1
|
Radice E, Potapov O, Shabat G, Martello E, Meineri G, Risso P, DI Pierro F, Olandese F. Innovative strategies for the rapid restoration of intestinal function in patients undergoing abdominal surgery: use of probiotics. Pilot study of 15 patients. Minerva Gastroenterol (Torino) 2025; 71:7-14. [PMID: 39639791 DOI: 10.23736/s2724-5985.24.03701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
BACKGROUND The clinical value of probiotics in patients undergoing abdominal surgery, particularly colorectal surgery, remains uncertain despite their well-documented health benefits. This pilot randomized controlled trial aimed to assess the effects of perioperative and postoperative oral administration of two probiotics, Clostridium butyricum CBM588® and Bifidobacterium longum ES1, on immune function, systemic inflammatory response, postoperative infections, and recovery after colorectal surgery. METHODS Fifteen adult patients underwent colorectal resection, with two groups receiving probiotics and one acting as a control. Blood and fecal samples were collected, and clinical parameters were assessed. RESULTS Results showed the safety of probiotics, resistance to antibiotics and gastric acid, and potential benefits in reducing postoperative infections and intestinal inflammation. CONCLUSIONS Future trials should provide more conclusive evidence on the efficacy and safety of perioperative probiotic administration in colorectal surgery, aiming for improved patient outcomes and reduced healthcare costs.
Collapse
|
2
|
Yamamoto Y, Furukawa S, Yoshida O, Miyake T, Shiraishi K, Hashimoto Y, Tange K, Kitahata S, Ninomiya T, Yagi S, Masakazu H, Suzuki S, Shibata N, Murakami H, Ohashi K, Tomida H, Takeshita E, Ikeda Y, Hiasa Y. Association between allergic diseases and mucosal healing in ulcerative colitis. Sci Rep 2025; 15:1272. [PMID: 39779831 PMCID: PMC11711406 DOI: 10.1038/s41598-025-85916-0] [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: 03/24/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025] Open
Abstract
Allergic diseases and ulcerative colitis (UC) share pathophysiological similarities. This study aimed to investigate the unclear association between allergic diseases and mucosal healing (MH), an important factor in the prognosis of UC. We studied 289 Japanese patients with UC. Information on allergic diseases (bronchial asthma, atopic dermatitis, pollen allergy, food allergy, and drug allergy), as diagnosed by physicians, was collected through self-reported questionnaires. The definition of MH was Mayo Endoscopic Score 0. The association between each allergic disease and its multimorbidity with MH was evaluated using multivariate logistic regression analyses. Pollen allergy was the most common allergic condition (36.3%). Pollen allergy and food allergy were independently associated with MH (pollen allergy adjusted OR: 1.82 [95% CI: 1.01-3.26]; food allergy adjusted OR: 3.47 [95% CI: 1.26-9.68]). The rates of MH for 0 and 3 or more allergic diseases were 24.6% and 4.2%, respectively. After adjustment for confounders, 3 or more allergic diseases were independently associated with MH (adjusted OR: 8.13 [95% CI: 2.17-34.04], p for trend = 0.020). This study demonstrates a significant positive association between specific allergic diseases (pollen and food allergies) and MH in UC patients, with a stronger association in cases of allergic multimorbidity.
Collapse
Affiliation(s)
| | - Shinya Furukawa
- Health Services Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan.
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Teruki Miyake
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kana Shiraishi
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yu Hashimoto
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kazuhiro Tange
- Department of Inflammatory Bowel Diseases and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shogo Kitahata
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Matsuyama, Ehime, Japan
| | - Tomoyuki Ninomiya
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Matsuyama, Ehime, Japan
| | - Sen Yagi
- Department of Internal Medicine, Saiseikai Imabari Hospital, Imabari, Ehime, Japan
| | - Hanayama Masakazu
- Department of Gastroenterology, Matsuyama Shimin Hospital, Matsuyama, Ehime, Japan
| | - Seiyuu Suzuki
- Department of Gastroenterology, Sumitomo Besshi Hospital, Niihama, Ehime, Japan
| | - Naozumi Shibata
- Department of Gastroenterology, Ehime Prefectural Niihama Hospital, Niihama, Ehime, Japan
| | - Hidehiro Murakami
- Department of Internal Medicine, Saiseikai Matsuyama Hospital, Matsuyama, Ehime, Japan
| | - Katsuhisa Ohashi
- OHASHI Clinic Participate in Gastro-Enterology and Ano-Proctology, Niihama, Ehime, Japan
| | - Hideomi Tomida
- Endoscopy Center, Ehime University Hospital, Toon, Ehime, Japan
| | - Eiji Takeshita
- Department of Inflammatory Bowel Diseases and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yoshio Ikeda
- Endoscopy Center, Ehime University Hospital, Toon, Ehime, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
3
|
Zhang W, Mackay CR, Gershwin ME. Immunomodulatory Effects of Microbiota-Derived Short-Chain Fatty Acids in Autoimmune Liver Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1629-1639. [PMID: 37186939 PMCID: PMC10188201 DOI: 10.4049/jimmunol.2300016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/01/2023] [Indexed: 05/17/2023]
Abstract
Nonpathogenic commensal microbiota and their metabolites and components are essential to maintain a tolerogenic environment and promote beneficial health effects. The metabolic environment critically impacts the outcome of immune responses and likely impacts autoimmune and allergic responses. Short-chain fatty acids (SCFAs) are the main metabolites produced by microbial fermentation in the gut. Given the high concentration of SCFAs in the gut and portal vein and their broad immune regulatory functions, SCFAs significantly influence immune tolerance and gut-liver immunity. Alterations of SCFA-producing bacteria and SCFAs have been identified in a multitude of inflammatory diseases. These data have particular significance in primary biliary cholangitis, primary sclerosing cholangitis, and autoimmune hepatitis because of the close proximity of the liver to the gut. In this focused review, we provide an update on the immunologic consequences of SCFA-producing microbiota and in particular on three dominant SCFAs in autoimmune liver diseases.
Collapse
Affiliation(s)
- Weici Zhang
- Division of Rheumatology, Allergy, and Clinical Immunology, School of Medicine, University of California Davis, CA, USA
| | - Charles R. Mackay
- Department of Microbiology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - M. Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, School of Medicine, University of California Davis, CA, USA
| |
Collapse
|
4
|
Štofilová J, Kvaková M, Kamlárová A, Hijová E, Bertková I, Guľašová Z. Probiotic-Based Intervention in the Treatment of Ulcerative Colitis: Conventional and New Approaches. Biomedicines 2022; 10:2236. [PMID: 36140337 PMCID: PMC9496552 DOI: 10.3390/biomedicines10092236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Although there are number of available therapies for ulcerative colitis (UC), many patients are unresponsive to these treatments or experience secondary failure during treatment. Thus, the development of new therapies or alternative strategies with minimal side effects is inevitable. Strategies targeting dysbiosis of gut microbiota have been tested in the management of UC due to the unquestionable role of gut microbiota in the etiology of UC. Advanced molecular analyses of gut microbiomes revealed evident dysbiosis in UC patients, characterized by a reduced biodiversity of commensal microbiota. Administration of conventional probiotic strains is a commonly applied approach in the management of the disease to modify the gut microbiome, improve intestinal barrier integrity and function, and maintain a balanced immune response. However, conventional probiotics do not always provide the expected health benefits to a patient. Their benefits vary significantly, depending on the type and stage of the disease and the strain and dose of the probiotics administered. Their mechanism of action is also strain-dependent. Recently, new candidates for potential next-generation probiotics have been discovered. This could bring to light new approaches in the restoration of microbiome homeostasis and in UC treatment in a targeted manner. The aim of this paper is to provide an updated review on the current options of probiotic-based therapies, highlight the effective conventional probiotic strains, and outline the future possibilities of next-generation probiotic and postbiotic supplementation and fecal microbiota transplantation in the management of UC.
Collapse
Affiliation(s)
- Jana Štofilová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, Trieda SNP 1, 040 11 Kosice, Slovakia
| | | | | | | | | | | |
Collapse
|
5
|
Liang L, Yang C, Liu L, Mai G, Li H, Wu L, Jin M, Chen Y. Commensal bacteria-derived extracellular vesicles suppress ulcerative colitis through regulating the macrophages polarization and remodeling the gut microbiota. Microb Cell Fact 2022; 21:88. [PMID: 35578339 PMCID: PMC9109417 DOI: 10.1186/s12934-022-01812-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/03/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The extracellular vesicles (EVs) traffic constitutes an essential pathway of cellular communication. And the molecules in EVs produced by procaryotes help in maintaining homeostasis, addressing microbial imbalance and infections, and regulating the immune system. Despite the fact that Clostridium butyricum (C. butyricum) is commonly used for treating ulcerative colitis (UC), the potential role of C. butyricum-secreted EVs in commensals-host crosstalk remains unclear. RESULTS Here, we performed flow cytometry, western blot, immunohistochemistry and 16S rRNA analysis to explore the role of C. butyricum-derived EVs on macrophage polarization and gut microbiota composition in a dextran sulfate sodium (DSS)-induced UC mouse model. The antibiotic cocktail-induced microbiome depletion and faecal transplantations were used to further investigate the mechanisms by which EVs regulate macrophage balance. Our findings showed that C. butyricum-derived EVs improved the remission of murine colitis and polarized the transformation of macrophages to the M2 type. Furthermore, C. butyricum-derived EVs restored gut dysbiosis and altered the relative abundance of Helicobacter, Escherichia-Shigella, Lactobacillus, Akkermansia and Bacteroides, which, in turn, faecal transplantations from EVs-treated mice relieved the symptoms of UC and improved the impact of EVs on the reprogramming of the M2 macrophages. CONCLUSION C. butyricum-derived EVs could protect against DSS-induced colitis by regulating the repolarization of M2 macrophages and remodelling the composition of gut microbiota, suggesting the potential efficacy of EVs from commensal and probiotic Clostridium species against UC.
Collapse
Affiliation(s)
- Liping Liang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Road, Guangzhou, 510515, China
| | - Chenghai Yang
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Le Liu
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Genghui Mai
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Road, Guangzhou, 510515, China
| | - Haolin Li
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Road, Guangzhou, 510515, China
| | - Lele Wu
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Ming Jin
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China
| | - Ye Chen
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Road, Guangzhou, 510515, China.
- Integrative Microecology Center, Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, 518100, China.
| |
Collapse
|
6
|
Li Y, Liu M, Liu H, Sui X, Liu Y, Wei X, Liu C, Cheng Y, Ye W, Gao B, Wang X, Lu Q, Cheng H, Zhang L, Yuan J, Li M. The Anti-Inflammatory Effect and Mucosal Barrier Protection of Clostridium butyricum RH2 in Ceftriaxone-Induced Intestinal Dysbacteriosis. Front Cell Infect Microbiol 2021; 11:647048. [PMID: 33842393 PMCID: PMC8027357 DOI: 10.3389/fcimb.2021.647048] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
This study aimed at determining the beneficial effect of Clostridium butyricum (CB) RH2 on ceftriaxone-induced dysbacteriosis. To this purpose, BALB/c mice were exposed to ceftriaxone (400 mg/ml) or not (control) for 7 days, and administered a daily oral gavage of low-, and high-dose CB RH2 (108 and 1010 CFU/ml, respectively) for 2 weeks. CB RH2 altered the diversity of gut microbiota, changed the composition of gut microbiota in phylum and genus level, decreased the F/B ratio, and decreased the pro-inflammatory bacteria (Deferribacteres, Oscillibacter, Desulfovibrio, Mucispirillum and Parabacteroides) in ceftriaxone-treated mice. Additionally, CB RH2 improved colonic architecture and intestinal integrity by improving the mucous layer and the tight junction barrier. Furthermore, CB RH2 also mitigated intestinal inflammation through decreasing proinflammatory factors (TNF-α and COX-2) and increasing anti-inflammatory factors (IL-10). CB RH2 had direct effects on the expansion of CD4+ T cells in Peyer’s patches (PPs) in vitro, which in turn affected their immune response upon challenge with ceftriaxone. All these data suggested that CB RH2 possessed the ability to modulate the intestinal mucosal and systemic immune system in limiting intestinal alterations to relieve ceftriaxone-induced dysbacteriosis.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Man Liu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - He Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xue Sui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Chunzheng Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yiqin Cheng
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Weikang Ye
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Binbin Gao
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xin Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiao Lu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Hao Cheng
- Marketing Department, Hangzhou Grand Biologic Pharmaceutical Inc., Hangzhou, China
| | - Lu Zhang
- Marketing Department, Hangzhou Grand Biologic Pharmaceutical Inc., Hangzhou, China
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
7
|
Xie Y, Zhou L, Li H, Li Y. Clostridium butyricum Supernatant Regulates the Expression of RORγt in HCT-116 Cells by Inhibiting the TLR2/MyD88/NF-κB Signaling Pathway. Curr Microbiol 2021; 78:1543-1550. [PMID: 33675405 DOI: 10.1007/s00284-021-02392-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 02/07/2021] [Indexed: 11/29/2022]
Abstract
In this study, we treated HCT-116 cells with Clostridium butyricum (C. butyricum) supernatant and observed its effects on the TLR2/MyD88/NF-κB signaling pathway and RORγt, to further explore the possible immune regulatory mechanism of C. butyricum. Our results showed that C. butyricum supernatant downregulated the mRNA and protein levels of TLR2, MyD88, NF-κBp65, and RORγt in HCT-116 cells and the protein levels of phospho-NF-κBp65. Partial blockage of TLR2 by CD282 weakened the inhibitory effects of C. butyricum supernatant on the above pathway components. Those component levels were still inhibited by C. butyricum supernatant after Pam3CSK4 activation of TLR2. In summary, C. butyricum supernatant can inhibit the TLR2/MyD88/NF-κB signaling pathway and the expression of RORγt in HCT-116 cells. These effects are at least partly achieved through inhibition of TLR2.
Collapse
Affiliation(s)
- Ying Xie
- Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Linyan Zhou
- Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Hui Li
- Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yan Li
- Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
8
|
Yi Z, Sadiq FA, Wang H, Zhao J, Zhang H, Lu W, Chen W. Establishment and evaluation of a method for efficient screening of Clostridium butyricum. Folia Microbiol (Praha) 2020; 65:917-924. [PMID: 32621129 DOI: 10.1007/s12223-020-00803-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/03/2020] [Indexed: 11/25/2022]
Abstract
At present, the traditional methods for the screening of Clostridium butyricum are not sufficiently selective and efficient. Therefore, it is necessary to establish a targeted and efficient screening method for the detection of C. butyricum. Bioinformatics was used in this study to find C. butyricum specific genes, and species-specific primers were designed based on the conserved regions of the targeted genes, followed by optimization of the PCR conditions. Methodological evaluation was carried out, and the results were compared with the traditional screening method based on Trypticase Sulfite Neomycin (TSN) selective medium. A high-efficiency PCR screening method, targeting C. butyricum species-specific primers, was established. The method was confirmed to have high specificity and sensitivity towards C. butyricum cut-off CFU 103. Compared with the traditional method, the screening success rate of C. butyricum strains increased from 0.61 to 81.91%. The PCR screening method could quickly and accurately detect C. butyricum in samples and dramatically improve screening efficiency.
Collapse
Affiliation(s)
- Zhi Yi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Faizan A Sadiq
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, 214122, Jiangsu Province, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, 214122, Jiangsu Province, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
- Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| |
Collapse
|
9
|
Xie Y, Zhou L, Yao X, Li Y. Protective Effects of Clostridium Butyricum in a Murine Model of Dextran Sodium Sulfate-Induced Colitis That Involve Inhibition of the TLR2 Signaling Pathway and T Helper 17 Cells. Am J Med Sci 2020; 360:176-191. [PMID: 32553747 DOI: 10.1016/j.amjms.2020.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/08/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND This study aimed to investigate the role of Clostridium butyricum (C. butyricum) in conjunction with the Toll-like receptor2 (TLR2) signaling pathway and T helper 17 (Th17) cells in dextran sodium sulfate (DSS)-induced colitis in mice. METHODS Forty 8-week-old BALB/c mice were randomly divided into 5 groups of 8 mice for 7 days: control, DSS (5% DSS), DSS+C. butyricum (1 × 109 CFU), DSS+C. butyricum (1 × 108 CFU) and DSS+C. butyricum (1 × 107 CFU) groups. We assessed the disease activity index (DAI) and histological damage scores. The expression levels of TLR2, myeloid differentiation factor 88 (MyD88), nuclear factor kappa-B p65 (NF-κBp65), interleukin (IL) 17 (IL17), IL23 and retineic acid receptor related orphan nuclear receptor gamma t (RORγt) were determined through immunohistochemical staining, western blot and quantitative real-time PCR (qRT-PCR). The expression levels of CD3+CD4+IL17+ cells in peripheral blood were measured by flow cytometry. RESULTS C. butyricum dose-dependently decreased DAI and histological damage scores in DSS mice and down-regulated the mRNA and protein levels of TLR2, MyD88 and NF-κBp65 in mouse colon tissue (all P < 0.05). In addition, C. butyricum dose-dependently decreased the levels of CD3+CD4+IL17+ cells in peripheral blood and down-regulated the mRNA and protein levels of IL17, IL23 and RORγt in mouse colon tissue (all P < 0.05). Moreover, the effect of C. butyricum on TLR2 was positively correlated with IL17, IL23 and RORγt. CONCLUSIONS C. butyricum exerts a dose-dependently protective effect on acute intestinal inflammation induced by DSS in mice, by inhibiting the TLR2 signaling pathway, down-regulating the expression of IL23 and RORγt, and inhibiting the secretion of IL17.
Collapse
Affiliation(s)
- Ying Xie
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Linyan Zhou
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xinjie Yao
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Yan Li
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
10
|
Gong J, Chehrazi-Raffle A, Placencio-Hickok V, Guan M, Hendifar A, Salgia R. The gut microbiome and response to immune checkpoint inhibitors: preclinical and clinical strategies. Clin Transl Med 2019; 8:9. [PMID: 30887236 PMCID: PMC6423251 DOI: 10.1186/s40169-019-0225-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 03/08/2019] [Indexed: 02/06/2023] Open
Abstract
There is growing interest in identifying predictive biomarkers for inhibitors of programmed cell death protein 1 receptor (PD-1), programmed death ligand 1 (PD-L1), and cytotoxic T-lymphocyte associated protein 4 (CTLA-4). Given the links between the stool microbiota, anticancer immunosurveillance, and general health, the composition of the gut microbiome has recently undergone investigation as a biomarker for immunotherapy. In this review, we highlight published results from preclinical and clinical studies to date supporting a relationship between the gut microbiome and antitumor efficacy of immune checkpoint inhibitors. Despite the promising and hypothesis-generating findings that have been produced in this arena to date, there remain some inconsistencies amongst present data that may need to be resolved to contribute to further development. Among these, a better understanding of the immunomodulatory function of the microbiome, standardization in sampling, sequencing techniques, and data analysis, and ensuring uniformity across various aspects of study design are warranted in conducting future prospective studies seeking to validate the gut microbiome as a potential biomarker of response to checkpoint blockade.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medicine, Division of Hematology/Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Alexander Chehrazi-Raffle
- Department of Internal Medicine, Harbor-UCLA Medical Center, 1000 W Carson St, Torrance, CA 90509 USA
| | - Veronica Placencio-Hickok
- Department of Medicine, Division of Hematology/Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Michelle Guan
- Department of Medicine, Division of Hematology/Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Andrew Hendifar
- Department of Medicine, Division of Hematology/Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Ravi Salgia
- Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Building 51, Room 101, 1500 E Duarte St, Duarte, CA 91010 USA
| |
Collapse
|
11
|
Li MG, Liu XY, Liu ZQ, Hong JY, Liu JQ, Zhou CJ, Hu TY, Xiao XJ, Ran PX, Zheng PY, Liu ZG, Yang PC. Bcl2L12 Contributes to Th2-Biased Inflammation in the Intestinal Mucosa by Regulating CD4+ T Cell Activities. THE JOURNAL OF IMMUNOLOGY 2018; 201:725-733. [DOI: 10.4049/jimmunol.1800139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
|
12
|
Galacto-Oligosaccharide/Polidextrose Enriched Formula Protects against Respiratory Infections in Infants at High Risk of Atopy: A Randomized Clinical Trial. Nutrients 2018; 10:nu10030286. [PMID: 29494489 PMCID: PMC5872704 DOI: 10.3390/nu10030286] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/20/2018] [Accepted: 02/24/2018] [Indexed: 01/06/2023] Open
Abstract
Background: Early nutrition affects the risk of atopy and infections through modifications of intestinal microbiota. The Prebiotics in the Prevention of Atopy (PIPA) study was a 24-month randomised, double-blind, placebo-controlled trial. It aimed to evaluate the effects of a galacto-oligosaccharide/polydextrose (GOS/PDX)-formula (PF) on atopic dermatitis (AD) and common infections in infants who were born to atopic parents and to investigate the relationship among early nutrition, gut microbiota and clinical outcomes. Methods: A total of 201 and 199 infants were randomized to receive a PF and standard formula (SF), respectively; 140 infants remained on exclusive breastfeeding (BF). Results: The cumulative incidence of AD and its intensity and duration were not statistically different among the three groups. The number of infants with at least one episode of respiratory infection (RI) and the mean number of episodes until 48 weeks of age were significantly lower in the PF group than in the SF group. The number of patients with recurrent RIs and incidence of wheezing lower RIs until 96 weeks were lower in the PF group than the SF group, but similar to the BF group. Bifidobacteria and Clostridium cluster I colonization increased over time in the PF group but decreased in the SF and BF groups. Bifidobacteria had a protective role in RIs, whereas Clostridium cluster I was associated with atopy protection. Conclusion: The early administration of PF protects against RIs and mediates a species-specific modulation of the intestinal microbiota. Trial registration: clinicaltrial.gov Identifier: NCT02116452.
Collapse
|
13
|
|