl Microbiol Biotechnol 2025; 109:117. [PMID: 40347262 PMCID: PMC12065738 DOI: 10.1007/s00253-025-13424-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 05/12/2025]
Abstract
Administering beneficial bacteria as probiotics to restore the intestinal microbiota and its metabolic functions, such as butyrogenesis, is a promising treatment strategy in ulcerative colitis (UC). This study aimed to investigate the effect of a combination of probiotics, consisting of the lactic acid bacterium Weizmannia coagulans SANK70258 and the lactate-utilizing butyrate-producing bacteria Anaerostipes caccae or Clostridium butyricum, on the colonic environment using an in vitro colonic microbiota culture model with fecal inoculums from seven patients with UC. Co-inoculated W. coagulans and A. caccae neither inhibited each other's growth nor significantly affected the relative abundance of other bacterial species; however, the growth of W. coagulans was significantly inhibited when co-inoculated with C. butyricum. The relative abundance of pro-inflammatory bacteria (Escherichia sp. and unclassified Enterobacteriaceae) and Bifidobacterium spp. significantly decreased in W. coagulans-C. butyricum co-inoculated cultures. Inoculation with any of the probiotics alone did not increase butyrate production, whereas co-inoculation of W. coagulans with A. caccae or C. butyricum significantly increased the butyrate levels. Overall, the results suggested that W. coagulans and lactate-utilizing butyrate-producing bacteria in combination have synergistic effects through cross-feeding and can effectively restore butyrogenesis in the colonic environment of patients with UC. KEY POINTS: • Effects of probiotics were evaluated using in vitro microbiota model of UC colon. • W. coagulans and lactate-utilizing butyrate producers have synergistic effects. • Co-inoculation of W. coagulans with A. caccae or C. butyricum enhanced butyrogenesis.
Collapse
Affiliation(s)
- Kentaro Inokuma
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Tomoya Shintani
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Jun Inoue
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Katsuaki Oyama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Yuta Noda
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Takayuki Maeda
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Ryouichi Yamada
- Science & Innovation Center, Mitsubishi Chemical Corporation, Yokohama, Kanagawa, 227-8502, Japan
| | - Yasushi Matsuki
- Strategic Planning Office, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan
| | - Yuzo Kodama
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-Cho, Nada-Ku, Kobe, 657-8501, Japan.
- Biomass Engineering Program, RIKEN, 1-7-22 Suehiro-Cho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
4
|
El-Son MAM, Elbahnaswy S, Khormi MA, Aborasain AM, Abdelhaffez HH, Zahran E. Harnessing the fish gut microbiome and immune system to enhance disease resistance in aquaculture. FISH & SHELLFISH IMMUNOLOGY 2025; 163:110394. [PMID: 40350102 DOI: 10.1016/j.fsi.2025.110394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
The increasing global reliance on aquaculture is challenged by disease outbreaks, exacerbated by antibiotic resistance, and environmental stressors. Traditional strategies, such as antibiotic treatments and chemical interventions, are becoming less effective, necessitating a shift toward microbiota-based disease control. The fish gut microbiome is a key determinant of immune homeostasis and pathogen resistance. However, previous reviews lack integration of microbiome engineering, machine learning, and next-generation sequencing in fish health strategies. This review encompasses recent advancements in microbiome research, including dietary strategies such as prebiotics, probiotics, synbiotics, and phytogenic feed additives. It synthesizes the latest metagenomic insights, microbiota modulation techniques, and AI-driven disease prediction models. It presents a novel conceptual framework for disease control using microbiome-based approaches in aquaculture. Additionally, we explore emerging methodologies, including microbiota transplantation and synthetic probiotics, to develop precision microbiome interventions. By bridging existing knowledge gaps, this review provides actionable insights into sustainable aquaculture practices through microbiome-driven disease resistance.
Collapse
Affiliation(s)
- Mai A M El-Son
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Samia Elbahnaswy
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohsen A Khormi
- Department of Biology, College of Science, Jazan University, P.O. Box 114, Jazan 45142, Kingdom of Saudi Arabia
| | - Ali M Aborasain
- Department of Biology, College of Science, Jazan University, P.O. Box 114, Jazan 45142, Kingdom of Saudi Arabia
| | - Hanan H Abdelhaffez
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut 20 71526, Egypt
| | - Eman Zahran
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
5
|
Wang Y, Chen S, Ye B, Yang Z, Liu Y, Lou G, Zhou C, Zheng M. Acalabrutinib alleviates metabolic dysfunction-associated steatotic liver disease by regulating bile acid metabolism. Int J Biochem Cell Biol 2025; 185:106786. [PMID: 40306481 DOI: 10.1016/j.biocel.2025.106786] [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: 04/23/2024] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a global epidemic of chronic liver disease currently lacking effective treatment. Evaluating the therapeutic effects of existing drugs on MASLD is a time and cost-effective strategy. Bruton's tyrosine kinase (BTK) is an inflammatory signaling molecule playing an important role in the progression of MASLD. Aclabrutinib, a BTK inhibitor approved for treating mantle-cell lymphoma and chronic lymphocytic leukemia, has not been investigated for its potential to treat MASLD. This study examined the therapeutic effects and mechanisms of aclabrutinib on MASLD using a high-fat diet-induced mouse model. Results demonstrated significant alleviation of pathological parameters associated with MASLD upon administration of aclabrutinib. TSE PhenoMaster results revealed that aclabrutinib increased energy expenditure in mice. Furthermore, aclabrutinib upregulated the expression of genes associated with thermogenesis and lipolysis in adipose tissues. Additionally, it inhibited the transcription of genes related to lipid absorption in the small intestine and liver, while increasing the expression of hormone-sensitive lipase, hepatic nuclear factor 4 alpha and fibroblast growth factor 21 in the liver. Further analysis indicated that aclabrutinib promoted the alternative pathway of bile acid synthesis while restoring gut microbiota homeostasis. The altered bile acid profiles upregulated G protein-coupled bile acid receptor 1 expression in adipose tissues as well as vitamin D receptor expression in liver and small intestine. Our findings suggest that by regulating bile acid metabolism and gut microbiota, aclabrutinib may promote thermogenesis and lipolysis, thereby alleviating MASLD. This study provides novel insights into clinical applications targeting BTK for treating MASLD.
Collapse
Affiliation(s)
- Yanbo Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shiwei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Bingjue Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Zhenggang Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Guohua Lou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Cheng Zhou
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Min Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
6
|
Kang A, Eor JY, Lee J, Kwak MJ, Lee DJ, Seo E, Lee WJ, Son SH, Song M, Kim JM, Kim HW, Yang J, Oh S, Kim Y. Lacticaseibacillus casei IDCC 3451 alleviates cognitive and behavioral functions by reshaping the gut microbiome and regulating intestinal barrier integrity in chronic stress animal models. Curr Res Food Sci 2025; 10:101051. [PMID: 40290371 PMCID: PMC12023876 DOI: 10.1016/j.crfs.2025.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Lacticaseibacillus casei IDCC 3451 (3451) was evaluated for its effects on the gut-brain axis using Caenorhabditis elegans (C. elegans) and mouse models of stress and inflammation. In C. elegans, 3451 extended lifespans by 25 %, improved motility, and chemotaxis, enhanced survival under pathogen challenge, and reduced amyloid beta accumulation by 42 %. Transcriptomic profiling revealed upregulation of genes involved in neurotransmitter signaling and serine/threonine pathways. In the unpredictable chronic mild stress (UCMS) mouse model, 3451 administration increased the time spent in the center of the open field by 65 % and reduced immobility in the forced swim test by 32 %, indicating anxiolytic and antidepressant effects. Serum levels of aspartate aminotransferase (AST) and gamma-glutamyl transferase (GGT) were decreased by 18 % and 24 %, respectively. Additionally, 3451 restored the expressions of 5HT1AR, GABAR, and tight junction proteins, including ZO-1 and Claudin1. Metabolomic analysis showed increased glycine and decreased palmitic acid levels, associated with an increased abundance of Ruminococcus and Akkermansia. In the dextran sulfate sodium (DSS)-induced colitis model, 3451 reduced the disease activity index by 36 %, improved colon histology, increased goblet cell preservation, and upregulated ZO-1 and IL-10 expression. Threonine levels were also increased and correlated with a higher abundance of Coprococcus. These findings demonstrate that 3451 improved behavioral and intestinal outcomes through coordinated modulation of host signaling, metabolite production, and gut microbial composition, highlighting its therapeutic potential for managing IBD and neurobehavioral disorders.
Collapse
Affiliation(s)
- Anna Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Junbeom Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Daniel Junpyo Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Eunsol Seo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Woong Ji Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Seon-hui Son
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| | - Minho Song
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, 34134, South Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Gyeonggi-do, South Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul, 05006, South Korea
| | - Jungwoo Yang
- Department of Microbiology, College of Medicine, Dongguk University, Gyeongju, 38066, South Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, 55069, South Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, South Korea
| |
Collapse
|
7
|
Mahmoud RES, Ateya A, Gadalla H, Alharbi HM, Alwutayd KM, Embaby EM. Growth Performance, Immuno-Oxidant Status, Intestinal Health, Gene Expression, and Histomorphology of Growing Quails Fed Diets Supplemented with Essential Oils and Probiotics. Vet Sci 2025; 12:341. [PMID: 40284844 PMCID: PMC12031328 DOI: 10.3390/vetsci12040341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
This study aimed to evaluate how natural dietary supplements, including essential oils (EOs) and probiotics, influence the growth performance, carcass traits, serum components, gut function, gene expression, and jejunal histomorphology of growing quails. A total of 240 unsexed 7-day-old growing Japanese quails were randomly assigned to four experimental groups (n = 60 per group), with each group further divided into six replicates (10 quails per replicate). The control group (S0) received a basal diet without incorporating any additives, while the experimental groups were supplemented with (i) essential oils (S1); (ii) probiotics (S2); or (iii) a mixture of EOs and probiotics (S3) at a level of 1.5 kg/ton and 0.55 g per kg diet, respectively, and the ratio of the mixture of EOs and probiotics was approximately 2.73:1. The results showed that, from 7 to 35 days of age, S3 quails showed increased growth performance, carcass weight, and serum total protein with a decreased lipid profile, outperforming the individual supplementation of either additive (p < 0.05). Importantly, EOs or probiotics enhanced immune-antioxidant status in growing quails, particularly those who were fed both EOs and probiotics, showing significantly increased levels of the serum immune parameters IgY and IgM as well as boosting T-AOC, SOD, and GPx levels when MDA content was lowered compared to S0 quails (p < 0.05). Additionally, in quails fed a mixture of EOs and probiotics, the primary pro-inflammatory factors TNF-α, IL-1β, and IL-6 were downregulated, and the anti-inflammatory factors TGF-β and IL-10 were elevated compared to the S0 group (p < 0.05). In this context, there was a notable increase in growth (IGF-I, myogenin, and AvUCP), immunological (IL-2, IL-4, IL-6, and AVBD), antioxidant (SOD, CAT, GPx, and ATOX1), and intestinal absorption (VEGF, MUC2, GLUT2, calbindin, and FABP6) markers in quails supplemented with EOs and/or probiotics when compared to the control group (p < 0.05). The combination of EOs and probiotics had the most noticeable impact on the markers' expression patterns compared to either additive alone (p < 0.05). Consistent with our results, quails given both EOs and probiotics showed significantly greater villi in terms of height and width when compared to the control group in intestinal histomorphology, the primary measure of intestinal wellness. In conclusion, quail diets could benefit from the use of EOs or probiotics as natural growth promoters since they improve growth performance, blood parameters associated with protein and lipid profiles, immune-antioxidant status and inflammation, and marker gene expression profiles of growth, immune, antioxidant, and intestinal absorption.
Collapse
Affiliation(s)
- Rania El Sayed Mahmoud
- Department of Nutrition & Clinical Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Ahmed Ateya
- Department of Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Hossam Gadalla
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Hanan M. Alharbi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (H.M.A.); (K.M.A.)
| | - Khairiah M. Alwutayd
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (H.M.A.); (K.M.A.)
| | - Eman M. Embaby
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| |
Collapse
|
8
|
Huang F, Yang N, Zhang Q, Luo C, Wang J, Yang Y, Yue B, Chen P, Zhang X. Marine-Derived Enterococcus faecalis HY0110 as a Next-Generation Functional Food Probiotic: Comprehensive In Vitro and In Vivo Bioactivity Evaluation and Synergistic Fermentation of Periplaneta americana Extract Powder. Foods 2025; 14:1181. [PMID: 40238337 PMCID: PMC11988638 DOI: 10.3390/foods14071181] [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: 03/11/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Addressing the escalating global burdens of inflammatory bowel disease and antimicrobial resistance demanded innovative food-based approaches to fortify gut health and suppress pathogens. We introduced a novel edible probiotic, Enterococcus faecalis HY0110, isolated from marine Thunnus thynnus. Through comprehensive in vitro, in vivo, and metabolomic analyses, we demonstrated its superior antibacterial effects compared to Lactobacillus rhamnosus GG, along with significantly enhanced antioxidant and free-radical scavenging capacities. Notably, elevated acetic acid production strongly correlated with its antimicrobial efficacy (R ≥ 0.999). HY0110 also exerted antiproliferative effects on HT-29 colorectal cancer cells by attenuating β-catenin and BCL-2 expression while upregulating pro-apoptotic markers P62 and c-PARP. In a DSS-induced colitis model, HY0110 alleviated inflammation, restored gut microbial homeostasis, and enhanced deterministic processes in community assembly dynamics. Furthermore, fermenting Periplaneta americana powder with HY0110 triggered extensive metabolic remodeling, notably a 668.73-fold rise in astragaloside A, plus increases in L-Leucyl-L-Alanine, S-lactoylglutathione, and 16,16-dimethyl prostaglandin A1. These shifts diminished harmful components and amplified essential amino acids and peptides to bolster immune modulation, redox balance, and anti-inflammatory responses. This work established a transformative paradigm for utilizing marine probiotics and novel entomological substrates in functional foods, presenting strategic pathways for precision nutrition and inflammatory disease management.
Collapse
Affiliation(s)
- Feiyun Huang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Nan Yang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610041, China;
| | - Qingqing Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Cuiling Luo
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Jingheng Wang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Yu Yang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Bisong Yue
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
| | - Peng Chen
- MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu 610065, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-Resources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610065, China; (F.H.); (Q.Z.); (C.L.); (J.W.); (Y.Y.); (B.Y.)
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu 610065, China
| |
Collapse
|
9
|
Liao Y, Wu S, Zhou G, Mei S, Ou B, Wen M, Yang Y, Wen G. Probiotic Bacillus cereus regulates metabolic disorders and activates the cholic acid-FXR axis to alleviate DSS-induced colitis. J Proteomics 2025; 312:105360. [PMID: 39631667 DOI: 10.1016/j.jprot.2024.105360] [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: 07/08/2024] [Revised: 11/30/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Inflammatory bowel disease is characterized by severe imbalance of intestinal flora and metabolic disorders. Recent studies have demonstrated that probiotics can effectively alleviate inflammatory bowel disease by restoring the intestinal flora structure and modulating the immune response. However, the role of probiotics in regulating intestinal metabolism disorders is still unclear. This study explores the role of probiotic B. cereus in alleviating DSS-induced colitis. The findings indicated probiotic B. cereus treatment mitigated tissue damage and apoptosis during inflammation. Metabolome and transcriptome analysis revealed B. cereus activated the cholic acid-FXR axis by increasing cholic acid levels, which promoted the gene expression level of NF-κB inhibitor α, reduced the IL-1β, IL-6, IL-18 and TNF-α concentrations. Furthermore, it effectively mitigated the DSS-induced disruption of bile acid metabolism, arginine metabolism, and linoleic acid metabolism. This study explores the effect and mechanisms of probiotic B. cereus on alleviating DSS-induced colitis. It aims to provide a theoretical basis for microbial therapy in inflammatory bowel disease. SIGNIFICANCE: This study used metabolome and transcriptome to reveal the roles and mechanisms, which probiotic Bacillus cereus modulates metabolic disorders and alleviate DSS-induced colitis. We identified the cholic acid-FXR axis as an important target for alleviating DSS-induced colitis. These findings provide new insights into microbial treatment strategies for IBD.
Collapse
Affiliation(s)
- Yixiao Liao
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China
| | - Shihui Wu
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China
| | - Guixian Zhou
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China
| | - Shihui Mei
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China
| | - Bingmin Ou
- School of Life Sciences, Zhaoqing University, Zhaoqing 526000, China
| | - Ming Wen
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China; Engineering Research Center of Animal Biological Products, Guiyang 550025, China
| | - Ying Yang
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China; Engineering Research Center of Animal Biological Products, Guiyang 550025, China.
| | - Guilan Wen
- College of Animal Science, Guizhou University, Guiyang 550025, China; Institute of Animal Diseases, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
10
|
Hassib L, Kanashiro A, Pedrazzi JFC, Vercesi BF, Higa S, Arruda Í, Soares Y, de Jesus de Souza A, Jordão AA, Guimarães FS, Ferreira FR. Should we consider microbiota-based interventions as a novel therapeutic strategy for schizophrenia? A systematic review and meta-analysis. Brain Behav Immun Health 2025; 43:100923. [PMID: 39839986 PMCID: PMC11745983 DOI: 10.1016/j.bbih.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
Schizophrenia is a chronic psychiatric disorder characterized by a variety of symptoms broadly categorized into positive, negative, and cognitive domains. Its etiology is multifactorial, involving a complex interplay of genetic, neurobiological, and environmental factors, and its neurobiology is associated with abnormalities in different neurotransmitter systems. Due to this multifactorial etiology and neurobiology, leading to a wide heterogeneity of symptoms and clinical presentations, current antipsychotic treatments face challenges, underscoring the need for novel therapeutic approaches. Recent studies have revealed differences in the gut microbiome of individuals with schizophrenia compared to healthy controls, establishing an intricate link between this disorder and gastrointestinal health, and suggesting that microbiota-targeted interventions could help alleviate clinical symptoms. Therefore, this meta-analysis investigates whether gut microbiota manipulation can ameliorate psychotic outcomes in patients with schizophrenia receiving pharmacological treatment. Nine studies (n = 417 participants) were selected from 81 records, comprising seven randomized controlled trials and two open-label studies, all with a low risk of bias, included in this systematic review and meta-analysis. The overall combined effect size indicated significant symptom improvement following microbiota treatment (Hedges' g = 0.48, 95% CI = 0.09 to 0.88, p = 0.004, I2 = 62.35%). However, according to Hedges' g criteria, the effect size was small (approaching moderate), and study heterogeneity was moderate based on I2 criteria. This review also discusses clinical and preclinical studies to elucidate the neural, immune, and metabolic pathways by which microbiota manipulation, particularly with Lactobacillus and Bifidobacterium genera, may exert beneficial effects on schizophrenia symptoms via the gut-brain axis. Finally, we address the main confounding factors identified in our systematic review, highlight key limitations, and offer recommendations to guide future high-quality trials with larger participant cohorts to explore microbiome-based therapies as a primary or adjunctive treatment for schizophrenia.
Collapse
Affiliation(s)
- Lucas Hassib
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Alexandre Kanashiro
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Bárbara Ferreira Vercesi
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sayuri Higa
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Íris Arruda
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Yago Soares
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Adriana de Jesus de Souza
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alceu Afonso Jordão
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | | |
Collapse
|
11
|
Du S, Sun R, Wang M, Fang Y, Wu Y, Yuan B, Jin Y. Synergistic effect of inulin hydrogels on multi-strain probiotics for prevention of ionizing radiation-induced injury. Int J Biol Macromol 2025; 287:138497. [PMID: 39647719 DOI: 10.1016/j.ijbiomac.2024.138497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/20/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
Prebiotics and probiotics are applied against multiple diseases including ionizing radiation-induced injury but their functions are not revealed enough. Here, we used a prebiotic, inulin hydrogels (IGs) to load multi-strain probiotics (MSPs) for protecting them from the gastrointestinal environment and improving their colonization in the gut; more importantly, they showed the synergistic effect against ionizing radiation-induced injury. Probiotics were embedded in a great number of channels of the IGs and used IGs as food. The MSP was composed of Clostridium butyricum (Cb), Bifidobacterium adolescentis (Ba), and Akkermansia muciniphila (Akk), which separately mainly produced butyl acid, acetic acid and lactic acid, and stimulated mucin proteins. Although the MSP showed higher effect against mouse radiation enteritis than the single probiotics and the similar effect to IGs, the IG/MSP-based synbiotic had the highest protection and improved many factors close to the normal levels, including animal physical activity, enteric barrier function, occludin and ZO-1 expressions, injury extension, the levels of pro-inflammatory factors (IL-6, TNF-α), gut microbiota, and short-chained fatty acids. Moreover, the synbiotic had strong protection against whole-body irradiation with high blood cell numbers, hemopoietic system recovery, and high levels of IL-3 and IL-10. IGs greatly synergized probiotics against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Shumin Du
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Rui Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Minting Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yubao Fang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yanping Wu
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yiguang Jin
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
12
|
Zhang T, Li X, Li J, Sun F, Duan L. Gut microbiome-targeted therapies as adjuvant treatments in inflammatory bowel diseases: a systematic review and network meta-analysis. J Gastroenterol Hepatol 2025; 40:78-88. [PMID: 39482823 DOI: 10.1111/jgh.16795] [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: 06/17/2024] [Revised: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND AND AIM Gut microbiome-targeted therapies (MTTs), including prebiotics, probiotics, synbiotics, and fecal microbiota transplantation (FMT), have been widely used in inflammatory bowel diseases (IBD), but the best MTTs has not yet been confirmed. We performed a network meta-analysis (NMA) to examine this in ulcerative colitis (UC) and Crohn's disease (CD). METHODS We searched for randomized controlled trials (RCTs) on the efficacy and safety of MTTs as adjuvant therapies for IBD until December 10, 2023. Data were pooled using a random effects model, with efficacy reported as pooled relative risks with 95% CIs, and interventions ranked according to means of surfaces under cumulative ranking values. RESULTS Thirty-eight RCTs met the inclusion criteria. Firstly, we compared the efficacy of MTTs in IBD patients. Only FMT and probiotics were superior to placebo in all outcomes, but FMT ranked best in improving clinical response rate and clinical and endoscopic remission rate, and probiotics ranked second in reducing clinical relapse rate showed significant efficacy, while prebiotics ranked first showed nonsignificant efficacy. Subsequently, we conducted NMA for specific MTT formulations in UC and CD separately, which revealed that FMT, especially combined FMT via colonoscopy and enema, showed significant efficacy and was superior in improving clinical response and remission rate of active UC patients. As for endoscopic remission and clinical relapse, multistrain probiotics based on specific genera of Lactobacillus and Bifidobacterium showed significant efficacy and ranked best in UC. In CD, we found that no MTTs were significantly better than placebo, but synbiotics comprising Bifidobacterium and fructo-oligosaccharide/inulin mix and Saccharomyces ranked best in improving clinical remission and reducing clinical relapse, respectively. Moreover, FMT was safe in both UC and CD. CONCLUSIONS FMT and multistrain probiotics showed superior efficacy in UC. However, the efficacy of MTTs varies among different IBD subtypes and disease stages; thus, the personalized treatment strategies of MTTs are necessary.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiaoang Li
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Jun Li
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Feng Sun
- China Center for Evidence Based Medical and Clinical Research, Peking University, Beijing, China
- Institute of Public Health, Peking University, Beijing, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
13
|
Joung JY, Choi K, Lee JH, Oh NS. Protective Potential of Limosilactobacillus fermentum Strains and Their Mixture on Inflammatory Bowel Disease via Regulating Gut Microbiota in Mice. J Microbiol Biotechnol 2024; 35:e2410009. [PMID: 39849930 PMCID: PMC11813365 DOI: 10.4014/jmb.2410.10009] [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: 10/12/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 01/25/2025]
Abstract
The aim of this study is to investigate the protective potential of Limosilactobacillus fermentum IM57, IR51, and IR62 strains, isolated from infant feces, and their mixture against inflammatory bowel disease (IBD). The strains exhibited robust antioxidant activities and anti-inflammatory properties in RAW 264.7 cells. Subsequently, the potential protective effects of each of these three strains, along with their mixture, were evaluated in a murine colitis model induced by dextran sodium sulfate (DSS). Noteworthy improvements in physiological parameters such as body weight, disease activity index, and colon length were observed in mice treated with the mixture followed by IR62. Additionally, administration of each strain and the mixture mitigated DSS-induced changes in gut microbiota composition with increased abundance of Lactobacillus, Bifidobacterium, Ruminococcus, and Muribaculum, compared to DSS-treated mice. Interestingly, the abundance of Muribaculum increased approximately 2.4-fold after administration of the mixture compared to before administration. Additionally, the concentration of short-chain fatty acids (SCFAs) was significantly reduced in DSS-treated group compared to the control group, while the mixture treatment group had the highest concentration of SCFAs. Furthermore, due to these changes in microbiota and the leading metabolites induced by treatment of the mixture, DSS-induced dysregulation of inflammationand barrier function-related mRNA expressions was significantly inhibited in the group fed with the mixture. Consequently, this study indicates that the multi-strain mixture of L. fermentum strains may play a crucial role in modulating gut microbiota, thereby alleviating IBD through the synergistic effect of the individual effects of the three strains.
Collapse
Affiliation(s)
- Jae Yeon Joung
- Department of Food and Animal Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Kayoung Choi
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
| | - Ju-Hoon Lee
- Department of Food and Animal Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Nam Su Oh
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
| |
Collapse
|
14
|
Zheng W, Ning K, Shi C, Zhou YF, Meng Y, Pan T, Chen Y, Xie Q, Xiang H. Xiaobugan decoction prevents CCl 4-induced acute liver injury by modulating gut microbiota and hepatic metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156113. [PMID: 39388924 DOI: 10.1016/j.phymed.2024.156113] [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: 01/29/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND The liver plays a crucial role in detoxification and metabolism. When its capacity to metabolize foreign substances is exceeded, it can lead to acute liver injury (ALI). Therefore, preventing liver disease and maintaining daily liver health are of utmost importance. Xiaobugan Decoction (XBGD), a traditional Chinese medicine (TCM) formula, is recorded in 'Fuxingjue', is used in folk practice to promote liver health and regulate respiration. However, the hepatoprotective mechanisms of XBGD remained unclear. PURPOSE We investigated the prophylactic and hepatoprotective effects of XBGD and explored its related molecular mechanisms using a mouse model of carbon tetrachloride (CCl4)-induced ALI. STUDY DESIGN AND METHODS XBGD composition was determined using analytical methods, and the main compounds were identified using ultra-high-performance liquid chromatography coupled with Q-Exactive focus mass spectrum (UHPLC-QE-MS) and high-performance liquid chromatography (HPLC). A CCl4-induced L02 cell injury model was employed to explore the protective effects of XBGD on liver cells, and a CCl4-induced ALI mouse model was used to investigate the hepatoprotective effects of XBGD. RESULTS Cellular experiments demonstrated that XBGD had a protective function against L02 cell damage by increasing cell viability, restoring alanine aminotransferase (ALT), aspartate aminotransferase (AST), and superoxide dismutase (SOD) levels, reducing malondialdehyde (MDA) content, and improving mitochondrial membrane potential (ΔΨm). In the mouse ALI model, XBGD prevented ALI by reducing ALT, AST, and alkaline phosphatase (ALP) levels and inhibiting oxidative stress. Quantitative real-time polymerase chain reaction (qPCR), immumohistochemical staining and western blotting results revealed that XBGD exerted hepatoprotective effects by reducing inflammatory responses and inhibiting cell apoptosis. Furthermore, 1H-NMR metabolomics indicated that XBGD regulates hepatic and intestinal metabolism, whereas 16S rDNA sequencing demonstrated the regulatory effects of XBGD on the gut microbiota. Correlation analysis highlighted the close relationship among gut microbiota, metabolites, and ALI indicators. CONCLUSIONS XBGD is a promising TCM for the prevention of CCl4-induced ALI via regulation of microbiota and metabolism. This study provides a new perspective on the development of hepatoprotective measures and the prevention of liver disease in daily life.
Collapse
Affiliation(s)
- Weiwei Zheng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Ke Ning
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Chao Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Yong-Fei Zhou
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Yao Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Tong Pan
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Yue Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China
| | - Qiuhong Xie
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China; National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun Jilin 130012, PR China; Institute of Changbai Mountain Resource and Health, Jilin University, Fusong Jilin 134504, PR China.
| | - Hongyu Xiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Jilin 130012, PR China; National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun Jilin 130012, PR China; Institute of Changbai Mountain Resource and Health, Jilin University, Fusong Jilin 134504, PR China.
| |
Collapse
|
15
|
Zainab SR, Khan JZ, Rehman MU, Shah FA, Tipu MK. Effect of Bacillus clausii in attenuating symptoms of DSS-induced ulcerative colitis by modulating NFkB pathway and oxidative stress in mice. Clin Exp Pharmacol Physiol 2024; 51:e70004. [PMID: 39513300 DOI: 10.1111/1440-1681.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024]
Abstract
Ulcerative colitis (UC) is a condition characterized by inflammation and ulcer formation in the colon and rectum due to genetic and environmental factors. It is a common condition, with a global prevalence rate exceeding 0.3%. Current treatments have limited efficacy and can cause unwanted side effects, leading to a high recurrence rate and reduced quality of life for patients. This study suggests that Bacillus clausii has a beneficial role in reducing intestinal inflammation and relieving colitis symptoms in mice. The study aimed to examine B. clausii's potential to reduce the progression and pathogenesis of dextran sulphate sodium (DSS)-induced UC. Bacillus clausii was administered to mice as a pre-treatment, post-treatment and adjunct treatment with sulfasalazine for 14 days. The study found that B. clausii effectively reduced the severity of colitis in mice when used preventatively. Administering B. clausii after the onset of colitis also effectively alleviated symptoms. Combining B. clausii with standard sulfasalazine as adjunct therapy was more effective in reducing intestinal inflammation than using a single therapy alone. B. clausii has shown the potential to prevent colon damage and decrease the likelihood and severity of the disease. Immunohistochemistry results revealed a decrease in the expression of pro-inflammatory cytokines such as IL-1β, TNF-α and NFkB in colon tissue. Additionally, mice that received B. clausii showed a significant increase in anti-oxidant levels and improved haematological markers. In conclusion, it must be emphasized that B. clausii possesses the potential to alleviate the symptoms of UC.
Collapse
Affiliation(s)
- Syeda Rida Zainab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Mujeeb Ur Rehman
- Department of Pharmacy, CECOS University of IT and Emerging Sciences, Peshawar, Pakistan
| | - Fawad Ali Shah
- Department of Pharmacology and Toxicology, College of Pharmacy Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| |
Collapse
|
16
|
Wang L, Wang H, Li X, Zhu M, Gao D, Hu D, Xiong Z, Li X, Qian P. Bacillus velezensis HBXN2020 alleviates Salmonella Typhimurium infection in mice by improving intestinal barrier integrity and reducing inflammation. eLife 2024; 13:RP93423. [PMID: 39560359 PMCID: PMC11575897 DOI: 10.7554/elife.93423] [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] [Indexed: 11/20/2024] Open
Abstract
Bacillus velezensis is a species of Bacillus that has been widely investigated because of its broad-spectrum antimicrobial activity. However, most studies on B. velezensis have focused on the biocontrol of plant diseases, with few reports on antagonizing Salmonella Typhimurium infections. In this investigation, it was discovered that B. velezensis HBXN2020, which was isolated from healthy black pigs, possessed strong anti-stress and broad-spectrum antibacterial activity. Importantly, B. velezensis HBXN2020 did not cause any adverse side effects in mice when administered at various doses (1×107, 1×108, and 1×109 CFU) for 14 days. Supplementing B. velezensis HBXN2020 spores, either as a curative or preventive measure, dramatically reduced the levels of S. Typhimurium ATCC14028 in the mice's feces, ileum, cecum, and colon, as well as the disease activity index (DAI), in a model of infection caused by this pathogen in mice. Additionally, supplementing B. velezensis HBXN2020 spores significantly regulated cytokine levels (Tnfa, Il1b, Il6, and Il10) and maintained the expression of tight junction proteins and mucin protein. Most importantly, adding B. velezensis HBXN2020 spores to the colonic microbiota improved its stability and increased the amount of beneficial bacteria (Lactobacillus and Akkermansia). All together, B. velezensis HBXN2020 can improve intestinal microbiota stability and barrier integrity and reduce inflammation to help treat infection by S. Typhimurium.
Collapse
Affiliation(s)
- Linkang Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Haiyan Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xinxin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Mengyuan Zhu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Dongyang Gao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Dayue Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Zhixuan Xiong
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, the Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
17
|
Félix J, Baca A, Taboada L, Álvarez-Calatayud G, De la Fuente M. Consumption of a Probiotic Blend with Vitamin D Improves Immunity, Redox, and Inflammatory State, Decreasing the Rate of Aging-A Pilot Study. Biomolecules 2024; 14:1360. [PMID: 39595538 PMCID: PMC11591724 DOI: 10.3390/biom14111360] [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: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
There is evidence of the effect of probiotic intake on the immune system. However, the effect probiotics may have on the rate of aging is unknown. The aim of this study is to determine the effect of a probiotic blend on immunity, redox state, inflammation, and the rate of aging or biological age. A group of 10 men and 14 women took, daily for 2 months, a sachet with three probiotics (Bifidobacterium animalis subsp. lactis BSO1, Lactobacillus reuteri LRE02, Lactobacillus plantarum LP14) and vitamin D. Before starting the treatment and after 2 months, peripheral blood was collected. Immune functions were assessed in isolated immune cells, and cytokine concentrations were also measured both in mononuclear cell cultures and plasma. Redox state parameters were also analyzed in whole blood cells. Finally, the Immunity Clock was applied to determine the biological age. Results show that the intake of this probiotic blend in general, in both men and women, improves immunity and decreases the oxidative and inflammatory state. In addition, it rejuvenates the biological age by 10 years on average. It can be concluded that this probiotic blend could be proposed as a good strategy to slow down the aging process, and to achieve healthy aging.
Collapse
Affiliation(s)
- Judith Félix
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, José Antonio Novais, 12, 28040 Madrid, Spain; (A.B.); (M.D.l.F.)
- Institute of Investigation Hospital 12 Octubre (imas12), 28041 Madrid, Spain
| | - Adriana Baca
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, José Antonio Novais, 12, 28040 Madrid, Spain; (A.B.); (M.D.l.F.)
| | - Luz Taboada
- General Medicine Area, Hospital HM Sanchinarro, 28040 Madrid, Spain;
| | - Guillermo Álvarez-Calatayud
- Gastroenterology and Child Nutrition Area, General University Hospital Gregorio Marañón, 28007 Madrid, Spain;
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28040 Madrid, Spain
| | - Mónica De la Fuente
- Department of Genetics, Physiology and Microbiology (Animal Physiology Unit), Faculty of Biological Sciences, Complutense University of Madrid, José Antonio Novais, 12, 28040 Madrid, Spain; (A.B.); (M.D.l.F.)
- Institute of Investigation Hospital 12 Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
18
|
You C, Xing J, Sun J, Zhang D, Yan Y, Dong Y. Anti-Inflammatory and Anti-Oxidant Impacts of Lentinan Combined with Probiotics in Ulcerative Colitis. Mol Biotechnol 2024; 66:2778-2791. [PMID: 37819465 DOI: 10.1007/s12033-023-00878-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023]
Abstract
Multi-methods have been developed to control ulcerative colitis. This research targeted to probe that lentinan combined with probiotics suppresses inflammation and oxidative stress responses in a dextran sulfate sodium (DSS)-induced colitis model. A mouse model of colitis was induced through oral administration with 2.5% DSS and treated with lentinan and probiotics independently or in combination. Then, bodyweight and Disease Activity Index (DAI) of mice were determined. Histopathology of colon tissue was analyzed, and apoptosis, inflammation and oxidative stress in the colon tissue of mice were observed. An HT-29 cell model of colitis was established by DSS stimulation and cultured with lentinan and/or probiotics to examine cell proliferation and apoptosis. The data discovered that after DSS induction of colitis, mice developed weight loss, increased DAI score, and shortened the length of colon. Also, severe histopathology of the colon, and increased apoptosis, inflammation and oxidative stress were recognizable. Lentinan could alleviate DSS-induced colitis, and the highest dose was the most significant. Probiotics could also relieve UC in mice, and mixed probiotics had a better therapeutic effect than single probiotics. Lentinan combined with probiotics could further alleviate DSS-induced colitis damage. In addition, lentinan combined with probiotics impaired apoptosis and enhanced proliferation of DSS-treated HT-29 cells. In a word, lentinan combined with probiotics reduces the inflammatory response and oxidative stress of UC.
Collapse
Affiliation(s)
- CuiYu You
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - JianFeng Xing
- School of Pharmacy, Xi'an Jiaotong University, Xi'an City, 710061, Shaanxi Province, China
| | - JinYao Sun
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Di Zhang
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - Yan Yan
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an City, 710061, Shaanxi Province, China
| | - YaLin Dong
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an City, 710061, Shaanxi Province, China.
| |
Collapse
|
19
|
Cai W, Pierzynowska K, Stiernborg M, Xu J, Nilsson IA, Svensson U, Melas PA, Lavebratt C. Multispecies synbiotics alleviate dextran sulfate sodium (DSS)-induced colitis: Effects on clinical scores, intestinal pathology, and plasma biomarkers in male and female mice. Clin Nutr ESPEN 2024; 63:74-83. [PMID: 38923468 DOI: 10.1016/j.clnesp.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by recurrent inflammation of the gastrointestinal tract and has been linked to an imbalance in gut bacteria. Synbiotics, which combine probiotics and prebiotics, are emerging as potential IBD treatments. AIM To examine the effects of four synbiotic formulations on intestinal inflammation and peripheral biomarkers in a rodent IBD model of both sexes. METHODS Colitis was induced in male and female C57BL/6 mice using 1% dextran sulfate sodium (DSS). Concurrently, a non-exposed control group was maintained. Starting on day 4 post-induction, DSS-exposed mice received one of four synbiotic preparations (Synbio1-4 composed of lactic acid bacteria, Bifidobacterium and dietary fibres), an anti-inflammatory drug used to treat IBD (mesalazine), or placebo (water) until day 14. Clinical symptoms and body weight were monitored daily. Blood samples (taken on days -3, 4, and 14, relative to DSS introduction), were used to analyze plasma biomarkers. At the end of the study, intestinal tissues underwent histological and morphological evaluation. RESULTS Compared to placebo, the Synbio1-, 2- and 3-treated groups had improved clinical scores by day 14. Synbio1 was the only preparation that led to clinical improvements to scores comparable to those of controls. The Synbio1-and 3-treated groups also demonstrated histological improvements in the colon. Plasma biomarker analyses revealed significant Synbio1-induced changes in plasma IL17A, VEGFD, and TNFRSF11B levels that correlated with improved clinical or histological scores. Sex-stratified analyses revealed that most therapeutic-like effects were more pronounced in females. CONCLUSION Our findings underscore the potential therapeutic benefits of specific synbiotics for IBD management. However, further research is needed to validate these outcomes in human subjects.
Collapse
Affiliation(s)
- Wenjie Cai
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | | | - Miranda Stiernborg
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | - Jingjing Xu
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | - Ida Ak Nilsson
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | | | - Philippe A Melas
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden
| | - Catharina Lavebratt
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
20
|
Das M, Dam S. Evaluation of probiotic efficacy of indigenous yeast strain, Saccharomyces cerevisiae Y-89 isolated from a traditional fermented beverage of West Bengal, India having protective effect against DSS-induced colitis in experimental mice. Arch Microbiol 2024; 206:398. [PMID: 39254791 DOI: 10.1007/s00203-024-04128-8] [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: 07/18/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Increasing awareness regarding health promotion and disease prevention has driven inclusion of fermented foods and beverages in the daily diet. These are the enormous sources of beneficial microbes, probiotics. This study aims to isolate yeast strains having probiotic potential and effectivity against colitis. Initially, ninety-two yeast strains were isolated from Haria, an ethnic fermented beverage of West Bengal, India. Primary screening was done by their acid (pH 4) and bile salt (0.3%) tolerance ability. Four potent isolates were selected and found effective against Entamoeba histolytica, as this human pathogen is responsible to cause colitis. They were identified as Saccharomyces cerevisiae. They showed luxurious growth even at 37 oC, tolerance up to 5% of NaCl, resistance to gastric juice and high bile salt (2.0%) and oro-gastrointestinal transit tolerance. They exhibited good auto-aggregation and co-aggregation ability and strong hydrophobicity. Finally, heat map and principal component analysis revealed that strain Y-89 was the best candidate. It was further characterised and found to have significant protective effects against DSS-induced colitis in experimental mice model. It includes improvement in colon length, body weight and organ indices; reduction in disease activity index; reduction in cholesterol, LDL, SGPT, SGOT, urea and creatinine levels; improvement in HDL, ALP, total protein and albumin levels; decrease in coliform count and restoration of tissue damage. This study demonstrates that the S. cerevisiae strain Y-89 possesses remarkable probiotic traits and can be used as a potential bio-therapeutic candidate for the prevention of colitis.
Collapse
Affiliation(s)
- Moubonny Das
- Department of Microbiology, The University of Burdwan, Burdwan, West Bengal, 713104, India
| | - Somasri Dam
- Department of Microbiology, The University of Burdwan, Burdwan, West Bengal, 713104, India.
| |
Collapse
|
21
|
Qian G, Zang H, Tang J, Zhang H, Yu J, Jia H, Zhang X, Zhou J. Lactobacillus gasseri ATCC33323 affects the intestinal mucosal barrier to ameliorate DSS-induced colitis through the NR1I3-mediated regulation of E-cadherin. PLoS Pathog 2024; 20:e1012541. [PMID: 39250508 PMCID: PMC11412683 DOI: 10.1371/journal.ppat.1012541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/19/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory bowel disease (IBD) is an immune system disorder primarily characterized by colitis, the exact etiology of which remains unclear. Traditional treatment approaches currently yield limited efficacy and are associated with significant side effects. Extensive research has indicated the potent therapeutic effects of probiotics, particularly Lactobacillus strains, in managing colitis. However, the mechanisms through which Lactobacillus strains ameliorate colitis require further exploration. In our study, we selected Lactobacillus gasseri ATCC33323 from the intestinal microbiota to elucidate the specific mechanisms involved in modulation of colitis. Experimental findings in a DSS-induced colitis mouse model revealed that L. gasseri ATCC33323 significantly improved physiological damage in colitic mice, reduced the severity of colonic inflammation, decreased the production of inflammatory factors, and preserved the integrity of the intestinal epithelial structure and function. It also maintained the expression and localization of adhesive proteins while improving intestinal barrier permeability and restoring dysbiosis in the gut microbiota. E-cadherin, a critical adhesive protein, plays a pivotal role in this protective mechanism. Knocking down E-cadherin expression within the mouse intestinal tract significantly attenuated the ability of L. gasseri ATCC33323 to regulate colitis, thus confirming its protective role through E-cadherin. Finally, transcriptional analysis and in vitro experiments revealed that L. gasseri ATCC33323 regulates CDH1 transcription by affecting NR1I3, thereby promoting E-cadherin expression. These findings contribute to a better understanding of the specific mechanisms by which Lactobacillus strains alleviate colitis, offering new insights for the potential use of L. gasseri as an alternative therapy for IBD, particularly in dietary supplementation.
Collapse
Affiliation(s)
- Guanru Qian
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hui Zang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jingtong Tang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jiankang Yu
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Huibiao Jia
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Xinzhuang Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| |
Collapse
|
22
|
Ryu HM, Islam SMS, Riaz B, Sayeed HM, Choi B, Sohn S. Immunomodulatory Effects of a Probiotic Mixture: Alleviating Colitis in a Mouse Model through Modulation of Cell Activation Markers and the Gut Microbiota. Int J Mol Sci 2024; 25:8571. [PMID: 39201260 PMCID: PMC11354276 DOI: 10.3390/ijms25168571] [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: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Ulcerative colitis (UC) is a persistent inflammatory intestinal disease that consistently affects the colon and rectum. Its exact cause remains unknown. UC causes a considerable challenge in healthcare, prompting research for novel therapeutic strategies. Although probiotics have gained popularity as possible candidates for managing UC, studies are still ongoing to identify the best probiotics or probiotic mixtures for clinical applications. This study aimed to determine the efficacy of a multi-strain probiotic mixture in mitigating intestinal inflammation in a colitis mouse model induced by dextran sulfate sodium. Specifically, a multi-strain probiotic mixture consisting of Tetragenococcus halophilus and Eubacterium rectale was used to study its impact on colitis symptoms. Anti-inflammatory effects were evaluated using ELISA and flow cytometry. The configuration of gut microbial communities was determined using 16S rRNA metagenomic analysis. According to this study, colitis mice treated with the probiotic mixture experienced reduced weight loss and significantly less colonic shortening compared to untreated mice. Additionally, the treated mice exhibited increased levels of forkhead box P3 (Foxp3) and interleukin 10, along with decreased expression of dendritic cell activation markers, such as CD40+, CD80+, and CD83+, in peripheral blood leukocytes and intraepithelial lymphocytes. Furthermore, there was a significant decrease in the frequencies of CD8+N.K1.1+ cells and CD11b+Ly6G+ cells. In terms of the gut microbiota, probiotic-mixture treatment of colitis mice significantly increased the abundance of the phyla Actinobacteria and Verrucomicrobia (p < 0.05). These results provide valuable insights into the therapeutic promise of multi-strain probiotics, shedding light on their potential to alleviate colitis symptoms. This research contributes to the ongoing exploration of effective probiotic interventions for managing inflammatory bowel disease.
Collapse
Affiliation(s)
- Hye-Myung Ryu
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - S. M. Shamsul Islam
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Bushra Riaz
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Hasan M. Sayeed
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| | - Bunsoon Choi
- Institute of Medical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Seonghyang Sohn
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (S.M.S.I.); (B.R.); (H.M.S.)
| |
Collapse
|
23
|
Haririzadeh Jouriani F, Torfeh M, Torkamaneh M, Sepehr A, Rohani M, Aghamohammad S. The preventive and therapeutic role of Lactobacillus spp. in in vitro model of inflammation via affecting autophagy signaling pathway. Immun Inflamm Dis 2024; 12:e1336. [PMID: 39189796 PMCID: PMC11348509 DOI: 10.1002/iid3.1336] [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: 07/08/2023] [Revised: 06/19/2024] [Accepted: 06/23/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Intestinal inflammation has various causes and leads to some inflammatory diseases, of which autophagy pathway dysfunction could be considered as one of them. Probiotics could have a positive effect on reducing inflammation by activating the autophagy pathway. To evaluate the precise effects of probiotics as preventive and therapeutic agents to control the symptoms of inflammatory diseases, we aimed to investigate the efficacy of Lactobacillus spp. in regulating the autophagy signaling pathway. METHODS A quantitative real-time polymerase chain reaction assay was used to analyze the expression of autophagy genes involved in the formation of phagophores, autophagosomes, and autolysosomes after exposing the HT-29 cell line to sonicated pathogens and adding Lactobacillus spp. before, after, and simultaneously with inflammation. A cytokine assay was also accomplished to evaluate the interleukin (IL)-6 and IL-1β level following the probiotic treatment. RESULTS Lactobacillus spp. generally increased autophagy gene expression and consumption of Lactobacillus spp. before, simultaneously, and after inflammation, ultimately leading to activate autophagy pathways. The proinflammatory cytokines including IL-6 and IL-1β decreased after probiotic treatment. CONCLUSIONS Our native probiotic Lactobacillus spp. showed beneficial effects on HT-29 cells by increasing autophagy gene expression and decreasing the proinflammatory cytokines production in all treatments. Therefore, this novel probiotic cocktail Lactobacillus spp. can prevent and treat inflammation-related diseases.
Collapse
Affiliation(s)
| | - Mahnaz Torfeh
- Department of BacteriologyPasteur Institute of IranTehranIran
| | | | - Amin Sepehr
- Department of BacteriologyPasteur Institute of IranTehranIran
| | - Mahdi Rohani
- Department of BacteriologyPasteur Institute of IranTehranIran
| | | |
Collapse
|
24
|
Vesci L, Tundo G, Soldi S, Galletti S, Stoppoloni D, Bernardini R, Modolea AB, Luberto L, Marra E, Giorgi F, Marini S. A Novel Lactobacillus brevis Fermented with a Vegetable Substrate (AL0035) Counteracts TNBS-Induced Colitis by Modulating the Gut Microbiota Composition and Intestinal Barrier. Nutrients 2024; 16:937. [PMID: 38612971 PMCID: PMC11013894 DOI: 10.3390/nu16070937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Crohn's and ulcerative colitis are common conditions associated with inflammatory bowel disease as well as intestinal flora and epithelial barrier dysfunction. A novel fermented Lactobacillus brevis (AL0035) herein assayed in a trinitro benzene sulfonic acid (TNBS)-induced colitis mice model after oral administration significantly counteracted the body weight loss and improves the disease activity index and histological injury scores. AL0035 significantly decreased the mRNA and protein expression of different pro-inflammatory cytokines (TNFalpha, IL-1beta, IL-6, IL-12, IFN-gamma) and enhanced the expression of IL-10. In addition, the probiotic promoted the expression of tight junction proteins, such as ZO-1, keeping the intestinal mucosal barrier function to attenuate colitis symptoms in mice. Markers of inflammation cascade such as myeloperoxidase (MPO) and PPAR-gamma measured in the colon were also modified by AL0035 treatment. AL0035 was also able to reduce different lymphocyte markers' infiltration in the colon (GATA-3, T-Bet, NK1.1) and monocyte chemoattractant protein-1 (MCP-1/CCL2), a key chemokine involved in the migration and infiltration of monocytes/macrophages in the immunological surveillance of tissues and inflammation. In colonic microbiota profile analysis through 16S rRNA sequencing, AL0035 increased the microbial diversity depleted by TNBS administration and the relative abundance of the Lactobacillaceae and Lachnospiraceae families, whereas it decreased the abundance of Proteobacteria. Altogether, these data indicated that AL0035 could lower the severity of colitis induced by TNBS by regulating inflammatory cytokines, increasing the expression of tight junction proteins and modulating intestinal microbiota, thus preventing tissue damage induced by colitis.
Collapse
Affiliation(s)
- Loredana Vesci
- Corporate R&D, Alfasigma S.p.A., Via Pontina km 30.400, Pomezia, 00071 Rome, Italy;
| | - Grazia Tundo
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| | - Sara Soldi
- AAT Advanced Analytical Technologies Srl, Via P. Majavacca 12, 29017 Fiorenzuola d’Arda, Italy; (S.S.); (S.G.)
| | - Serena Galletti
- AAT Advanced Analytical Technologies Srl, Via P. Majavacca 12, 29017 Fiorenzuola d’Arda, Italy; (S.S.); (S.G.)
| | | | - Roberta Bernardini
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
- Centro Interdipartimentale di Medicina Comparata, Tecniche Alternative ed Acquacoltura (CIMETA), University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Anamaria Bianca Modolea
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| | - Laura Luberto
- Takis Castel Romano, 00128 Rome, Italy; (D.S.); (L.L.); (E.M.)
| | - Emanuele Marra
- Takis Castel Romano, 00128 Rome, Italy; (D.S.); (L.L.); (E.M.)
| | - Fabrizio Giorgi
- Corporate R&D, Alfasigma S.p.A., Via Pontina km 30.400, Pomezia, 00071 Rome, Italy;
| | - Stefano Marini
- Department of Translational Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (R.B.); (A.B.M.); (S.M.)
| |
Collapse
|
25
|
Xu J, Xu H, Guo X, Zhao H, Wang J, Li J, He J, Huang H, Huang C, Zhao C, Li Y, Zhou Y, Peng Y, Nie Y. Pretreatment with an antibiotics cocktail enhances the protective effect of probiotics by regulating SCFA metabolism and Th1/Th2/Th17 cell immune responses. BMC Microbiol 2024; 24:91. [PMID: 38500062 PMCID: PMC10946100 DOI: 10.1186/s12866-024-03251-2] [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: 10/24/2023] [Accepted: 03/07/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Probiotics are a potentially effective therapy for inflammatory bowel disease (IBD); IBD is linked to impaired gut microbiota and intestinal immunity. However, the utilization of an antibiotic cocktail (Abx) prior to the probiotic intervention remains controversial. This study aims to identify the effect of Abx pretreatment from dextran sulfate sodium (DSS)-induced colitis and to evaluate whether Abx pretreatment has an enhanced effect on the protection of Clostridium butyricum Miyairi588 (CBM) from colitis. RESULTS The inflammation, dysbiosis, and dysfunction of gut microbiota as well as T cell response were both enhanced by Abx pretreatment. Additionally, CBM significantly alleviated the DSS-induced colitis and impaired gut epithelial barrier, and Abx pretreatment could enhance these protective effects. Furthermore, CBM increased the benefit bacteria abundance and short-chain fatty acids (SCFAs) level with Abx pretreatment. CBM intervention after Abx pretreatment regulated the imbalance of cytokines and transcription factors, which corresponded to lower infiltration of Th1 and Th17 cells, and increased Th2 cells. CONCLUSIONS Abx pretreatment reinforced the function of CBM in ameliorating inflammation and barrier damage by increasing beneficial taxa, eliminating pathogens, and inducing a protective Th2 cell response. This study reveals a link between Abx pretreatment, microbiota, and immune response changes in colitis, which provides a reference for the further application of Abx pretreatment before microbiota-based intervention.
Collapse
Affiliation(s)
- Jing Xu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Hailan Zhao
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jiaqi Wang
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jianhong Li
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Jie He
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Hongli Huang
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Chong Zhao
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Yingfei Li
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Youlian Zhou
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, China.
| |
Collapse
|
26
|
Guo X, Liu L, Zhao W, Li X, Wang X, Ning A, Cao J, Zhang W, Cao L, Zhong M. The protective effect of Schisandra chinensis (Turcz.) Baill. polysaccharide on DSS-induced ulcerative colitis in mice via the modulation of gut microbiota and inhibition of NF-κB activation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:196-206. [PMID: 37555248 DOI: 10.1002/jsfa.12905] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Schisandra chinensis (Turcz.) Baill, a fruit utilized in traditional Chinese medicine (TCM), has a long history of medical application. It has been used to treat diseases of the gastrointestinal tract. Schisandra chinensis (Turcz.) Baill polysaccharide (SACP) is an important biologically active ingredient that has been shown to have a variety of beneficial effects including immune regulation and anti-oxidative properties. Ulcerative colitis (UC) is a complicated gastrointestinal inflammatory disease. We explore the protective effect of SACP against UC. RESULTS Schisandra chinensis (Turcz.) Baill polysaccharide significantly reduced the disease activity index (DAI) and levels of myeloperoxidase(MPO) and malondialdehyde (MDA) in colonic tissue. It also alleviated weight loss and histopathological damage of mice. The expression of MUC2 and occludin proteins was increased and the barrier function of the colonic mucosa was enhanced by SACP treatment. NF-κB pathway activation was also inhibited and the production of pro-inflammatory cytokines was decreased whereas anti-inflammatory cytokines were increased. 16SrDNA sequencing of fecal flora showed that SACP increased the abundance of Muribaculaceaeunclassified, LachnospiraceaeNK4A136group and reduced the abundance of Bacteroides and Erysipelatoclostridium. CONCLUSION Schisandra chinensis (Turcz.) Baill polysaccharide can protect against Dextran Sulfate Sodium Salt (DSS)-induced ulcerative colitis in mice. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaorong Guo
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
- The Liaoning Province People's Hospital, Shenyang, P.R. China
| | - Lei Liu
- Laboratory of Pathogenic Biology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Wenqi Zhao
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Xingyun Li
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Xiaoli Wang
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Anhong Ning
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Jing Cao
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Wei Zhang
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| | - Liang Cao
- Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, P.R. China
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Science, Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
27
|
Kim SH, Keum B, Kwak S, Byun J, Shin JM, Kim TH. Therapeutic Applications of Extracellular Vesicles in Inflammatory Bowel Disease. Int J Mol Sci 2024; 25:745. [PMID: 38255819 PMCID: PMC10815267 DOI: 10.3390/ijms25020745] [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: 11/29/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
The treatment landscape for inflammatory bowel disease (IBD) has undergone substantial advancements with the introduction of biologics. However, a considerable number of patients either show an immediate lack of response or lose responsiveness over time, necessitating the development of innovative and effective treatment approaches. Extracellular vesicles (EVs) are small lipid bilayer-enclosed structures that facilitate cell-to-cell molecular transfer and are integral to the pathogenesis of IBD. They play pivotal roles in maintaining the integrity of the intestinal epithelial barrier and the expulsion of cellular metabolites. The potential use of EVs as drug carriers or therapeutic agents has opened up a plethora of clinical applications. This review investigates the creation and content of EVs, their role in IBD development, and advances in their isolation and analytical techniques. Furthermore, the therapeutic promise they hold for IBD is explored, along with the latest research on their roles as IBD drug delivery systems.
Collapse
Affiliation(s)
- Sang Hyun Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (S.H.K.)
| | - Bora Keum
- Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (S.H.K.)
| | - Sooun Kwak
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Junhyoung Byun
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jae Min Shin
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Tae Hoon Kim
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
28
|
Ning K, Shi C, Chi YY, Zhou YF, Zheng W, Duan Y, Tong W, Xie Q, Xiang H. Portulaca oleracea L. polysaccharide alleviates dextran sulfate sodium-induced ulcerative colitis by regulating intestinal homeostasis. Int J Biol Macromol 2024; 256:128375. [PMID: 38000581 DOI: 10.1016/j.ijbiomac.2023.128375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
Portulaca oleracea L. (purslane) is a vegetable that contains a variety of active compounds with nutritional properties and has the potential to treat ulcerative colitis (UC). However, the mechanisms underlying the effects of Portulaca oleracea L. polysaccharide (POP) in alleviating UC remain unclear. In this study, we prepared an aqueous extract of purslane and separated a fraction with molecular weight >10 kDa using membrane separation. This fraction was used to isolate POP. The effect of POP on gut microbiota and colon transcriptome in dextran sulfate sodium-induced UC model mice was evaluated. POP treatment reduced inflammation and oxidative stress imbalance in UC mice. In addition, POP improved the intestinal barrier and regulated intestinal homeostasis. Importantly, POP was found to regulate gut microbiota, maintain the levels of retinol and short-chain fatty acids in the gut, promote the proliferation and differentiation of B cells in the colon, and increase the expression of immunoglobulin A. These results provide novel insights into the role of POP in regulating intestinal homeostasis, which should guide further development of POP as a functional food.
Collapse
Affiliation(s)
- Ke Ning
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Chao Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Yan-Yu Chi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Yong-Fei Zhou
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Weiwei Zheng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Yameng Duan
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Weiwei Tong
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China
| | - Qiuhong Xie
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China; Institute of Changbai Mountain Resource and Health, Jilin University, Fusong 134504, PR China.
| | - Hongyu Xiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, PR China; Institute of Changbai Mountain Resource and Health, Jilin University, Fusong 134504, PR China.
| |
Collapse
|
29
|
Kim HJ, Jeon HJ, Kim JY, Shim JJ, Lee JH. Lactiplantibacillus plantarum HY7718 Improves Intestinal Integrity in a DSS-Induced Ulcerative Colitis Mouse Model by Suppressing Inflammation through Modulation of the Gut Microbiota. Int J Mol Sci 2024; 25:575. [PMID: 38203747 PMCID: PMC10779067 DOI: 10.3390/ijms25010575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Inflammatory bowel disease (IBD), a chronic condition that causes persistent inflammation in the digestive system, is closely associated with the intestinal microbiome. Here, we evaluated the effects of Lactiplantibacillus plantarum HY7718 (HY7718) on IBD symptoms in mice with dextran sulfate sodium (DSS)-induced colitis. Oral administration of HY7718 led to significant improvement in the disease activity index score and the histological index, as well as preventing weight loss, in model mice. HY7718 upregulated the expression of intestinal tight junction (TJ)-related genes and downregulated the expression of genes encoding pro-inflammatory cytokines and genes involved in the TLR/MyD88/NF-κB signaling pathway. Additionally, HY7718 reduced the blood levels of pro-inflammatory cytokines, as well as reversing DSS-induced changes to the composition of the intestinal microbiome. HY7718 also increased the percentage of beneficial bacteria (Lactiplantibacillus and Bifidobacterium), which correlated positively with the expression of intestinal TJ-related genes. Finally, HY7718 decreased the population of pathogens such as Escherichia, which correlated with IBD symptoms. The data suggest that HY7718 improves intestinal integrity in colitis model mice by regulating the expression of TJ proteins and inflammatory cytokines, as well as the composition of the intestinal microflora. Thus, L. plantarum HY7718 may be suitable as a functional supplement that improves IBD symptoms and gut health.
Collapse
Affiliation(s)
| | | | - Joo-Yun Kim
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (H.-J.K.); (H.-J.J.); (J.-J.S.); (J.-H.L.)
| | | | | |
Collapse
|
30
|
Zheng W, Shi C, Meng Y, Peng J, Zhou Y, Pan T, Ning K, Xie Q, Xiang H. Integrated network analysis and metabolomics reveal the molecular mechanism of Yinchen Sini decoction in CCl 4-induced acute liver injury. Front Pharmacol 2023; 14:1221046. [PMID: 37818184 PMCID: PMC10561237 DOI: 10.3389/fphar.2023.1221046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Objective: Yinchen Sini decoction (YCSND), a traditional Chinese medicine (TCM) formula, plays a crucial role in the treatment of liver disease. However, the bioactive constituents and pharmacological mechanisms of action remain unclear. The present study aimed to reveal the molecular mechanism of YCSND in the treatment of acute liver injury (ALI) using integrated network analysis and metabolomics. Methods: Ultra-high-performance liquid chromatography coupled with Q-Exactive focus mass spectrum (UHPLC-QE-MS) was utilized to identify metabolites in YCSND, and high-performance liquid chromatography (HPLC) was applied to evaluate the quality of four botanical drugs in YCSND. Cell damage and ALI models in mice were established using CCl4. 1H-NMR metabolomics approach, along with histopathological observation using hematoxylin and eosin (H&E), biochemical measurements, and reverse transcription quantitative real-time PCR (RT-qPCR), was applied to evaluate the effect of YCSND on CCl4- induced ALI. Network analysis was conducted to predict the potential targets of YCSND in ALI. Result: Our results showed that 89 metabolites in YCSND were identified using UHPLC-QE-MS. YCSND protected against ALI by reducing the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), and malondialdehyde (MDA) contents and increasing those of superoxide dismutase (SOD), and glutathione (GSH) both in vivo and in vitro. The 1H-NMRmetabolic pattern revealed that YCSND reversed CCl4-induced metabolic abnormalities in the liver. Additionally, the Kyoto Encyclopedia of Genes and Genome (KEGG) pathway enrichment analysis identified five pathways related to liver injury, including the PI3K-AKT, MAPK, HIF-1, apoptosis, and TNF signaling pathways. Moreover, RT-qPCR showed YCSND regulated the inflammatory response (Tlr4, Il6, Tnfα, Nfκb1, Ptgs2, and Mmp9) and apoptosis (Bcl2, Caspase3, Bax, and Mapk3), and inhibited PI3K-AKT signaling pathway (Pi3k and Akt1). Combined network analysis and metabolomics showed a link between the key targets (Tlr4, Ptgs2, and Mmp9) and vital metabolites (choline, xanthine, lactate, and 3-hydroxybutyric acid) of YCSND in ALI. Conclusion: Overall, the results contribute to the understanding of the therapeutic effects of YCSND on ALI, and indicate that the integrated network analysis and metabolomics could be a powerful strategy to reveal the pharmacological effects of TCM.
Collapse
Affiliation(s)
- Weiwei Zheng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Chao Shi
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yao Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Jian Peng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yongfei Zhou
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Tong Pan
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Ke Ning
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Qiuhong Xie
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong, Jilin, China
| | - Hongyu Xiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong, Jilin, China
| |
Collapse
|
31
|
Han M, Liao W, Dong Y, Fei T, Gai Z. Sustained ameliorative effect of Lactobacillus acidophilus LA85 on dextran sulfate sodium-induced colitis in mice. J Food Sci 2023; 88:3893-3904. [PMID: 37548631 DOI: 10.1111/1750-3841.16723] [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: 03/13/2023] [Revised: 05/28/2023] [Accepted: 07/14/2023] [Indexed: 08/08/2023]
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease associated with immune system dysfunction caused by gut dysbiosis. This study aimed to investigate the alleviating effect of Lactobacillus acidophilus LA85 on colitis and its underlying mechanism using mouse models of dextran sulfate sodium (DSS)-induced UC. The UC mouse models were established by treating C57BL/6J male mice with 2.5% (w/v) DSS in drinking water for 7 days. These mice received supplementation with either L. acidophilus LA85 (1 × 109 colony-forming units/day) or 200 µL of sterile water once daily (LA85-treated and UC model mice, respectively). The disease activity index (DAI), colon length, and histological changes in the colons of mice were then analyzed at Day 21, and the effects of L. acidophilus LA85 on the gut microbiota and serum inflammatory cytokines were also investigated. Compared with the UC model mice, L. acidophilus LA85-treated UC mice showed significant reductions in a variety of colitis symptoms, including weight loss, the DAI score, colon shortening, and colon tissue damage. Lactobacillus acidophilus LA85 supplementation also significantly decreased the serum concentrations of tumor necrosis factor α and interleukin-6 while increasing the serum concentration of IL-10. Furthermore, LA85 supplementation improved the diversity and composition of the gut microbiota, both of which had been decreased by DSS. In particular, L. acidophilus LA85-treated UC mice showed higher relative abundances of Akkermansia and Romboutsia than the UC model mice. These results demonstrate that L. acidophilus LA85 can alleviate inflammatory diseases of the intestine, such as inflammatory bowel disease, by regulating immune responses and restoring the gut microbiota. PRACTICAL APPLICATION: Ulcerative colitis is a type of inflammatory bowel disease caused by imbalance of gut microbiota. This study showed that L. acidophilus LA85 can alleviate DSS-induced colitis in mice through regulation of inflammatory cytokines, protection of intestinal barrier, and regulation of specific gut microbiota. L. acidophilus LA85 is a promising probiotic candidate for the treatment of UC.
Collapse
Affiliation(s)
- Mei Han
- Shanghai Business School, Shanghai, China
| | - Wenyan Liao
- State Key Laboratory of Dairy Biotechnology, Technology Center Bright Dairy & Food Co., Ltd., Shanghai, China
| | - Yao Dong
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Teng Fei
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare Probiotics Co., Ltd., Suzhou, China
| |
Collapse
|
32
|
Yang J, Li D, Zhang M, Lin G, Hu S, Xu H. From the updated landscape of the emerging biologics for IBDs treatment to the new delivery systems. J Control Release 2023; 361:568-591. [PMID: 37572962 DOI: 10.1016/j.jconrel.2023.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/14/2023]
Abstract
Inflammatory bowel diseases (IBDs) treatments have shifted from small-molecular therapeutics to the oncoming biologics. The first-line biologics against the moderate-to-severe IBDs are mainly involved in antibodies against integrins, cytokines and cell adhesion molecules. Besides, other biologics including growth factors, antioxidative enzyme, anti-inflammatory peptides, nucleic acids, stem cells and probiotics have also been explored at preclinical or clinical studies. Biologics with variety of origins have their unique potentials in attenuating immune inflammation or gut mucosa healing. Great advances in use of biologics for IBDs treatments have been archived in recent years. But delivering issues for biologic have also been confronted due to their liable nature. In this review, we will focus on biologics for IBDs treatments in the recent publications; summarize the current landscapes of biologics and their promise to control disease progress. Alternatively, the confronted challenges for delivering biologics will also be analyzed. To combat these drawbacks, some new delivering strategies are provided: firstly, designing the functional materials with high affinity toward biologics; secondly, the delivering vehicle systems to encapsulate the liable biologics; thirdly, the topical adhering delivery systems as enema. To our knowledge, this review is the first study to summarize the updated usage of the oncoming biologics for IBDs, their confronted challenges in term of delivery and the potential combating strategies.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengjiao Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Gaolong Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
33
|
Zhao Z, Sun M, Cui X, Chen J, Liu C, Zhang X. Bacillus coagulans MZY531 alleviates intestinal mucosal injury in immunosuppressive mice via modulating intestinal barrier, inflammatory response, and gut microbiota. Sci Rep 2023; 13:11181. [PMID: 37429891 DOI: 10.1038/s41598-023-38379-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023] Open
Abstract
Bacillus coagulans has a potential role in improving intestinal injury. However, the specific mechanism is still unclear. In this study, the protective effect of B. coagulans MZY531 on intestinal mucosa injury in cyclophosphamide (CYP)-induced immunosuppressed mice were investigated. The results indicated that the immune organ (thymus and spleen) indices of B. coagulans MZY531 treatment groups were significantly increased compared to the CYP group. B. coagulans MZY531 administration promotes the expression of immune proteins (IgA, IgE, IgG, and IgM). B. coagulans MZY531 could upregulate the ileum levels of IFN-γ, IL-2, IL-4, and IL-10 in immunosuppressed mice. Moreover, B. coagulans MZY531 restores the villus height and crypt depth of the jejunum and alleviates injury of intestinal endothelial cells caused by CYP. Furthermore, the western blotting results showed that B. coagulans MZY531 ameliorated CYP-induced intestinal mucosal injury and inflammatory via up-regulates the ZO-1 pathway and down-regulates the expression of the TLR4/MyD88/NF-κB pathway. After treatment with B. coagulans MZY531, the relative abundance of Firmicutes phylum was dramatically increased, as well as the genera of Prevotella and Bifidobacterium, and reducing harmful bacteria. These findings suggested that B. coagulans MZY531 has a potential immunomodulatory activity on chemotherapy-induced immunosuppression.
Collapse
Affiliation(s)
- Zhongwei Zhao
- Medical College, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Manqing Sun
- Medical College, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
| | - Xinmu Cui
- Medical College, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
| | - Jiaxin Chen
- Medical College, Yanbian University, Yanji, 133002, Jilin, People's Republic of China
| | - Chunhong Liu
- College of Special Education, Changchun University, Changchun, 130022, People's Republic of China
| | - Xuewu Zhang
- Medical College, Yanbian University, Yanji, 133002, Jilin, People's Republic of China.
| |
Collapse
|
34
|
Ning K, Duan Y, Tong W, Chen Y, Zhang Q, Xie Q, Xiang H. Protective Effects of Different Molecular Weights of Purslane ( Portulaca oleracea L.) Aqueous Extract on DSS-Induced Ulcerative Colitis in Mice. Antioxidants (Basel) 2023; 12:1400. [PMID: 37507939 PMCID: PMC10376347 DOI: 10.3390/antiox12071400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/29/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Purslane, a common wild vegetable, contains active substances with various biological functions. However, its effects have been under-investigated in ulcerative colitis (UC). Therefore, this study investigated the therapeutic effects of purslane macromolecular (POEM) and small molecular extracts (POES) on dextran sulfate sodium (DSS)-induced UC in mice. Membrane separation was used to obtain extracts of different molecular weights, and their compositional differences were compared using liquid chromatography-mass spectrometry (LC/MS). POEM contained more proteins and polysaccharides, whereas POES contained more organic acids and alkaloids. These differences in composition were directly responsible for the different degrees of remission of the alleviated UC in model mice. POEM may alleviate UC by regulating the antioxidant capacity and the gut microbiota, whereas the major alleviatory effect of POES was primarily related to the regulation of antioxidant capacity. The POEM and POES effects identified in this study provide a theoretical basis for the development of purslane as a functional food.
Collapse
Affiliation(s)
- Ke Ning
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yameng Duan
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Weiwei Tong
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yue Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qinghui Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qiuhong Xie
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong 134504, China
| | - Hongyu Xiang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong 134504, China
| |
Collapse
|
35
|
Zheng W, Zhao Z, Yang Y, Ding L, Yao W. The synbiotic mixture of lactulose and Bacillus coagulans protects intestinal barrier dysfunction and apoptosis in weaned piglets challenged with lipopolysaccharide. J Anim Sci Biotechnol 2023; 14:80. [PMID: 37301956 DOI: 10.1186/s40104-023-00882-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/10/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Lactulose as an effective prebiotic protects intestinal mucosal injury. Bacillus coagulans is widely used in feed additives because of its ability to promote intestinal health. Our previous study suggests that the combination of lactulose and Bacillus coagulans may be a good candidate as alternative for antibiotic growth promoters. However, the in vivo effects of lactulose and Bacillus coagulans on growth and intestinal health under immune challenge in piglets remains unclear. The objective of this study is to explore the protective effects of synbiotic containing lactulose and Bacillus coagulans on the intestinal mucosal injury and barrier dysfunction under immune challenge in weaned piglets. METHODS Twenty four weaned piglets were assigned to 4 groups. Piglets in the CON-saline and LPS-LPS group were fed the basal diet, while others were fed either with chlortetracycline (CTC) or synbiotic mixture of lactulose and Bacillus coagulans for 32 d before injection of saline or lipopolysaccharide (LPS). Piglets were sacrificed 4 h after LPS injection to collect samples to determine intestinal morphology, integrity and barrier functions as well as relative genes and proteins. RESULTS Our data showed that no differences were observed in the growth performance of the four test groups. LPS injection induced higher serum diamine oxidase activities, D-lactic acid levels, and endotoxin status, lower villus height and ratio of villus height to crypt depth, greater mRNA and lower protein expression related tight junction in both jejunum and ileum. In addition, a higher apoptosis index, and protein expression of Bax and caspase-3 were also observed in the LPS challenge group. Interestingly, dietary synbiotic mixture with lactulose and Bacillus coagulans protected against LPS-induced intestinal damage, barrier dysfunction and higher apoptosis as well as CTC. CONCLUSIONS Our data suggest that dietary supplementation of synbiotic mixture with lactulose and Bacillus coagulans showed resilience to LPS-induced intestinal morphological damage, barrier dysfunction and aggressive apoptosis in piglets as well as the protective effects of CTC. These results indicate that synbiotic mixture of lactulose and Bacillus coagulans showed beneficial effects on performance and resilience to acute immune stress in weaned piglets.
Collapse
Affiliation(s)
- Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Zuyan Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Yunnan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Liren Ding
- National Experimental Teaching Center for Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China.
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing, 210095, Jiangsu, China.
| |
Collapse
|
36
|
Chen L, Ou Q, Kou X. Extracellular vesicles and their indispensable roles in pathogenesis and treatment of inflammatory bowel disease: A comprehensive review. Life Sci 2023; 327:121830. [PMID: 37286163 DOI: 10.1016/j.lfs.2023.121830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Inflammatory bowel disease (IBD) is a global disease with rising incidence worldwide, and its debilitating symptoms and dissatisfactory therapies have brought heavy burdens for patients. Extracellular vesicles (EVs), a heterogeneous population of lipid bilayer membranes containing abundant bioactive molecules, have been indicated to play important roles in the pathogenesis and treatment of many diseases. However, to our knowledge, comprehensive reviews summarizing the various roles of diverse source-derived EVs in the pathogenesis and treatment of IBD are still lacking. This review, not only summarizes the EV characteristics, but also focuses on the multiple roles of diverse EVs in IBD pathogenesis and their treatment potential. In addition, hoping to push forward the research frontiers, we point out several challenges that the researchers are faced, about EVs in current IBD research and future therapeutic applications. We also put forward our prospects on future exploration regarding EVs in IBD treatment, including developing IBD vaccines and paying more attention on apoptotic vesicles. This review is aimed to enrich the knowledge on the indispensable roles of EVs in IBD pathogenesis and treatment, providing ideas and reference for future therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Linling Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong 510055, China.
| |
Collapse
|
37
|
Ren R, Zhao AQ, Chen L, Wu S, Hung WL, Wang B. Therapeutic effect of Lactobacillus plantarum JS19 on mice with dextran sulfate sodium induced acute and chronic ulcerative colitis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:4143-4156. [PMID: 36573836 DOI: 10.1002/jsfa.12414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 05/03/2023]
Abstract
BACKGROUND Ulcerative colitis is associated with intestinal inflammation and dysbiosis. Previous studies have shown that probiotics are potential agents for treatment of inflammatory bowel disease (IBD). Jiang-shui is a traditional fermented vegetable that is rich in lactic acid bacteria (LABs), but the preventive effect of LABs in jiang-shui on IBD is not yet fully understood. RESULTS We isolated 38 LAB strains from jiang-shui, and Lactobacillus plantarum JS19 exhibited the strongest antioxidant activity among them. Our data indicate that oral administration of L. plantarum JS19 significantly inhibited body weight loss, colon shortening and damage, and reduced the disease activity index score in the mice with dextran sulfate sodium (DSS)-induced colitis. In addition, L. plantarum JS19 also alleviated inflammatory responses and oxidative stress through reducing lipid peroxidation, tumor necrosis factor-α expression, and myeloperoxidase activity and enhancing the antioxidant enzyme activity. Importantly, L. plantarum JS19 significantly rebalanced DSS-induced dysbiosis of gut microbiota. CONCLUSION L. plantarum JS19 may be used as a potential probiotic to prevent IBD, particularly ulcerative colitis. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Rong Ren
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Ai-Qing Zhao
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Li Chen
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Shan Wu
- Research and Development Center, Xi'an Yinqiao Dairy (Group) Co., Ltd, Xi'an, China
| | - Wei-Lun Hung
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Bini Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
38
|
Shu LZ, Ding YD, Xue QM, Cai W, Deng H. Direct and indirect effects of pathogenic bacteria on the integrity of intestinal barrier. Therap Adv Gastroenterol 2023; 16:17562848231176427. [PMID: 37274298 PMCID: PMC10233627 DOI: 10.1177/17562848231176427] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/01/2023] [Indexed: 06/06/2023] Open
Abstract
Bacterial translocation is a pathological process involving migration of pathogenic bacteria across the intestinal barrier to enter the systemic circulation and gain access to distant organs. This phenomenon has been linked to a diverse range of diseases including inflammatory bowel disease, pancreatitis, and cancer. The intestinal barrier is an innate structure that maintains intestinal homeostasis. Pathogenic infections and dysbiosis can disrupt the integrity of the intestinal barrier, increasing its permeability, and thereby facilitating pathogen translocation. As translocation represents an essential step in pathogenesis, a clear understanding of how barrier integrity is disrupted and how this disruption facilitates bacterial translocation could identify new routes to effective prophylaxis and therapy. In this comprehensive review, we provide an in-depth analysis of bacterial translocation and intestinal barrier function. We discuss currently understood mechanisms of bacterial-enterocyte interactions, with a focus on tight junctions and endocytosis. We also discuss the emerging concept of bidirectional communication between the intestinal microbiota and other body systems. The intestinal tract has established 'axes' with various organs. Among our regulatory systems, the nervous, immune, and endocrine systems have been shown to play pivotal roles in barrier regulation. A mechanistic understanding of intestinal barrier regulation is crucial for the development of personalized management strategies for patients with bacterial translocation-related disorders. Advancing our knowledge of barrier regulation will pave the way for future research in this field and novel clinical intervention strategies.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Yi-Dan Ding
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Qing-Ming Xue
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Wei Cai
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
- Department of Pathology, the Fourth Affiliated
Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated
Hospital of Nanchang University, No. 133 South Guangchang Road, Nanchang
330003, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang
University, Nanchang, China
| |
Collapse
|
39
|
Fei Y, Wang R, Lu J, Peng S, Yang S, Wang Y, Zheng K, Li R, Lin L, Li M. Probiotic intervention benefits multiple neural behaviors in older adults with mild cognitive impairment. Geriatr Nurs 2023; 51:167-175. [PMID: 36990042 DOI: 10.1016/j.gerinurse.2023.03.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023]
Abstract
Probiotic supplements were shown to improve cognitive function in Alzheimer's disease (AD) patients. However, it is still unclear whether this applies to older individuals with mild cognitive impairment (MCI). We aimed to explore the effects of probiotic supplementation on multiple neural behaviors in older adults with MCI. Forty-two MCI patients (age > 60 years) were randomly divided into two groups and consumed either probiotics (n=21) or placebo (n=21) for 12 weeks. Various scale scores, gut microbiota measures and serological indicators were recorded pre- and posttreatment. After 12 weeks of intervention, cognitive function and sleep quality were improved in the probiotic group compared with those in the control group, and the underlying mechanisms were associated with changes in the intestinal microbiota. In conclusion, our study demonstrated that probiotic treatment enhanced cognitive function and sleep quality in older MCI patients, thus providing important insights into the clinical prevention and treatment of MCI.
Collapse
|
40
|
Moon HJ, Oh SH, Park KB, Cha YS. Kimchi and Leuconostoc mesenteroides DRC 1506 Alleviate Dextran Sulfate Sodium (DSS)-Induced Colitis via Attenuating Inflammatory Responses. Foods 2023; 12:foods12030584. [PMID: 36766113 PMCID: PMC9914003 DOI: 10.3390/foods12030584] [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: 01/06/2023] [Revised: 01/18/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Ulcerative colitis (UC) is caused by inflammation only in the mucosa of the colon, and its incidence is increasing worldwide. The intake of probiotics is known to have a beneficial effect on the development of UC. In this study, we investigated the alleviating effects of kimchi (KC), a fermented food rich in probiotics, and Leuconostoc mesenteroides DRC 1506 (DRC) isolated from kimchi on UC. A freeze-dried kimchi suspension and DRC were orally given to mice at a dose of 1 × 109 CFU/day for 3 weeks. Furthermore, 3% dextran sulfate sodium (DSS) in drinking water was given to induce UC. The KC and DRC groups reduced symptoms of colitis, such as disease activity index, decrease in colon length, colon weight-to-length ratio, and pathological damage to the colon caused by DSS treatment. The KC and DRC groups decreased the levels of pro-inflammatory cytokine (TNF-α) and increased anti-inflammatory cytokine (IL-10) in the colon tissues. At the mRNA and protein expression levels in the colon tissue, KC and DRC groups downregulated inflammatory factors and upregulated tight junction-related factors. Therefore, DRC, as well as KC supplementation, are potent in alleviating UC by improving the inflammatory response and mucosal barrier function in the colon.
Collapse
Affiliation(s)
- Hye-Jung Moon
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Suk-Heung Oh
- Department of Food & Biotechnology & Woosuk Institute of Smart Convergence Life Care, Woosuk University, Wanju 55338, Republic of Korea
| | - Ki-Bum Park
- Institute of Kimchi Technology, Daesang Co., Icheon 17384, Republic of Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
- K-Food Research Center, Jeonbuk National University, Jeonju 54896, Republic of Korea
- Correspondence: ; Tel.: +82-63-270-3822
| |
Collapse
|
41
|
He X, Ye G, Xu S, Chen X, He X, Gong Z. Effects of three different probiotics of Tibetan sheep origin and their complex probiotics on intestinal damage, immunity, and immune signaling pathways of mice infected with Clostridium perfringens type C. Front Microbiol 2023; 14:1177232. [PMID: 37138630 PMCID: PMC10149710 DOI: 10.3389/fmicb.2023.1177232] [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: 03/01/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Tibetan sheep have unique intestinal microorganisms in their intestines that are adapted to the highland alpine and anoxic environment. To further clarify the probiotic properties of Tibetan sheep-derived probiotics, we selected three Tibetan sheep-derived probiotic isolates (Enterococcus faecalis EF1-mh, Bacillus subtilis BS1-ql, and Lactobacillus sakei LS-ql) to investigate the protective mechanisms of monocultures and their complex strains against Clostridium perfringens type C infection in mice. We established a model of C. perfringens type C infection and used histology and molecular biology to analyze the effects and mechanisms of different probiotic treatments on mice after C. perfringens type C infection. After supplementation with either probiotics or complex probiotics, mice were improved in terms of weight reduction and reduced the levels of cytokines in serum and increased the levels of intestinal sIgA, and supplementation with complex probiotics was effective. In addition, both probiotic and complex probiotic supplementation effectively improved the damage of intestinal mucosa and spleen tissue. The relative expressions of Muc 2, Claudin-1, and Occludin genes were increased in the ileum. The three probiotics and the compound probiotics treatment significantly reduced the relative mRNA expression of toll-like/MyD88/NF-κB/MAPK. The effect of probiotic treatment was similar to the results of engramycin treatment, but the effect of engramycin treatment on intestinal sIgA was not significant. Our results clarify the immunomodulatory effects of the three probiotic isolates and the complex probiotics on C. perfringens infection, and the repair of the intestinal mucosal barrier.
Collapse
|
42
|
Li L, Qiu N, Meng Y, Wang C, Mine Y, Keast R, Guyonnet V. Preserved egg white alleviates DSS-induced colitis in mice through the reduction of oxidative stress, modulation of infl ammatory cytokines, NF-κB, MAPK and gut microbiota composition. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
43
|
Lactobacillus gasseri JM1 Isolated from Infant Feces Alleviates Colitis in Mice via Protecting the Intestinal Barrier. Nutrients 2022; 15:nu15010139. [PMID: 36615796 PMCID: PMC9823819 DOI: 10.3390/nu15010139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic and recurrent inflammatory bowel disease, and the intestinal barrier is an important line of defense against intestinal disease. Herein, we investigated the effect of Lactobacillus gasseri JM1 at different doses (1 × 106, 1 × 107, 1 × 108 CFU/day) on colitis mice and explored the possible mechanism. The results showed that L. gasseri JM1 alleviated DSS-induced colitis in mice, with reductions in disease activity index (DAI), histological scores and myeloperoxidase activity as well as alleviation of colonic shortening. Furthermore, L. gasseri JM1 regulated the levels of inflammatory cytokines TNF-α, IL-6, IL-1β, and IL-10; restored the expression of Claudin-3, Occludin, ZO-1, and MUC2; and increased the number of goblet cells and acidic mucin. The 16S rDNA sequencing results indicated that intervention with L. gasseri JM1 balanced the gut microbiota structure by elevating the abundance of beneficial bacteria (Oscillospira, Clostridium and Ruminococcus) and decreasing that of harmful bacteria (Shigella and Turicibacter). Meanwhile, the contents of short-chain fatty acids (SCFAs) increased. In conclusion, L. gasseri JM1 could alleviate intestinal barrier damage in colitis mice by modulating the tight junction structures, intestinal mucus layer, inflammatory cytokines, gut microbiota, and SCFAs. It can be considered a potential preventive strategy to alleviate colitis injury.
Collapse
|
44
|
Hu Y, Chen Z, Xu C, Kan S, Chen D. Disturbances of the Gut Microbiota and Microbiota-Derived Metabolites in Inflammatory Bowel Disease. Nutrients 2022; 14:5140. [PMID: 36501169 PMCID: PMC9735443 DOI: 10.3390/nu14235140] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/10/2022] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is characterized as a chronic and recurrent inflammatory disease whose pathogenesis is still elusive. The gut microbiota exerts important and diverse effects on host physiology through maintaining immune balance and generating health-benefiting metabolites. Many studies have demonstrated that IBD is associated with disturbances in the composition and function of the gut microbiota. Both the abundance and diversity of gut microbiota are dramatically decreased in IBD patients. Furthermore, some particular classes of microbiota-derived metabolites, principally short-chain fatty acids, tryptophan, and its metabolites, and bile acids have also been implicated in the pathogenesis of IBD. In this review, we aim to define the disturbance of gut microbiota and the key classes of microbiota-derived metabolites in IBD pathogenesis. In addition, we also focus on scientific evidence on probiotics, not only on the molecular mechanisms underlying the beneficial effects of probiotics on IBD but also the challenges it faces in safe and appropriate application.
Collapse
Affiliation(s)
- Yongjia Hu
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Zhouzhou Chen
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Chengchen Xu
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Shidong Kan
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, No. 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| |
Collapse
|
45
|
Srivastava P, Kim KS. Membrane Vesicles Derived from Gut Microbiota and Probiotics: Cutting-Edge Therapeutic Approaches for Multidrug-Resistant Superbugs Linked to Neurological Anomalies. Pharmaceutics 2022; 14:2370. [PMID: 36365188 PMCID: PMC9692612 DOI: 10.3390/pharmaceutics14112370] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Multidrug-resistant (MDR) superbugs can breach the blood-brain barrier (BBB), leading to a continuous barrage of pro-inflammatory modulators and induction of severe infection-related pathologies, including meningitis and brain abscess. Both broad-spectrum or species-specific antibiotics (β-lactamase inhibitors, polymyxins, vancomycin, meropenem, plazomicin, and sarecycline) and biocompatible poly (lactic-co-glycolic acid) (PLGA) nanoparticles have been used to treat these infections. However, new therapeutic platforms with a broad impact that do not exert off-target deleterious effects are needed. Membrane vesicles or extracellular vesicles (EVs) are lipid bilayer-enclosed particles with therapeutic potential owing to their ability to circumvent BBB constraints. Bacteria-derived EVs (bEVs) from gut microbiota are efficient transporters that can penetrate the central nervous system. In fact, bEVs can be remodeled via surface modification and CRISPR/Cas editing and, thus, represent a novel platform for conferring protection against infections breaching the BBB. Here, we discuss the latest scientific research related to gut microbiota- and probiotic-derived bEVs, and their therapeutic modifications, in terms of regulating neurotransmitters and inhibiting quorum sensing, for the treatment of neurodegenerative diseases, such as Parkinson's and Alzheimer's diseases. We also emphasize the benefits of probiotic-derived bEVs to human health and propose a novel direction for the development of innovative heterologous expression systems to combat BBB-crossing pathogens.
Collapse
Affiliation(s)
| | - Kwang-sun Kim
- Department of Chemistry and Chemistry Institute of Functional Materials, Pusan National University, Busan 46241, Korea
| |
Collapse
|
46
|
Pan T, Zheng S, Zheng W, Shi C, Ning K, Zhang Q, Xie Y, Xiang H, Xie Q. Christensenella regulated by Huang-Qi-Ling-Hua-San is a key factor by which to improve type 2 diabetes. Front Microbiol 2022; 13:1022403. [PMID: 36312936 PMCID: PMC9597676 DOI: 10.3389/fmicb.2022.1022403] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
There is a lot of evidence that oral hypoglycemic drugs work by affecting gut microbes, but the key strains responsible for this effect are not well known. Huang-Qi-Ling-Hua-San (HQLHS), composed of Astragalus Membranaceus, Ganoderma lucidum, Inonotus obliquus, and Momordica charantia L., is a specially designed Chinese medicine formula to treat type 2 diabetes (T2D). In this study, a mouse model of T2D induced by high-fat diet and streptozotocin was used to explore the mechanism of HQLHS in improving hyperglycemia and hyperlipidemia through multiple rounds of animal experiments, such as HQLHS feeding, fecal microbiota transplantation (FMT), and live bacteria feeding, so as to explore the potential target intestinal flora in its hypoglycemic effect. Results show that such specific taxa as Bifidobacterium, Turicibacter, Alistipes, Romboutsia, and Christensenella were identified to be preferably enriched by HQLHS and then assumed to be the target microbes. Herein, FMT was used to test if the upregulated beneficial bacteria by HQLHS play a therapeutic role. The strain Christensenella minuta DSM 22607 and the strain Christensenella timonensis DSM 102800 were selected to test the beneficial effect of Christensenella taxa on T2D. Diabetic animals supplemented with these strains showed the improvement in blood glucose and lipid metabolism, the promotion of GLP-1 secretion, the increase in antioxidant capacity, the inhibition of hepatic gluconeogenesis, the suppression of intestinal glucose absorption, the enhancement of intestinal barrier, reduced LPS-induced inflammation, and the reduction of branched amino acids (BCAAs) content in the liver. Overall, these data demonstrate that Christensenella plays a beneficial role in T2D and is a target for the action of HQLHS therapy.
Collapse
Affiliation(s)
- Tong Pan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Shujun Zheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Weiwei Zheng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Chao Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Ke Ning
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Qinghui Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Yanbo Xie
- Jilin Provincial Key Laboratory of Agricultural Biotechnology, Jilin Academy of Agricultural Sciences, Changchun, China
- *Correspondence: Hongyu Xiang, ; Qiuhong Xie, ; Yanbo Xie
| | - Hongyu Xiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong, China
- *Correspondence: Hongyu Xiang, ; Qiuhong Xie, ; Yanbo Xie
| | - Qiuhong Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
- Institute of Changbai Mountain Resource and Health, Jilin University, Fusong, China
- *Correspondence: Hongyu Xiang, ; Qiuhong Xie, ; Yanbo Xie
| |
Collapse
|
47
|
Li L, Liu T, Gu Y, Wang X, Xie R, Sun Y, Wang B, Cao H. Regulation of gut microbiota-bile acids axis by probiotics in inflammatory bowel disease. Front Immunol 2022; 13:974305. [PMID: 36211363 PMCID: PMC9539765 DOI: 10.3389/fimmu.2022.974305] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/31/2022] [Indexed: 12/02/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic and relapsing inflammation of gastrointestinal tract, with steadily increased incidence and prevalence worldwide. Although the precise pathogenesis remains unclear, gut microbiota, bile acids (BAs), and aberrant immune response play essential roles in the development of IBD. Lately, gut dysbiosis including certain decreased beneficial bacteria and increased pathogens and aberrant BAs metabolism have been reported in IBD. The bacteria inhabited in human gut have critical functions in BA biotransformation. Patients with active IBD have elevated primary and conjugated BAs and decreased secondary BAs, accompanied by the impaired transformation activities (mainly deconjugation and 7α-dehydroxylation) of gut microbiota. Probiotics have exhibited certain positive effects by different mechanisms in the therapy of IBD. This review discussed the effectiveness of probiotics in certain clinical and animal model studies that might involve in gut microbiota-BAs axis. More importantly, the possible mechanisms of probiotics on regulating gut microbiota-BAs axis in IBD were elucidated, which we focused on the elevated gut bacteria containing bile salt hydrolase or BA-inducible enzymes at genus/species level that might participate in the BA biotransformation. Furthermore, beneficial effects exerted by activation of BA-activated receptors on intestinal immunity were also summarized, which might partially explain the protect effects and mechanisms of probiotics on IBD. Therefore, this review will provide new insights into a better understanding of probiotics in the therapy targeting gut microbiota-BAs axis of IBD.
Collapse
|
48
|
Wang H, Fan C, Zhao Z, Zhai Z, Hao Y. Anti-inflammatory effect of Bifidobacterium animalis subsp. lactis A6 on DSS-induced colitis in mice. J Appl Microbiol 2022; 133:2063-2073. [PMID: 35737740 DOI: 10.1111/jam.15681] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/02/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
AIMS This study aimed to investigate the protective effect of Bifidobacterium animalis subsp. lactis A6 on dextran sodium sulphate (DSS)-induced colitis in C57BL/6J mice. METHODS AND RESULTS Mice were randomly divided into three groups (n = 8 per group). Each group was administered with PBS (Control and DSS group) or B. lactis A6 with a dosage of ~4.0 × 109 CFU day-1 (DSS + A6 group) for 21 consecutive days. The DSS and DSS + A6 group mice were ad libitum drinking 2.5% DSS water during day 15-21, while the Control group mice were given normal water. The administration of B. lactis A6 significantly inhibited DSS-induced bodyweight loss and colon shortening (p < 0.001), but showed no significant influence on the spleen enlargement (p > 0.05). The intestinal barrier integrity was improved by reducing colonic damage, recovering mucus layer loss and enhancing tight junction expression including ZO-1, occludin and claudin-1. In addition, B. lactis A6 attenuated the oxidative stress by decreasing MDA and increasing SOD and GSH levels in colon tissues. Moreover, B. lactis A6 suppressed DSS-induced inflammatory responses via downregulating TNF-α, IL-1β and IL-6 levels and upregulating IL-10 level in colon tissues. CONCLUSION B. lactis A6 effectively alleviated DSS-induced colitis by maintaining intestinal barrier integrity, reducing oxidative stress and inhibiting inflammatory responses. SIGNIFICANCE AND IMPACT OF THE STUDY This study suggests that B. lactis A6 could act as a candidate probiotic for UC treatment.
Collapse
Affiliation(s)
- Hui Wang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Chengfei Fan
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhaoer Zhao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zhengyuan Zhai
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
49
|
Wang Q, Guo X, Yue Q, Zhu S, Guo L, Li G, Zhou Q, Xiang Y, Chen G, Yin W, Sun J. Exploring the role and mechanism of gut microbiota in methamphetamine addiction using antibiotic treatment followed by fecal microbiota transplantation. Anat Rec (Hoboken) 2022; 306:1149-1164. [PMID: 36054423 DOI: 10.1002/ar.25055] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/11/2022] [Accepted: 07/12/2022] [Indexed: 11/09/2022]
Abstract
Recently, the role of the gut microbiota in the context of drug addiction has attracted the attention of researchers; however, the specific effects and underlying mechanisms require further exploration. To accomplish this, C57BL/6 mice were firstly treated with methamphetamine (MA). Conditioned place preference (CPP) behavior changes, gut permeability and function, microglial activation, and inflammatory cytokine expression were systematically analyzed in antibiotics-treated mice with microbiota depletion and in fecal microbiota transplantation mice with microbiota reconstitution. MA treatment altered microbiota composition and caused gut dysbiosis. Depletion of gut microbiota with antibiotics inhibited MA-induced CPP formation, and fecal microbiota transplantation reversed this inhibition. Mechanistic analyses indicated that antibiotic treatment decreased gut permeability and neuroinflammation, while fecal microbiota transplantation offset the impact of antibiotic treatment. Additionally, MA-induced microglial activation was suppressed by antibiotics but restored by microbiota transplantation, and this correlated well with the CPP score. Compared to antibiotic treatment, microbiota transplantation significantly increased 5-HT4 receptor expression in both the nucleus accumbens and the hippocampus. Furthermore, when fecal microbiota from healthy mice was transplanted into MA-treated mice, the CPP scores decreased. Our results provide a novel avenue for understanding MA addiction and suggest a potential future intervention strategy.
Collapse
Affiliation(s)
- Qiuting Wang
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Xiuwen Guo
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Qingwei Yue
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Shaowei Zhu
- Department of Neurology Qilu Hospital of Shandong University Jinan China
| | - Liying Guo
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Guibao Li
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Qidi Zhou
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Yunzhi Xiang
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Ganggang Chen
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Wei Yin
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medicine Shandong University Jinan China
| |
Collapse
|
50
|
Zhou B, Te Ba, Wang L, Gao Y, He Q, Yan Z, Wang H, Shen G. Combination of sodium butyrate and probiotics ameliorates severe burn-induced intestinal injury by inhibiting oxidative stress and inflammatory response. Burns 2022; 48:1213-1220. [PMID: 34903409 DOI: 10.1016/j.burns.2021.11.009] [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: 06/14/2021] [Revised: 09/28/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Burns are a common traumatic injuries with considerable morbidity and mortality rates. Post-burn intestinal injuries are closely related to oxidative stress and inflammatory response. The aim of the current study was to investigate the combined effect of sodium butyrate (NaB) and probiotics (PROB) on severe burn-induced oxidative stress and inflammatory response and the underlying mechanism of action. Sprague-Dawley rats with severe burns were treated with NaB with or without PROB. Pathomorphology of skin and small intestine tissue was observed using hematoxylin and eosin staining and severe burn-induced apoptosis in small intestine tissue was examined via terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. The release of factors related to inflammation was quantified using ELISA kits and qRT-PCR and levels of oxidative stress markers were evaluated using biochemical assays. Furthermore, mitochondrial morphological changes in small intestinal epithelial cells were observed using transmission electron microscopy. In addition, the underlying mechanism associated with the combined effect of NaB and PROB on severe burn-induced oxidative stress and inflammatory response was investigated using western blotting. The combination of NaB and PROB exerted protective effects against severe burn-induced intestinal barrier injury by reducing the levels of diamine oxidase and intestinal fatty acid binding protein. Combined NaB and PROB treatment inhibited severe burn-induced oxidative stress by increasing superoxide dismutase levels and decreasing those of malondialdehyde and myeloperoxidase levels. Severe burn-induced inflammation was suppressed by combined NaB and PROB administration, as demonstrated by the decreased mRNA expression of tumor necrosis factor-α, interleukin-6, interleukin-1β, and high mobility group box-1 in the small intestine. In addition, this study showed that combined NaB and PROB administration increased nuclear factor-erythroid 2-related factor 2 (Nrf2) protein expression and decreased the phosphorylation of nuclear factor (NF)-κB and extracellular signal-regulated kinase 1/2 (ERK 1/2). In conclusion, our findings indicate that combined NaB and PROB treatment may inhibit severe burn-induced inflammation and oxidative stress in the small intestine by regulating HMGB1/NF-κB and ERK1/2/Nrf2 signaling, thereby providing a new therapeutic strategy for intestinal injury induced by severe burn.
Collapse
Affiliation(s)
- Biao Zhou
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Te Ba
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Lingfeng Wang
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Yixuan Gao
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Qiaoling He
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Zengqiang Yan
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Hongyu Wang
- Department of Burns, The Third Affiliated Hospital of Inner Mongolia Medical University, Burns Institute of Inner Mongolia, Baotou 014010, PR China
| | - Guoliang Shen
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China.
| |
Collapse
|