1
|
Ahrend H, Buchholtz A, Stope MB. Microbiome and Mucosal Immunity in the Intestinal Tract. In Vivo 2025; 39:17-24. [PMID: 39740876 PMCID: PMC11705094 DOI: 10.21873/invivo.13801] [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: 08/16/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
The human bowel is exposed to numerous biotic and abiotic external noxious agents. Accordingly, the digestive tract is frequently involved in malfunctions within the organism. Together with the commensal intestinal flora, it regulates the immunological balance between inflammatory defense processes and immune tolerance. Pathological changes in this system often cause chronic inflammatory bowel diseases including Crohn's disease and ulcerative colitis. This review article highlights the complex interaction between commensal microorganisms, the intestinal microbiome, and the intestinal epithelium-localized local immune system. The main functions of the human intestinal microbiome include (i) protection against pathogenic microbial colonization, (ii) maintenance of the barrier function of the intestinal epithelium, (iii) degradation and absorption of nutrients and (iv) active regulation of the intestinal immunity. The local intestinal immune system consists primarily of macrophages, antigen-presenting cells, and natural killer cells. These cells regulate the commensal intestinal microbiome and are in turn regulated by signaling factors of the epithelial cells and the microbiome. Deregulated immune responses play an important role and can lead to both reduced activity of the commensal microbiome and pathologically increased activity of harmful microorganisms. These aspects of chronic inflammatory bowel disease have become the focus of attention in recent years. It is therefore important to consider the immunological-microbial context in both the diagnosis and treatment of inflammatory bowel diseases. A promising holistic approach would include the most comprehensive possible diagnosis of the immune and microbiome status of the patient, both at the time of diagnostics and during therapy.
Collapse
Affiliation(s)
- Hannes Ahrend
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Anja Buchholtz
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, Research Laboratories, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Gu W, Zhang H, Zhang Z, Xu M, Li X, Han Z, Fu X, Li X, Wang X, Zhang C. Continuous Oral Administration of the Superantigen Staphylococcal Enterotoxin C2 Activates Intestinal Immunity and Modulates the Gut Microbiota in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405039. [PMID: 39248343 PMCID: PMC11538665 DOI: 10.1002/advs.202405039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/28/2024] [Indexed: 09/10/2024]
Abstract
Staphylococcal Enterotoxin C2 (SEC2), a classical superantigen, is an antitumor immunotherapy agent. However, the injectable formulation of SEC2 limits its clinical application. Here, it is reported that oral administration of SEC2 activates the intestinal immune system and benefits intestinal health in a mouse model. These results indicate that intact SEC2 is detected in the stomach, intestine, and serum after oral administration. Continuous oral administration of SEC2 activates immune cells in gut-associated lymphoid tissues, promoting extensive differentiation and proliferation of CD4+ and CD8+ T cells and CD19+ B cells, leading to increased production of cytokines and secretory immunoglobulin A. SEC2 also enhances intestinal barrier function, as demonstrated by an increased villus length/crypt depth ratio and elevated expression of mucins and tight junction proteins. Additionally, SEC2 indirectly influenced gut microbiota, reinforcing potential probiotics and short-chain fatty acid synthesis. Enhanced differentiation of T and B cells in the spleen, coupled with elevated serum interleukin-2 levels, suggests systemic immune enhancement following oral administration of SEC2. These findings provide a scientific basis for the development of SEC2 as an oral immunostimulant for immune enhancement and anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wu Gu
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- University of Chinese Academy of SciencesNo.1 Yanqihu East Rd, Huairou DistrictBeijing101408P. R. China
| | - Huiwen Zhang
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Best Health (Guangdong) Bio‐Technology Co., Ltd.Center Building, Minke Park, Xinhui Economic Development ZoneJiangmen529100P. R. China
| | - Zhichun Zhang
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- University of Chinese Academy of SciencesNo.1 Yanqihu East Rd, Huairou DistrictBeijing101408P. R. China
| | - Mingkai Xu
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
| | - Xiang Li
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
| | - Zhiyang Han
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- University of Chinese Academy of SciencesNo.1 Yanqihu East Rd, Huairou DistrictBeijing101408P. R. China
| | - Xuanhe Fu
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
- Department of ImmunologyShenyang Medical CollegeNo. 146 Huanghe North StreetShenyang110034P. R. China
| | - Xu Li
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
| | - Xiujuan Wang
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
| | - Chenggang Zhang
- Institute of Applied EcologyChinese Academy of Sciences72 WenHua RoadShenyang110016P. R. China
- Key Laboratory of Superantigen Research of Liao Ning ProvinceNo. 72 WenHua RoadShenyang110016P. R. China
| |
Collapse
|
3
|
Xia J, Cui Y, Guo Y, Liu Y, Deng B, Han S. The Function of Probiotics and Prebiotics on Canine Intestinal Health and Their Evaluation Criteria. Microorganisms 2024; 12:1248. [PMID: 38930630 PMCID: PMC11205510 DOI: 10.3390/microorganisms12061248] [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: 06/03/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Maintaining homeostasis within the intestinal microbiota is imperative for assessing the health status of hosts, and dysbiosis within the intestinal microbiota is closely associated with canine intestinal diseases. In recent decades, the modulation of canine intestinal health through probiotics and prebiotics has emerged as a prominent area of investigation. Evidence indicates that probiotics and prebiotics play pivotal roles in regulating intestinal health by modulating the intestinal microbiota, fortifying the epithelial barrier, and enhancing intestinal immunity. This review consolidates literature on using probiotics and prebiotics for regulating microbiota homeostasis in canines, thereby furnishing references for prospective studies and formulating evaluation criteria.
Collapse
Affiliation(s)
| | | | | | | | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.X.); (Y.C.); (Y.G.); (Y.L.)
| | - Sufang Han
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.X.); (Y.C.); (Y.G.); (Y.L.)
| |
Collapse
|
4
|
Zhao M, Liang X, Meng Y, Lu H, Lin K, Gong P, Liu T, Yi H, Pan J, Zhang Y, Zhang Z, Zhang L. Probiotics induce intestinal IgA secretion in weanling mice potentially through promoting intestinal APRIL expression and modulating the gut microbiota composition. Food Funct 2024; 15:4862-4873. [PMID: 38587236 DOI: 10.1039/d4fo00962b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Intestinal infections are strongly associated with infant mortality, and intestinal immunoglobulin A (IgA) is important to protect infants from intestinal infections after weaning. This study aims to screen probiotics that can promote the production of intestinal IgA after weaning and further explore their potential mechanisms of action. In this study, probiotics promoting intestinal IgA production were screened in weanling mouse models. The results showed that oral administration of Bifidobacterium bifidum (B. bifidum) FL228.1 and Bifidobacterium bifidum (B. bifidum) FL276.1 significantly enhanced IgA levels in the small intestine and upregulated the expression of a proliferation-inducing ligand (APRIL) and its upstream regulatory factor toll-like receptor 4 (TLR4). Furthermore, B. bifidum FL228.1 upregulated the relative abundance of Lactobacillus, while B. bifidum FL276.1 increased the relative abundance of Marvinbryantia and decreased Mucispirillum, further elevating intestinal IgA levels. In summary, B. bifidum FL228.1 and B. bifidum FL276.1 can induce IgA production in the intestinal tract of weanling mice by promoting intestinal APRIL expression and mediating changes in the gut microbiota, thus playing a significant role in enhancing local intestinal immunity in infants.
Collapse
Affiliation(s)
- Maozhen Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Xi Liang
- College of Public Health, Qingdao University, Qingdao, 266000, China
| | - Yang Meng
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Haiyan Lu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Kai Lin
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Pimin Gong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Tongjie Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Jiancun Pan
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihar, 161000, China
| | - Yongjiu Zhang
- Heilongjiang Feihe Dairy Co., Ltd., Qiqihar, 161000, China
| | - Zhe Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China.
| |
Collapse
|
5
|
Haneishi Y, Treppiccione L, Maurano F, Luongo D, Miyamoto J, Rossi M. High Fat Diet-Wheat Gliadin Interaction and its Implication for Obesity and Celiac Disease Onset: In Vivo Studies. Mol Nutr Food Res 2024; 68:e2300779. [PMID: 38632845 DOI: 10.1002/mnfr.202300779] [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: 11/03/2023] [Revised: 03/03/2024] [Indexed: 04/19/2024]
Abstract
The intestinal immune system plays a crucial role in obesity and insulin resistance. An altered intestinal immunity is associated with changes to the gut microbiota, barrier function, and tolerance to luminal antigens. Lipid metabolism and its unbalance can also contribute to acute and chronic inflammation in different conditions. In celiac disease (CD), the serum phospholipid profile in infants who developed CD is dramatically different when compared to that of infants at risk of CD not developing the disease. In a mouse model of gluten sensitivity, oral wheat gliadin challenge in connection with inhibition of the metabolism of arachidonic acid, an omega-6 polyunsaturated fatty acid, specifically induces the enteropathy. Recent evidence suggests that gluten may play a role also for development of life-style related diseases in populations on a high fat diet (HFD). However, the mechanisms behind these effects are not yet understood. Exploratory studies in mice feed HFD show that wheat gliadin consumption affects glucose and lipid metabolic homeostasis, alters the gut microbiota, and the immune cell profile in liver.
Collapse
Affiliation(s)
- Yuri Haneishi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | | | - Francesco Maurano
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| | - Diomira Luongo
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mauro Rossi
- Institute of Food Sciences, CNR, via Roma 64, Avellino, 83100, Italy
| |
Collapse
|
6
|
Zhu Y, Tang H, Xie W, Chen S, Zeng H, Lan C, Guan J, Ma C, Yang X, Wang Q, Wei L, Zhang Z, Yu X. The multilevel extensive diversity across the cynomolgus macaque captured by ultra-deep adaptive immune receptor repertoire sequencing. SCIENCE ADVANCES 2024; 10:eadj5640. [PMID: 38266093 PMCID: PMC10807814 DOI: 10.1126/sciadv.adj5640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
The extent to which AIRRs differ among and within individuals remains elusive. Via ultra-deep repertoire sequencing of 22 and 25 tissues in three cynomolgus macaques, respectively, we identified 84 and 114 novel IGHV and TRBV alleles, confirming 72 (85.71%) and 100 (87.72%) of them. The heterogeneous V gene usage patterns were influenced, in turn, by genetics, isotype (for BCRs only), tissue group, and tissue. A higher proportion of intragroup shared clones in the intestinal tissues than those in other tissues suggests a close intra-intestinal adaptive immunity network. Significantly higher mutation burdens in the public clones and the inter-tissue shared IgM and IgD clones indicate that they might target the shared antigens. This study reveals the extensive heterogeneity of the AIRRs at various levels and has broad fundamental and clinical implications. The data generated here will serve as an invaluable resource for future studies on adaptive immunity in health and diseases.
Collapse
Affiliation(s)
- Yan Zhu
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haipei Tang
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Wenxi Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sen Chen
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huikun Zeng
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Division of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Chunhong Lan
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junjie Guan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Cuiyu Ma
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiujia Yang
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Qilong Wang
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Zhenhai Zhang
- Center for Precision Medicine, Medical Research Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, China
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Division of Nephrology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
7
|
Campidelli C, Bruxelle JF, Collignon A, Péchiné S. Immunization Strategies Against Clostridioides difficile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:117-150. [PMID: 38175474 DOI: 10.1007/978-3-031-42108-2_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is an important healthcare but also a community-associated disease. CDI is considered a public health threat and an economic burden. A major problem is the high rate of recurrences. Besides classical antibiotic treatments, new therapeutic strategies are needed to prevent infection, to treat patients, and to prevent recurrences. If fecal transplantation has been recommended to treat recurrences, another key approach is to elicit immunity against C. difficile and its virulence factors. Here, after a summary concerning the virulence factors, the host immune response against C. difficile, and its role in the outcome of disease, we review the different approaches of passive immunotherapies and vaccines developed against CDI. Passive immunization strategies are designed in function of the target antigen, the antibody-based product, and its administration route. Similarly, for active immunization strategies, vaccine antigens can target toxins or surface proteins, and immunization can be performed by parenteral or mucosal routes. For passive immunization and vaccination as well, we first present immunization assays performed in animal models and second in humans and associated clinical trials. The different studies are presented according to the mode of administration either parenteral or mucosal and the target antigens and either toxins or colonization factors.
Collapse
Affiliation(s)
- Camille Campidelli
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Jean-François Bruxelle
- CIRI-Centre International de Recherche en Infectiologie, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1111, CNRS UMR5308, ENS Lyon, Lyon, France
| | - Anne Collignon
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Severine Péchiné
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| |
Collapse
|
8
|
Cela L, Brindisi G, Gravina A, Pastore F, Semeraro A, Bringheli I, Marchetti L, Morelli R, Cinicola B, Capponi M, Gori A, Pignataro E, Piccioni MG, Zicari AM, Anania C. Molecular Mechanism and Clinical Effects of Probiotics in the Management of Cow's Milk Protein Allergy. Int J Mol Sci 2023; 24:9781. [PMID: 37372929 DOI: 10.3390/ijms24129781] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Cow's milk protein allergy (CMPA) is the most common food allergy (FA) in infancy, affecting approximately 2% of children under 4 years of age. According to recent studies, the increasing prevalence of FAs can be associated with changes in composition and function of gut microbiota or "dysbiosis". Gut microbiota regulation, mediated by probiotics, may modulate the systemic inflammatory and immune responses, influencing the development of allergies, with possible clinical benefits. This narrative review collects the actual evidence of probiotics' efficacy in the management of pediatric CMPA, with a specific focus on the molecular mechanisms of action. Most studies included in this review have shown a beneficial effect of probiotics in CMPA patients, especially in terms of achieving tolerance and improving symptoms.
Collapse
Affiliation(s)
- Ludovica Cela
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Giulia Brindisi
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Gravina
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesca Pastore
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Antonio Semeraro
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Ivana Bringheli
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Lavinia Marchetti
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Rebecca Morelli
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Bianca Cinicola
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Martina Capponi
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandra Gori
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Elia Pignataro
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Grazia Piccioni
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| | - Caterina Anania
- Department of Maternal Infantile and Urological Science, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
9
|
Seguel M, Budischak SA, Jolles AE, Ezenwa VO. Helminth-associated changes in host immune phenotype connect top-down and bottom-up interactions during co-infection. Funct Ecol 2023; 37:860-872. [PMID: 37214767 PMCID: PMC10195069 DOI: 10.1111/1365-2435.14237] [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: 06/19/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022]
Abstract
1. Within-host parasite interactions can be mediated by the host and changes in host phenotypes often serve as indicators of the presence or intensity of parasite interactions. 2. Parasites like helminths induce a range of physiological, morphological, and immunological changes in hosts that can drive bottom-up (resource-mediated) or top-down (immune-mediated) interactions with co-infecting parasites. Although top-down and bottom-up interactions are typically studied in isolation, the diverse phenotypic changes induced by parasite infection may serve as a useful tool for understanding if, and when, these processes act in concert. 3. Using an anthelmintic treatment study of African buffalo (Syncerus caffer), we tracked changes in host immunological and morphological phenotypes during helminth-coccidia co-infection to investigate their role in driving independent and combinatorial bottom-up and top-down parasite interactions. We also examined repercussions for host fitness. 4. Clearance of a blood-sucking helminth, Haemonchus, from the host gastrointestinal tract induced a systemic Th2 immune phenotype, while clearance of a tissue-feeding helminth, Cooperia, induced a systemic Th1 phenotype. Furthermore, the Haemonchus-associated systemic Th2 immune phenotype drove simultaneous top-down and bottom-up effects that increased coccidia shedding by changing the immunological and morphological landscapes of the intestine. 5. Higher coccidia shedding was associated with lower host body condition, a lower chance of pregnancy, and older age at first pregnancy, suggesting that coccidia infection imposed significant condition and reproductive costs on the host. 6. Our findings suggest that top-down and bottom-up interactions may commonly co-occur and that tracking key host phenotypes that change in response to infection can help uncover complex pathways by which parasites interact.
Collapse
Affiliation(s)
- Mauricio Seguel
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Sarah A. Budischak
- W.M. Keck Science Department, Claremont McKenna, Pitzer, and Scripps Colleges, Claremont, USA
| | - Anna E. Jolles
- Department of Biomedical Sciences and Department of Integrative Biology, Oregon State University, Corvallis, OR, USA
| | - Vanessa O. Ezenwa
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
Li Y, Li H, Wang R, Yu Y, Liu X, Tian Z. Protective effect of sodium butyrate on intestinal barrier damage and uric acid reduction in hyperuricemia mice. Biomed Pharmacother 2023; 161:114568. [PMID: 36948133 DOI: 10.1016/j.biopha.2023.114568] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023] Open
Abstract
PURPOSE The goal of this study was to examine the role of sodium butyrate in preserving the intestinal mucosal barrier and reducing hyperuricemia (HUA). METHODS First, we established a mouse model of HUA via intraperitoneal injection of potassium oxonate together with a yeast-rich diet to detect the levels of serum uric acid (UA) and fecal short-chain fatty acids (SCFAs). Then, in vitro, different concentrations of UA and sodium butyrate (NaB) were used to treat LS174T and Caco2 cells. The effects of UA and NaB on the gut barrier were determined based on the expression levels of MUC2, ZO-1, and Occludin.Finally, C57BL/6 mice were used to model HUA, and these mice were administered 200 mg·kg-1·d-1 NaB by gavage to counter the HUA. The effect of NaB on HUA in the intestinal tract was elucidated by determining serum UA levels, inflammatory parameters, epithelial barrier integrity, and via histological analysis. RESULTS The data showed that the content of fecal SCFAs in HUA mice decreased. Additionally, in LS174T and Caco2 cells, NaB reversed the decrease of ZO-1, Occludin, and MUC2 protein expression caused by high UA levels. Furthermore, NaB decreased serum UA of HUA mice, and reversed both the decreased expression of MUC2, ZO-1, Occludin, and ABCG2 proteins and the increased level of inflammatory factors in the intestinal tissues of these mice. CONCLUSION The HUA mouse model showed intestinal barrier damage. NaB protected the intestinal barrier of HUA mice and reduced the serum UA level.
Collapse
Affiliation(s)
- Yukun Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hanqing Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Rong Wang
- Plastic Surgery Institute of Weifang Medical University, Weifang, China
| | - Yajie Yu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Yuan Y, Sepúlveda MS, Bi B, Huang Y, Kong L, Yan H, Gao Y. Acute polyethylene microplastic (PE-MPs) exposure activates the intestinal mucosal immune network pathway in adult zebrafish (Danio rerio). CHEMOSPHERE 2023; 311:137048. [PMID: 36419273 DOI: 10.1016/j.chemosphere.2022.137048] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/03/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Polyethylene is one of the most important plastic types with the highest consumption in the world. Plastics are prone to photodegradation and turn into microplastics, which are magnified as they move across trophic levels. Microplastics would be able to penetrate into lymph even cross cell membranes, causing harm to the lymphatic and/or circulatory systems, accumulating in secondary organs, and impacting the immune system and cell health. The objective of this study was to test that the activation of the intestinal immune network might be caused by disruption of intestinal microbiota after exposure to different polyethylene microplastics (PE-MPs) concentrations (1, 10, 100, and 1000 μg/mL) in adult zebrafish (Danio rerio) for 7 days. The concentrations of PE-MPs (100 and 1000 μg/mL) exposure decreased the goblet cell coverage. The intestinal microbial diversity index (Shannon and Simpson) was increased at 100 and 1000 μg/mL PE-MPs concentrations. The relative abundance of intestinal dominant microbiota phylum Proteobacteria and Actinobacteria increased significantly (P < 0.05); however, phylum Fusobacteria decreased significantly (P < 0.05). The relative abundance of intestinal microbiota at level of genera showed varying degrees of elevation such as Acinetobacter (6.31-fold), Plesiomonas (4.80-fold), Flavobacterium (10.54-fold) and Pseudomonas (5.17-fold) in 1000 μg/mL PE-MPs. Intestinal innate immunity-complement C3 and C4 content first increased and then declined in a dose-dependent manner. Expression of genes from the intestinal immune network for mucosal immunoglobulin production were increased also in a dose-dependent manner. The expression of immune-related genes (pigr, il10 and ighv4-5) were positively correlated with the relative abundance of genera Plesiomonas. In conclusion, PE-MPs increase the infection probability in the intestinal mucosa by altering the abundance of intestinal dominant microbiota at the level of phylum. PE-MPs exposure activated the intestinal immune network pathway for mucosal immunoglobulin production at a concentration of 100 or 1000 μg/mL for 7 days.
Collapse
Affiliation(s)
- Yin Yuan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Marisol S Sepúlveda
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN 47907, United States
| | - Baoliang Bi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory for Plateau Fishery Resources Conservation and Sustainable Utilization of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Yadong Huang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Lingfu Kong
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory for Plateau Fishery Resources Conservation and Sustainable Utilization of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Hui Yan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory for Plateau Fishery Resources Conservation and Sustainable Utilization of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China
| | - Yu Gao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Key Laboratory for Plateau Fishery Resources Conservation and Sustainable Utilization of Yunnan Province, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
12
|
Wang Y, Takano T, Zhou Y, Wang R, Toshimitsu T, Sashihara T, Tanokura M, Miyakawa T, Nakajima-Adachi H, Hachimura S. Orally administered Lactiplantibacillus plantarum OLL2712 decreased intestinal permeability, especially in the ileum: Ingested lactic acid bacteria alleviated obesity-induced inflammation by collaborating with gut microbiota. Front Immunol 2023; 14:1123052. [PMID: 36911680 PMCID: PMC9995389 DOI: 10.3389/fimmu.2023.1123052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Chronic inflammation caused by dietary obesity has been considered to induce lifestyle-related diseases and functional ingredients with anti-inflammatory effects are attracting attention. Although multiple studies on obesity had proved the anti-inflammatory effects of ingestion of lactic acid bacteria (LAB) and other functional ingredients on adipose tissue, the precise effects on the intestine, especially on the individual intestinal segments have not been made clear. In this study, we elucidated the mechanisms of Lactiplantibacillus plantarum (basonym: Lactobacillus plantarum) OLL2712 in suppressing obesity-induced inflammation using high fat diet (HFD)-fed mice obesity model. Methods We orally administered heat-treated LAB to HFD-fed mice model, and investigated the inflammatory changes in adipose tissue and intestinal immune cells. We also analyzed gut microbiota, and evaluated the inflammation and permeability of the duodenum, jejunum, ileum and colon; four intestinal segments differing in gut bacteria composition and immune response. Results After 3-week LAB administration, the gene expression levels of proinflammatory cytokines were downregulated in adipose tissue, colon, and Peyer's patches (PP)-derived F4/80+ cells. The LAB treatment alleviated obesity-related gut microbiota imbalance. L. plantarum OLL2712 treatment helps maintain intestinal barrier function, especially in the ileum, possibly by preventing ZO-1 and Occludin downregulation. Discussion Our results suggest that the oral administration of the LAB strain regulated the gut microbiota, suppressed intestinal inflammation, and improved the gut barrier, which could inhibit the products of obesity-induced gut dysbiosis from translocating into the bloodstream and the adipose tissue, through which the LAB finally alleviated the inflammation caused by dietary obesity. Barrier improvement was observed, especially in the ileum, suggesting collaborative modulation of the intestinal immune responses by ingested LAB and microbiota.
Collapse
Affiliation(s)
- Yimei Wang
- Research Center for Food Safety, The University of Tokyo, Tokyo, Japan
| | - Tomohiro Takano
- Research Center for Food Safety, The University of Tokyo, Tokyo, Japan
| | - Yingyu Zhou
- Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | - Rong Wang
- Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | | | | | - Masaru Tanokura
- Research Center for Food Safety, The University of Tokyo, Tokyo, Japan.,Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, The University of Tokyo, Tokyo, Japan.,Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | - Satoshi Hachimura
- Research Center for Food Safety, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
DuPont HL, Jiang ZD, Alexander AS, DuPont AW, Brown EL. Intestinal IgA-Coated Bacteria in Healthy- and Altered-Microbiomes (Dysbiosis) and Predictive Value in Successful Fecal Microbiota Transplantation. Microorganisms 2022; 11:microorganisms11010093. [PMID: 36677385 PMCID: PMC9862469 DOI: 10.3390/microorganisms11010093] [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: 11/21/2022] [Revised: 12/17/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
IgA-coated bacteria in the gut (IgA-biome) provide a homeostatic function in healthy people through inhibition of microbial invaders and by protecting the epithelial monolayer of the gut. The laboratory methods used to detect this group of bacteria require flow cytometry and DNA sequencing (IgA-Seq). With dysbiosis (reduced diversity of the microbiome), the IgA-biome also is impaired. In the presence of enteric infection, oral vaccines, or an intestinal inflammatory disorder, the IgA-biome focuses on the pathogenic bacteria or foreign antigens, while in other chronic diseases associated with dysbiosis, the IgA-biome is reduced in capacity. Fecal microbiota transplantation (FMT), the use of fecal product from well-screened, healthy donors administered to patients with dysbiosis, has been successful in engrafting the intestine with healthy microbiota and metabolites leading to improve health. Through FMT, IgA-coated bacteria have been transferred to recipients retaining their immune coating. The IgA-biome should be evaluated in FMT studies as these mucosal-associated bacteria are more likely to be associated with successful transplantation than free luminal organisms. Studies of the microbiome pre- and post-FMT should employ metagenomic methods that identify bacteria at least at the species level to better identify organisms of interest while allowing comparisons of microbiota data between studies.
Collapse
Affiliation(s)
- Herbert L. DuPont
- Center for Infectious Diseases, Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX 77030, USA
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
- Kelsey Research Foundation, Houston, TX 77005, USA
- Correspondence: ; Tel.: +1-713-500-9366
| | - Zhi-Dong Jiang
- Center for Infectious Diseases, Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX 77030, USA
| | | | - Andrew W. DuPont
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
| | - Eric L. Brown
- Center for Infectious Diseases, Division of Epidemiology, Human Genetics and Environmental Sciences, University of Texas School of Public Health, Houston, TX 77030, USA
| |
Collapse
|
14
|
Zheng H, Zhang C, Wang Q, Feng S, Fang Y, Zhang S. The impact of aging on intestinal mucosal immune function and clinical applications. Front Immunol 2022; 13:1029948. [PMID: 36524122 PMCID: PMC9745321 DOI: 10.3389/fimmu.2022.1029948] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022] Open
Abstract
Immune cells and immune molecules in the intestinal mucosa participate in innate and adaptive immunity to maintain local and systematic homeostasis. With aging, intestinal mucosal immune dysfunction will promote the emergence of age-associated diseases. Although there have been a number of studies on the impact of aging on systemic immunity, relatively fewer studies have been conducted on the impact of aging on the intestinal mucosal immune system. In this review, we will briefly introduce the impact of aging on the intestinal mucosal barrier, the impact of aging on intestinal immune cells as well as immune molecules, and the process of interaction between intestinal mucosal immunity and gut microbiota during aging. After that we will discuss potential strategies to slow down intestinal aging in the elderly.
Collapse
Affiliation(s)
- Han Zheng
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qianqian Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyan Feng
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Fang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China,*Correspondence: Shuo Zhang,
| |
Collapse
|
15
|
Han B, Ma Y, Liu Y. Fucoxanthin Prevents the Ovalbumin-Induced Food Allergic Response by Enhancing the Intestinal Epithelial Barrier and Regulating the Intestinal Flora. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10229-10238. [PMID: 35947424 DOI: 10.1021/acs.jafc.2c04685] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This study aimed to determine whether fucoxanthin alleviated ovalbumin (OVA)-induced food allergy (FA) and explored the possible mechanisms. The results indicated that supplementation with fucoxanthin at 10.0-20.0 mg/kg per day for 7 weeks inhibited food anaphylaxis and the production of immunoglobulin (Ig) E, IgG, histamine, and related cytokines while alleviating allergic symptoms in sensitized mice. Fucoxanthin enhanced the intestinal epithelial barrier by up-regulating tight junction (TJ) protein expression and promoting regenerating islet-derived protein III-gamma (RegIIIγ) and secretory IgA (sIgA) secretion. In addition, fucoxanthin induced the secretion of anti-inflammatory factors (interleukin (IL)-10 and transforming growth factor β (TGF-β)) by regulatory T (Treg) cells and decreased the pro-inflammatory factor levels (IL-4, tumor necrosis factor-α (TNF-α), IL-17, and IL-1β), ameliorating intestinal inflammation. Compared with the model group, beneficial bacteria, such as Lactobacillaceae, increased in the intestinal flora, while pathogenic bacteria like Helicobacteraceae, Desulfovibrionaceae, and Streptococcaceae decreased. Therefore, fucoxanthin may effectively prevent FA by enhancing the intestinal epithelial barrier and reshaping the intestinal flora.
Collapse
Affiliation(s)
- Bing Han
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yu Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
- Collaborative Innovation Center of Provincial and Ministerial Co-construction for Marine Food Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
16
|
Ma L, Yu J, Zhang H, Zhao B, Zhang J, Yang D, Luo F, Wang B, Jin B, Liu J. Effects of Immune Cells on Intestinal Stem Cells: Prospects for Therapeutic Targets. Stem Cell Rev Rep 2022; 18:2296-2314. [DOI: 10.1007/s12015-022-10347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
|
17
|
Inatomi T, Honma M. Effects of probiotics on loperamide-induced constipation in rats. Sci Rep 2021; 11:24098. [PMID: 34916548 PMCID: PMC8677781 DOI: 10.1038/s41598-021-02931-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 11/24/2021] [Indexed: 12/23/2022] Open
Abstract
The role of probiotics in mitigating constipation, gut immunity, and gut microbiota has not been well studied. We aimed to evaluate the effects of probiotics on loperamide (LP)-induced constipation in Sprague-Dawley rats. Altogether, 150 male Sprague-Dawley rats (age 8 weeks) were used in the experiments following a 12-day acclimatisation period and were randomly divided into three treatment groups (groups 1, 2, and 3). Spastic constipation was induced via oral LP administration (3 mg/kg) for 6 days, 1 h before administering each test compound in groups 1 and 2. A probiotic solution (4 mL/kg body weight) was orally administered once a day for 6 days in group 2. In group 1, a phosphate buffer solution was orally administered once a day for 6 days, 1 h after each LP administration. In group 3, a phosphate buffer solution was orally administered once a day for 6 days. In the probiotic group, faecal parameters improved; faecal n-butyric acid, acetic acid, and IgA concentrations were increased; intestinal transit time was shortened; and disturbance of intestinal microbiota was inhibited. Our findings suggest that this probiotic was useful in improving various symptoms caused by constipation.
Collapse
Affiliation(s)
- Takio Inatomi
- Inatomi Animal Hospital, 1-1-24 Denenchofu, Ota-ku, Tokyo, 145-0071, Japan.
| | - Mihoko Honma
- Kusama Animal Health Laboratory, 2240 Tsunehiro, Kashima-shi, Saga, 849-1301, Japan
| |
Collapse
|
18
|
Cai G, Wusiman A, Gu P, Mao N, Xu S, Zhu T, He J, Liu Z, Wang D. Supplementation of Alhagi honey polysaccharides contributes to the improvement of the intestinal immunity regulating the structure of intestinal flora in mice. Food Funct 2021; 12:9693-9707. [PMID: 34664596 DOI: 10.1039/d1fo01860d] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alhagi honey polysaccharides (AH), a main active component of Alhagi honey, are known to possess excellent pharmacological activities and have been widely used as dietary supplements in traditional Chinese medicine for thousands of years. This study is aimed to investigate the heath effect of AH on murine intestinal mucosal immune function and composition of the gut microbiome. ICR mice received daily intragastric administration of AH (three dosages, 200 mg kg-1, 400 mg kg-1, and 800 mg kg-1) or saline for 7 consecutive days. Results indicated an improvement in the intestinal barrier function through increases in secretory immunoglobulin A (sIgA) and β-defensins. Simultaneously, AH also significantly stimulated IL-2, IL-4, IL-6, IL-10, IL-17, IFN-γ, and TNF-α cytokine secretion as compared to the control samples. Moreover, hematoxylin and eosin staining showed that AH enhanced the number of intraepithelial lymphocytes (IELs) in the small intestine. An obvious increase in the ratio of IgA+ cells of AH-treatment samples in the lamina propria was also detected by immunohistochemical staining. In addition, the CD3+, CD4+ and CD8+ T-cell ratio in mesenteric lymph nodes and Peyer's patches in the AH-treatment was significantly higher than that in the control group. Furthermore, 16S rDNA gene sequencing was used to monitor the dynamic changes in the gut microbiota. The result revealed that AH significantly increased the indexes of Shannon and obviously decreased the indexes of Simpson, suggesting the enhancement of the diversity and richness of the intestinal microbiome. Moreover, AH modulated the gut microbiome via increasing the abundance of probiotics and decreasing the levels of pathogenic bacteria. In summary, these results indicated that AH could be used as a prebiotic to enhance murine intestinal mucosal immunity and to modulate the gut microbiome.
Collapse
Affiliation(s)
- Gaofeng Cai
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Adelijiang Wusiman
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830000, China
| | - Pengfei Gu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Shuwen Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Tianyu Zhu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Jin He
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P.R. China
| |
Collapse
|
19
|
Kurokawa M, Kurokawa R, Hagiwara A, Gonoi W, Harayama S, Koizumi K, Yoshino K, Hishima T, Baba A, Ota Y, Abe O, Takaki Y. CT imaging findings of anti-PD-1 inhibitor-related enterocolitis. Abdom Radiol (NY) 2021; 46:3033-3043. [PMID: 33638055 DOI: 10.1007/s00261-021-02986-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE Immune checkpoint inhibitors promote the antitumor activity of T cells; however, there is a risk of side effects. The aim of this study was to characterize the computed tomography (CT) findings of one such side effect, anti-programmed cell death-1 antibody-related enterocolitis (αPD-1-EC). METHODS This single-institution retrospective study included 21 patients with αPD-1-EC who underwent CT between January 2015 and April 2020. Two board-certified radiologists independently evaluated the CT findings, including the pattern of intestinal wall enhancement, maximum bowel wall thickness, maximum appendiceal diameter, and involvement of enterocolitis in each intestinal segment. Symptoms and their severity were also investigated. RESULTS Pancolitis and skip lesions involving both the rectosigmoid colon and the cecum were found in 9 patients each (42.9%). The rectum was the most frequently involved lesion (18/21, 85.7%), and appendiceal involvement was found in 11 patients (52.4%). The most frequent wall enhancement pattern was the gray pattern (i.e., mild homogeneous enhancement of the thickened bowel wall). The mean maximum diameter of the involved appendix was 9.6 ± 4.5 mm (range 4.5-18 mm). Frequent symptoms included diarrhea (21/21), fever (8/21), and abdominal pain (7/21). Other concomitant immune-related adverse events were found in 6 patients. CONCLUSIONS Pancolitis, skip lesions, and appendiceal involvement were frequent in patients with αPD-1-EC. When combining these characteristic findings with other clinical findings, such as low-grade diarrhea, other concomitant immune-related adverse events, and anti-PD-1 therapy administration, CT may be a useful diagnostic tool for αPD-1-EC.
Collapse
Affiliation(s)
- Mariko Kurokawa
- Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Ryo Kurokawa
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Akifumi Hagiwara
- Department of Radiology, Juntendo University School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Wataru Gonoi
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinjiro Harayama
- Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Koichi Koizumi
- Department of Gastroenterology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Koji Yoshino
- Department of Dermato Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Tsunekazu Hishima
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Akira Baba
- Department of Radiology, The Jikei University School of Medicine, 3-19-18 Nishishinbashi, Minato-ku, Tokyo, 105-0003, Japan
| | - Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, Michigan Medicine, 1500E Medical Center Drive, Ann Arbor, MI, 48109, USA
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasunobu Takaki
- Department of Radiology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| |
Collapse
|
20
|
Zhao L, Hu S, Davila ML, Yang J, Lin YD, Albanese JM, Lo Y, Wang Y, Kennett MJ, Liu Q, Xiong N. Coordinated co-migration of CCR10 + antibody-producing B cells with helper T cells for colonic homeostatic regulation. Mucosal Immunol 2021; 14:420-430. [PMID: 32773769 PMCID: PMC7870723 DOI: 10.1038/s41385-020-0333-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 02/04/2023]
Abstract
In the intestine, IgA antibody-secreting B cells (IgA-ASCs) and helper T cells coordinate to maintain local homeostasis while their dysregulation could lead to development of intestinal inflammatory diseases. However, mechanisms underlying the coordinated localization and function of the B and T cells into the intestine, particularly the colon, are poorly understood. We herein report the first evidence that the gut-homing chemokine receptor CCR10+ IgA-ASCs form conjugates with helper T cells, preferentially regulatory T cells, at their differentiation sites of gut-associated lymphoid organs for their coordinated co-localization into the colon to promote local homeostasis. In CCR10-knockout mice, defective migration of IgA-ASCs also resulted in defective T-cell migration and homeostasis, and development of inflammatory symptoms in the colon. Antigen-specific interaction of CCR10+ IgA-ASCs and T cells is crucial for their homeostatic establishment in the colon. On the other hand, in IgA-knockout mice, preferential expansion of CCR10+ IgG1-ASCs with regulatory functions compensated for CCR10+ IgA-ASCs to help maintain colonic homeostasis. The preferential expansion of specific subclasses of CCR10+ IgG-ASCs with regulatory functions was also found in asymptomatic IgA-deficient patients. These findings suggest coordinated cell migration as a novel mechanism underlying localization and function of B and T cells in colonic homeostatic regulation.
Collapse
Affiliation(s)
- Luming Zhao
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Shaomin Hu
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA,Current address: Department of Pathology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | - Micha L. Davila
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Jie Yang
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Current address: Precision for Medicine-Houston Site, 2575 West Bellfort, Suite 190, Houston, TX 77054, USA
| | - Yang-Ding Lin
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Joseph M. Albanese
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Yungtai Lo
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Yanhua Wang
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Mary J. Kennett
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA
| | - Qiang Liu
- Department of Pathology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East 210th Street, Bronx, NY 10467, USA
| | - Na Xiong
- Center for Molecular Immunology and Infectious Disease, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, 115 Henning Building, University Park, PA 16802, USA.,Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA,Department of Medicine-Division of Dermatology and Cutaneous Surgery, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Correspondence to: Na Xiong, Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229. ; Tel: 2104505362
| |
Collapse
|
21
|
Liu Y, Chen L, Wang L, Xiong Y. Effects of intestinal lymphatic ligation on intestinal immunity in rats with severe acute pancreatitis. FEBS Open Bio 2021; 11:1109-1121. [PMID: 33576136 PMCID: PMC8016124 DOI: 10.1002/2211-5463.13115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/28/2021] [Accepted: 02/10/2021] [Indexed: 01/30/2023] Open
Abstract
Severe acute pancreatitis (SAP) is one of the most common diseases of the gastrointestinal tract, characterized by a complicated pathogenesis, multiple organ failure, and high mortality. The primary aim of the present study was to observe the effect of intestinal lymphatic ligation on intestinal injury and modification in rats with SAP. Male Sprague‐Dawley (SD) rats were randomly divided into: (a) Saline group (SO); (b) SAP group; and (c) SAP + ligation group. We evaluated the effect of mesenteric lymphatic duct ligation on the pancreas and intestine tissue by HE. The histopathology of the pancreas in SAP + ligation rats was alleviated slightly compared with SAP rats, but aggravated in the intestine of SAP + ligation rats. Treatment of mesenteric lymphatic duct ligation resulted in an increase in the levels of tumor necrosis factor (TNF)‐α, interleukin (IL)‐1β, and myeloperoxidase compared with the small intestinal tissues of SAP rats. In addition, the expression of nucleotide‐binding oligomerization domain‐like receptors 3, apoptosis‐associated speck‐like protein containing a caspase recruitment domain (CARD) (ASC), and caspase‐1 in the intestine were higher in the SAP + ligation group. The ratio of Th1/Th2 and regulatory T cells (Tregs) in the mesenteric lymph nodes of the SAP group was lower than those in the SAP + ligation group. The present results indicated that ligation of the mesenteric lymph duct can effectively prevent intestinal inflammatory mediators entering the body through the mesenteric lymph duct, but these mediators assembled in the intestine where they induced an excessive immune response and intestinal injury during SAP.
Collapse
Affiliation(s)
- Yuanqi Liu
- College of Comprehensive Health Management, Xihua University, Chengdu, China
| | - Li Chen
- Department of Pharmacy, The Affiliated T.C.M. Hospital of Southwest Medical University, Luzhou, China
| | - Lulu Wang
- College of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yuxia Xiong
- College of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Yun L, Wu T, Li W, Zhang M. Wheat germ glycoprotein regionally modulates immunosuppressed mouse intestinal immunity function from early life to adulthood. Food Funct 2021; 12:97-106. [PMID: 33305774 DOI: 10.1039/d0fo02754e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Wheat germ glycoprotein (WGP) is widely used due to its nutritional benefits and biological activity. This study evaluated the effects of WGP on intestinal-immunosuppressed mice from early life to adulthood and detected the underlying mechanism. The results revealed that WGP demonstrated no clinical side effects on the body index, serum total IgA level, protein expression and the morphology of intestine in newborn mice. In the phase of life, compared with the cyclophosphamide-treated group (CG), WGP clearly promoted the secretion of sIgA and effectively regulated the cytokine gene (IL-2, IFN-γ, TNF-α, IL-4, IL-6, IL-5, IL-17, and TGF-β1) expression in the intestine. Furthermore, WGP promoted the expression of CD40L and CD40, phosphorylation of IKKα/β and transcription of NF-κB-p65. The data as reported in this present analysis suggest that WGP can improve the intestinal immunity of newborn mice to adulthood via the CD40L-CD40-IKKα/β-NF-κB p65 signaling pathway.
Collapse
Affiliation(s)
- Liyuan Yun
- Tianjin Agricultural University, Tianjin 300191, China.
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Wen Li
- State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Min Zhang
- Tianjin Agricultural University, Tianjin 300191, China. and State Key Laboratory of Food Nutrition and Safety, Food Biotechnology Engineering Research Center of Ministry of Education, College of food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
23
|
Bai Y, Huang F, Zhang R, Ma Q, Dong L, Su D, Chi J, Zhang M. Longan pulp polysaccharide protects against cyclophosphamide-induced immunosuppression in mice by promoting intestinal secretory IgA synthesis. Food Funct 2021; 11:2738-2748. [PMID: 32175536 DOI: 10.1039/c9fo02780g] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aimed to explore the effect of longan pulp polysaccharide (LP) on the systemic immunity and intestinal mucosal immunity of immunosuppressive mice. The synthesis process and secretion of intestinal secretory IgA (SIgA) were investigated. Results showed that LP increased the thymus index, spleen index, and serum IgA level in cyclophosphamide (CTX)-treated mice. SIgA secretion in the intestinal lumen was increased by LP as well. The underlying mechanism comes down to the facts as follow: LP increased intestinal cytokines expression and TGFβRII that is associated with pathways of IgA class switch recombination (CSR). By improving protein expression of mucosal addressin cell-adhesion molecule-1 (MAdCAM-1) and integrin α4β7, LP was beneficial to gut homing of IgA+ plasma cells. LP increased IgA, polymeric immunoglobulin receptor (pIgR), and secretory component (SC) to fortify the SIgA secretion. This study suggested that moderate consumption of LP is helpful for improving systemic immunity and intestinal mucosal immunity via promotion of intestinal SIgA to strengthen the mucosal barrier.
Collapse
Affiliation(s)
- Yajuan Bai
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Fei Huang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Ruifen Zhang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Qin Ma
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Lihong Dong
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Dongxiao Su
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, PR China
| | - Jianwei Chi
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| | - Mingwei Zhang
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, PR China.
| |
Collapse
|
24
|
Kazemi A, Soltani S, Nasri F, Clark CCT, Kolahdouz-Mohammadi R. The effect of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin production: a systematic review and meta-analysis of clinical trials. Int J Food Sci Nutr 2020; 72:632-649. [PMID: 33307904 DOI: 10.1080/09637486.2020.1857710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this systematic review and meta-analysis was to evaluate the effect of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin production. We searched PubMed, Scopus, Web of Science, National Institute of Health Clinical Trials Register, and Cochrane Central Register of Clinical Trials, up to February 2020. All clinical trials that investigated the effects of oral intake of probiotics, parabiotics, synbiotics, fermented foods and other microbial forms on immunoglobulin (Ig)A, IgE, Japanese cedar pollen (JCP)-specific IgE, IgG, and IgM, for a duration of >7 days were included. Fifty-nine studies met the inclusion criteria, of these 54 studies were included in the analysis. The results indicated a significant increase in salivary IgA secretion rate (SMD = 0.21, 95% CI 0.02-0.39), while no significant effect was observed on other Igs. In conclusion, mentioned supplementation induced a small but significant effect on salivary secretion rate of IgA.
Collapse
Affiliation(s)
- Asma Kazemi
- Nutrition Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Nasri
- Department Immunology, School of medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cain C T Clark
- Centre Intelligent Healthcare, Coventry University, Coventry, CV15FB, UK
| | - Roya Kolahdouz-Mohammadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Pietrzak B, Tomela K, Olejnik-Schmidt A, Mackiewicz A, Schmidt M. Secretory IgA in Intestinal Mucosal Secretions as an Adaptive Barrier against Microbial Cells. Int J Mol Sci 2020; 21:ijms21239254. [PMID: 33291586 PMCID: PMC7731431 DOI: 10.3390/ijms21239254] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Secretory IgA (SIgA) is the dominant antibody class in mucosal secretions. The majority of plasma cells producing IgA are located within mucosal membranes lining the intestines. SIgA protects against the adhesion of pathogens and their penetration into the intestinal barrier. Moreover, SIgA regulates gut microbiota composition and provides intestinal homeostasis. In this review, we present mechanisms of SIgA generation: T cell-dependent and -independent; in different non-organized and organized lymphoid structures in intestinal lamina propria (i.e., Peyer’s patches and isolated lymphoid follicles). We also summarize recent advances in understanding of SIgA functions in intestinal mucosal secretions with focus on its role in regulating gut microbiota composition and generation of tolerogenic responses toward its members.
Collapse
Affiliation(s)
- Bernadeta Pietrzak
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
- Correspondence: (B.P.); (M.S.)
| | - Katarzyna Tomela
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznań, Poland; (K.T.); (A.M.)
| | - Agnieszka Olejnik-Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 8 Rokietnicka Street, 60-806 Poznań, Poland; (K.T.); (A.M.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary Street, 61-866 Poznań, Poland
| | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, 48 Wojska Polskiego, 60-627 Poznań, Poland;
- Correspondence: (B.P.); (M.S.)
| |
Collapse
|
26
|
Age-related chemokine alterations affect IgA secretion and gut immunity in female mice. Biogerontology 2020; 21:609-618. [DOI: 10.1007/s10522-020-09877-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/30/2020] [Indexed: 01/09/2023]
|
27
|
Wang SZ, Yu YJ, Adeli K. Role of Gut Microbiota in Neuroendocrine Regulation of Carbohydrate and Lipid Metabolism via the Microbiota-Gut-Brain-Liver Axis. Microorganisms 2020; 8:microorganisms8040527. [PMID: 32272588 PMCID: PMC7232453 DOI: 10.3390/microorganisms8040527] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/10/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota play an important role in maintaining intestinal health and are involved in the metabolism of carbohydrates, lipids, and amino acids. Recent studies have shown that the central nervous system (CNS) and enteric nervous system (ENS) can interact with gut microbiota to regulate nutrient metabolism. The vagal nerve system communicates between the CNS and ENS to control gastrointestinal tract functions and feeding behavior. Vagal afferent neurons also express receptors for gut peptides that are secreted from enteroendocrine cells (EECs), such as cholecystokinin (CCK), ghrelin, leptin, peptide tyrosine tyrosine (PYY), glucagon-like peptide-1 (GLP-1), and 5-hydroxytryptamine (5-HT; serotonin). Gut microbiota can regulate levels of these gut peptides to influence the vagal afferent pathway and thus regulate intestinal metabolism via the microbiota-gut-brain axis. In addition, bile acids, short-chain fatty acids (SCFAs), trimethylamine-N-oxide (TMAO), and Immunoglobulin A (IgA) can also exert metabolic control through the microbiota-gut-liver axis. This review is mainly focused on the role of gut microbiota in neuroendocrine regulation of nutrient metabolism via the microbiota-gut-brain-liver axis.
Collapse
Affiliation(s)
- Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang 421001, China;
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
- Molecular Medicine, Research Institute, The Hospital for Sick Children and Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Yi-Jing Yu
- Molecular Medicine, Research Institute, The Hospital for Sick Children and Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children and Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
- Correspondence: ; Tel.: +1-416-813-8682; Fax: +1-416-813-6257
| |
Collapse
|
28
|
Prawiro SR, Poeranto S, Amalia A, Widyani EL, Indraswari G, Soraya M, Dwi Pradipto SR, Prasetya A, Hidayat GR, Alitha Putri SN. Generating mucosal and systemic immune response following vaccination of vibrio cholerae adhesion molecule against shigella flexneri infection. Indian J Med Microbiol 2020; 38:37-45. [PMID: 32719207 DOI: 10.4103/ijmm.ijmm_19_411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction Previous studies have shown 37.8 kDa pili subunit protein of Vibrio cholerae and 49.8 kDa pili subunit protein of Shigella flexneri can act as an adhesion molecule to initiate infection. These molecules also have the ability to agglutinate blood. The present study assessed mucosal and systemic immunity following vaccination using 37.8 kDa V. cholerae and protection against S. flexneri. Subjects and Methods Haemagglutination test was performed after purification of V. cholerae protein, followed by an anti-haemagglutination test. The intestinal weight and colony count were used to validate the protective effect on balb/c mice which were divided into the naive group, Shigella-positive control group, Vibrio-positive control group, V. cholerae infected-Vibrio-vaccinated group and S. flexneri-infected-Vibrio-vaccinated group. Th17, Treg, interleukin (IL) IL-17A, β-defensin and secretory-immunoglobulin A (s-IgA) were also measured to determine the systemic and mucosal immunity after vaccination. Results The haemagglutination and anti-haemagglutination tests showed that the 37.8 kDa protein could inhibit 49.8 kDa of the S. flexneri pili subunit. Decreased intestinal weight and colony count of vaccinated group compared to naive group also support cross reaction findings. Vaccination also generates higher level of Th17, Treg, IL-17A, β-defensin and s-IgA significantly. Conclusions 37.8 kDa subunit pili can act as a homologous vaccine candidate to prevent V. cholerae and S. flexneri infection.
Collapse
Affiliation(s)
- Sumarno Reto Prawiro
- Department of Clinical Microbiology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Sri Poeranto
- Department of Clinical Parasitology, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Aisyah Amalia
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Elsa Larissa Widyani
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Genitri Indraswari
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Merika Soraya
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Septha Rully Dwi Pradipto
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Adrian Prasetya
- Master Program in Biomedical Sciences, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | - Guntur Rizal Hidayat
- Bachelor Medical Program, Faculty of Medicine, University of Brawijaya, Malang, Indonesia
| | | |
Collapse
|
29
|
Arciniega-Martínez IM, Drago-Serrano ME, Salas-Pimentel M, Ventura-Juárez J, Reséndiz-Albor AA, Campos-Rodríguez R. Anterior subdiaphragmatic vagotomy decreases the IgA antibody response in the small intestines of BALB/c mice. J Neuroimmunol 2019; 337:577072. [DOI: 10.1016/j.jneuroim.2019.577072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/04/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
|
30
|
Xie SZ, Liu B, Ye HY, Li QM, Pan LH, Zha XQ, Liu J, Duan J, Luo JP. Dendrobium huoshanense polysaccharide regionally regulates intestinal mucosal barrier function and intestinal microbiota in mice. Carbohydr Polym 2019; 206:149-162. [DOI: 10.1016/j.carbpol.2018.11.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/15/2018] [Accepted: 11/01/2018] [Indexed: 02/07/2023]
|
31
|
Pérez-López JA, Rojas-Hernández S, Campos-Rodríguez R, Arciniega-Martínez IM, Cruz-Hernández TR, Reséndiz-Albor AA, Drago-Serrano ME. Posterior Subdiaphragmatic Vagotomy Downmodulates the IgA Levels in the Small Intestine of BALB/c Mice. Neuroimmunomodulation 2019; 26:292-300. [PMID: 31918430 DOI: 10.1159/000505097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/28/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The posterior vagus nerve trunk innervates the entire small intestine, and elucidating its modulatory role in the IgA response was the aim of this study. METHODS Two groups of six male BALB/c mice underwent sham or posterior subdiaphragmatic vagotomy and were euthanized on the 14th postoperative day; then, the small intestines were dissected. The intestinal fluid was harvested for antibody analysis by ELISA, and cell suspensions from Peyer's patches and lamina propria were prepared for cytofluorometric analysis of plasma cells and T lymphocytes. The CD4+ T cells were labeled for the intracellular IgA-producing interleukins (ILs)-4, -5, -6, and -10; transforming growth factor (TGF)-β; and the inflammatory cytokines tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and IL-12. In the intestinal tissue samples, myeloperoxidase (MPO) visualization and the enzymatic activity were assessed by immunohistochemistry and ELISA, respectively. The data were analyzed by Student's t test, and the differences were considered significant at p < 0.05. RESULTS In the vagotomy group, the IgA levels and the CD4+ T cells labeled with mediators that promote IgA secretion, including IL-4 (only at lamina propria), TNF-α, and IFN-γ, were decreased, whereas the lamina propria IgA+ plasma cells and MPO presence/activity were increased; changes in the IgM levels, IgM+ plasma cells, and CD4+ T cells labeled with TGF-β, which have a role in class switch recombination, were not observed. CONCLUSION The downmodulating impact of vagotomy on IgA levels may result from defective IgA secretion without affecting class switch recombination, whereas vagotomy evoked a proinflammatory response regarding MPO. These findings may reflect the role of the vagus nerve on the control of the IgA response in the small intestine.
Collapse
Affiliation(s)
- José Alfredo Pérez-López
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Saúl Rojas-Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rafael Campos-Rodríguez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Ivonne Maciel Arciniega-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Teresita Rocío Cruz-Hernández
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Aldo Arturo Reséndiz-Albor
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, Mexico City, Mexico,
| |
Collapse
|
32
|
Xu Y, Sun J, Cui Y, Yu S, He J, Liu P, Zhang Q. Age‐related changes in the morphology and the distribution of IgA and IgG in the pharyngeal tonsils of yaks (Bos grunniens). J Morphol 2018; 280:214-222. [DOI: 10.1002/jmor.20933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/23/2018] [Accepted: 11/26/2018] [Indexed: 02/04/2023]
Affiliation(s)
- Yuanfang Xu
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Juan Sun
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Junfeng He
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Penggang Liu
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| | - Qian Zhang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary MedicineCollege of Veterinary Medicine, Gansu Agricultural University Lanzhou Gansu China
| |
Collapse
|
33
|
Matsuo K, Nagakubo D, Yamamoto S, Shigeta A, Tomida S, Fujita M, Hirata T, Tsunoda I, Nakayama T, Yoshie O. CCL28-Deficient Mice Have Reduced IgA Antibody-Secreting Cells and an Altered Microbiota in the Colon. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:800-809. [PMID: 29237777 DOI: 10.4049/jimmunol.1700037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 11/14/2017] [Indexed: 02/05/2023]
Abstract
CCL28 induces the migration of IgA Ab-secreting cells (ASCs) via CCR10 and also displays a potent antimicrobial activity in vitro. To explore the role of CCL28 in vivo, we generated CCL28-deficient mice. The mice exhibited a significant reduction and abnormal distribution of IgA ASCs in the lamina propria of the colon. The concentrations of total and Ag-specific IgA in the fecal extracts of CCL28-deficient mice were also drastically reduced. The average amount of IgA secreted by a single IgA ASC derived from the colon was also substantially reduced in CCL28-deficient mice. Furthermore, CCL28 was found to significantly increase the average amount of IgA secreted by a single IgA ASC derived from the colon in vitro. In contrast, the generation of IgA ASCs in Peyer's and cecal patches was not significantly impaired in CCL28-deficient mice. We also found a relative increase in the Class Bacilli in the fecal extracts of CCL28-deficient mice and demonstrated a potent antimicrobial activity of CCL28 against Bacillus cereus and Enterococcus faecalis, both of which belong to Class Bacilli. Thus, CCL28 may also suppress the outgrowth of some bacterial species by its direct antimicrobial activity. Finally, CCL28-deficient mice exhibited a highly aggravated dextran sodium sulfate-induced colitis that was ameliorated by pretreatment with antibiotics. Collectively, CCL28 plays a pivotal role in the homing, distribution, and function of IgA ASCs in the colon and may also affect the intestinal microbiota through its direct antimicrobial activity.
Collapse
Affiliation(s)
- Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Daisuke Nagakubo
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Shinya Yamamoto
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Akiko Shigeta
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Shuta Tomida
- Department of Biobank, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama 700-8558, Japan
| | - Mitsugu Fujita
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Takako Hirata
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan;
| | - Osamu Yoshie
- Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka 589-8511, Japan; and
| |
Collapse
|
34
|
Immunization Strategies Against Clostridium difficile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1050:197-225. [PMID: 29383671 DOI: 10.1007/978-3-319-72799-8_12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
C. difficile infection (CDI) is an important healthcare- but also community-associated disease. CDI is considered a public health threat and an economic burden. A major problem is the high rate of recurrences. Besides classical antibiotic treatments, new therapeutic strategies are needed to prevent infection, to treat patients and prevent recurrences. If fecal transplantation has been recommended to treat recurrences, another key approach is to restore immunity against C. difficile and its virulence factors. Here, after a summary concerning the virulence factors, the host immune response against C. difficile and its role in the outcome of disease, we review the different approaches of passive immunotherapies and vaccines developed against CDI. Passive immunization strategies are designed in function of the target antigen, the antibody-based product and its administration route. Similarly, for active immunization strategies, vaccine antigens can target toxins or surface proteins and immunization can be performed by parenteral or mucosal routes. For passive immunization and vaccination as well, we first present immunization assays performed in animal models and second in humans and associated clinical trials. The different studies are presented according to the mode of administration either parenteral or mucosal and the target antigens, either toxins or colonization factors.
Collapse
|
35
|
Badia-Boungou F, Sane F, Alidjinou EK, Ternois M, Opoko PA, Haddad J, Stukens C, Lefevre C, Gueorguieva I, Hamze M, Ismail M, Weill J, Monabéka HG, Bouenizabila E, Moukassa D, Abena AA, Hober D. Marker of coxsackievirus-B4 infection in saliva of patients with type 1 diabetes. Diabetes Metab Res Rev 2017; 33. [PMID: 28719027 DOI: 10.1002/dmrr.2916] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/20/2017] [Accepted: 07/03/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Coxsackieviruses B (CV-B) are enteroviruses that have been reported to play a role in the pathogenesis of type 1 diabetes. Enteroviral RNA was detected in the gut mucosa of patients. The mucosal immunity is an interconnected network; therefore, the response to enteroviruses possibly present in the gastrointestinal mucosa can be reflected by specific antibodies in the saliva. In the present study, the anti-CV-B neutralizing activity of saliva samples from patients with type 1 diabetes was investigated. METHODS Saliva samples were collected from patients and controls of 3 countries, and plasma was obtained from some of them. The anti-CV-B activity of clinical samples was determined by neutralization of the cytopathic effect induced by challenging viruses in vitro and expressed as titre value. RESULTS Overall prevalence and levels of anti-CV-B4 activity of saliva were higher in patients (n = 181) than in controls (n = 135; P = .0002; titre values ≥ 16: odds ratio = 4.22 95% CI: 1.90-9.38 P = .0002). It has been shown that IgA1 played a role in this activity. There was no correlation between the saliva and the plasma anti-CV-B4 neutralizing activity. The neutralizing activity of saliva against CV-B1, CV-B2, CV-B3, and CV-B5 existed rarely, if at all. Increased levels of anti-CV-B4 activity were observed all along a 4 year follow-up period in patients but not in matched controls (P = .01). CONCLUSION There is an anti-CV-B4 activity in saliva of patients with type 1 diabetes that may be a useful marker to study the role of CV-B in the pathogenesis of the disease.
Collapse
Affiliation(s)
- F Badia-Boungou
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
| | - F Sane
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| | - E K Alidjinou
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| | - M Ternois
- Univ. Lille, Faculté d'odontologie et de chirurgie dentaire, Lille, France
| | - P A Opoko
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
| | - J Haddad
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - C Stukens
- CHU Hôpital Jeanne de Flandres, Lille, France
| | - C Lefevre
- CHU Hôpital Jeanne de Flandres, Lille, France
| | | | - M Hamze
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - M Ismail
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - J Weill
- CHU Hôpital Jeanne de Flandres, Lille, France
| | - H G Monabéka
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
- CHU de Brazzaville, Brazzaville, Republic of Congo
| | | | - D Moukassa
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
| | - A A Abena
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
| | - D Hober
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| |
Collapse
|
36
|
Abstract
免疫球蛋白A(immunoglobulin A, IgA)在黏膜的免疫功能中起关键作用, 是维持肠道黏膜稳态的重要物质. 分泌型IgA(secretory IgA, SIgA)的分泌组分保护免疫球蛋白不被蛋白水解酶降解, SIgA在肠道内各种免疫因子、免疫细胞以及其他免疫球蛋白的参与下完成肠道内的免疫监视、免疫自稳、免疫调控. 本文主要就肠道中SIgA的结构、合成转运、分泌调节、作用和其相关临床疾病的研究进展作一综述.
Collapse
|
37
|
Felgner J, Jain A, Nakajima R, Liang L, Jasinskas A, Gotuzzo E, Vinetz JM, Miyajima F, Pirmohamed M, Hassan-Hanga F, Umoru D, Jibir BW, Gambo S, Olateju K, Felgner PL, Obaro S, Davies DH. Development of ELISAs for diagnosis of acute typhoid fever in Nigerian children. PLoS Negl Trop Dis 2017. [PMID: 28640809 PMCID: PMC5498068 DOI: 10.1371/journal.pntd.0005679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Improved serodiagnostic tests for typhoid fever (TF) are needed for surveillance, to facilitate patient management, curb antibiotic resistance, and inform public health programs. To address this need, IgA, IgM and IgG ELISAs using Salmonella enterica serovar Typhi (S. Typhi) lipopolysaccharide (LPS) and hemolysin E (t1477) protein were conducted on 86 Nigerian pediatric TF and 29 non-typhoidal Salmonella (NTS) cases, 178 culture-negative febrile cases, 28 "other" (i.e., non-Salmonella) pediatric infections, and 48 healthy Nigerian children. The best discrimination was achieved between TF and healthy children. LPS-specific IgA and IgM provided receiver operator characteristic areas under the curve (ROC AUC) values of 0.963 and 0.968, respectively, and 0.978 for IgA+M combined. Similar performance was achieved with t1477-specific IgA and IgM (0.968 and 0.968, respectively; 0.976 combined). IgG against LPS and t1477 was less accurate for discriminating these groups, possibly as a consequence of previous exposure, although ROC AUC values were still high (0.928 and 0.932, respectively). Importantly, discrimination between TF and children with other infections was maintained by LPS-specific IgA and IgM (AUC = 0.903 and 0.934, respectively; 0.938 combined), and slightly reduced for IgG (0.909), while t1477-specific IgG performed best (0.914). A similar pattern was seen when comparing TF with other infections from outside Nigeria. The t1477 may be recognized by cross-reactive antibodies from other acute infections, although a robust IgG response may provide some diagnostic utility in populations where incidence of other infections is low, such as in children. The data are consistent with IgA and IgM against S. Typhi LPS being specific markers of acute TF.
Collapse
Affiliation(s)
- Jiin Felgner
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Aarti Jain
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Rie Nakajima
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Li Liang
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Algis Jasinskas
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Eduardo Gotuzzo
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
- Hospital Nacional Cayetano Heredia, Lima, Peru
| | - Joseph M. Vinetz
- Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Fabio Miyajima
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Munir Pirmohamed
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | - Safiya Gambo
- Department of Pediatrics, Murtala Specialist Hospital, Kano, Nigeria
| | | | - Philip L. Felgner
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
| | - Stephen Obaro
- Department of Pediatrics, Aminu Kano Teaching Hospital, Kano, Nigeria
- University of Abuja Teaching Hospital, Gwagwalada, Nigeria
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- International Foundation against Infectious Diseases in Nigeria (IFAIN), Abuja, Nigeria
| | - D. Huw Davies
- Division of Infectious Diseases, School of Medicine, University of California Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Salas-Cuestas F, Bautista-Molano W, Bello-Gualtero JM, Arias I, Castillo DM, Chila-Moreno L, Valle-Oñate R, Herrera D, Romero-Sánchez C. Higher Levels of Secretory IgA Are Associated with Low Disease Activity Index in Patients with Reactive Arthritis and Undifferentiated Spondyloarthritis. Front Immunol 2017; 8:476. [PMID: 28496443 PMCID: PMC5406393 DOI: 10.3389/fimmu.2017.00476] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 04/05/2017] [Indexed: 01/31/2023] Open
Abstract
Introduction Both reactive arthritis (ReA) and undifferentiated spondyloarthritis (uSpA) belong to the group of autoinflammatory diseases called spondyloarthritis (SpA). Hypotheses have been proposed about a relationship between the intestinal mucosa and inflammation of joint tissues. The role of immunoglobulin IgA or secretory immunoglobulin A (SIgA) in the inflammatory and/or clinical activity of patients with SpA remains poorly understood. Objective To evaluate the status of total IgA and SIgA, and the association among the levels of SIgA, IgA, IgA anti-Chlamydia trachomatis, and anti-Shigella spp. with the disease activity measures, erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels, was compared in a cohort of patients with ReA and uSpA and healthy subjects. Methods This was a cross-sectional study. The serum concentrations of SIgA, IgA anti-C. trachomatis, anti-Shigella spp., and total IgA were measured. Disease activity was measured in each patient by means of Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) and Ankylosing Spondylitis Disease Activity Score (ASDAS). Statistical analysis did include as bivariate evaluation, comparisons by Student’s t-test, Kruskal–Wallis test, and U Mann–Whitney test, with a multivariate evaluation by principal components analysis (PCA). A correlation analysis was carried out using the Pearson correlation coefficient and a linear regression models. All analysis were made using Stata version 11.2® for Windows, R V3.3.21. Statistical significance was defined a p-value <0.05. Results In all, 46 patients (78.2% men; mean age, 34.8 ± 12.3 years) and 53 controls (41% men; mean age, 32 ± 11.4 years) were included in the study. The mean serum levels of SIgA were higher in SpA patients than in healthy subjects (p < 0.001). Only SIgA levels correlated with disease activity: BASDAI (r = −0.42, p = 0.0046), ASDAS-CRP (r = −0.37, p = 0.014), and ASDAS-ESR (r = −0.45, p = 0.0021). The negative correlation between SIgA and all activity indices was higher in HLA-B27-positive patients (BASDAI r = −0.70, p = 0.0009, ASDAS-CRP r = −0.58, p = 0.0093, and ASDAS-ESR r = −0.57, p = 0.0083). The PCA showed three factors: the first component was constituted by variables referred as clinical activity measures, the second did include the serological activity markers, and the last component was compounded by age and symptoms time. Conclusion Elevated serum levels of SIgA were found to be related with low disease activity in patients with ReA and uSpA.
Collapse
Affiliation(s)
| | - Wilson Bautista-Molano
- Faculty of Medicine, Universidad Militar Nueva Granada, Bogotá, Colombia.,Unit of Oral Basic Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Juan M Bello-Gualtero
- Faculty of Medicine, Universidad Militar Nueva Granada, Bogotá, Colombia.,Department of Rheumatology and Immunology, Hospital Militar Central, Bogotá, Colombia
| | - Ivonne Arias
- School of Medicine, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Diana Marcela Castillo
- Unit of Oral Basic Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Lorena Chila-Moreno
- Unit of Oral Basic Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Rafael Valle-Oñate
- Faculty of Medicine, Universidad Militar Nueva Granada, Bogotá, Colombia.,Department of Rheumatology and Immunology, Hospital Militar Central, Bogotá, Colombia
| | - Daniel Herrera
- School of Medicine, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Consuelo Romero-Sánchez
- Faculty of Medicine, Universidad Militar Nueva Granada, Bogotá, Colombia.,Unit of Oral Basic Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia.,Department of Rheumatology and Immunology, Hospital Militar Central, Bogotá, Colombia
| |
Collapse
|
39
|
Liu S, Pei F, Wang X, Li D, Zhao L, Song Y, Chen Z, Liu B. The immune impact of mimic endoscopic retrograde appendicitis therapy and appendectomy on rabbits of acute appendicitis. Oncotarget 2017; 8:66528-66539. [PMID: 29029533 PMCID: PMC5630433 DOI: 10.18632/oncotarget.16236] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/28/2017] [Indexed: 01/28/2023] Open
Abstract
This study was conducted to evaluate the immune impact of mimic endoscopic retrograde appendicitis therapy and appendectomy on rabbits of acute suppurative appendicitis and to determine whether TLR4/MYD88/NF-κB signaling pathway was activated in this process. 48 rabbits were assigned into 4 groups: group I, the mimic endoscopic retrograde appendicitis therapy group; group II, the appendectomy group; group III, the model group; and group IV, the blank group. White blood cells decreased, while levels of C-reactive protein, tumor necrosis factor-α, interleukin-6, interleukin-4, and interleukin-10 increased on the 2nd day in group I and II. IgA in feces decreased at 2 weeks, while fecal microbiota changed at 2 and 4 weeks after appendectomy. CD8+ cells in appendix of group I increased within 8 weeks. Upregulated expression of TLR4, MYD88, and nuclear NF-κB were detected on the 2nd day in group I and II. Mimic endoscopic retrograde appendicitis therapy and appendectomy are effective ways for acute suppurative appendicitis. Mimic endoscopic retrograde appendicitis therapy was more preferable due to its advantage in maintaining intestinal immune function. TLR4/MYD88/NF-κB signaling pathway was activated in acute phase of appendicitis.
Collapse
Affiliation(s)
- Suqin Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Fenghua Pei
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhong Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Deliang Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lixia Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanyan Song
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhendong Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bingrong Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
40
|
Immunogenic Properties of Lactobacillus plantarum Producing Surface-Displayed Mycobacterium tuberculosis Antigens. Appl Environ Microbiol 2016; 83:AEM.02782-16. [PMID: 27815271 DOI: 10.1128/aem.02782-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/27/2016] [Indexed: 12/18/2022] Open
Abstract
Tuberculosis (TB) remains among the most deadly diseases in the world. The only available vaccine against tuberculosis is the bacille Calmette-Guérin (BCG) vaccine, which does not ensure full protection in adults. There is a global urgency for the development of an effective vaccine for preventing disease transmission, and it requires novel approaches. We are exploring the use of lactic acid bacteria (LAB) as a vector for antigen delivery to mucosal sites. Here, we demonstrate the successful expression and surface display of a Mycobacterium tuberculosis fusion antigen (comprising Ag85B and ESAT-6, referred to as AgE6) on Lactobacillus plantarum The AgE6 fusion antigen was targeted to the bacterial surface using two different anchors, a lipoprotein anchor directing the protein to the cell membrane and a covalent cell wall anchor. AgE6-producing L. plantarum strains using each of the two anchors induced antigen-specific proliferative responses in lymphocytes purified from TB-positive donors. Similarly, both strains induced immune responses in mice after nasal or oral immunization. The impact of the anchoring strategies was reflected in dissimilarities in the immune responses generated by the two L. plantarum strains in vivo The present study comprises an initial step toward the development of L. plantarum as a vector for M. tuberculosis antigen delivery. IMPORTANCE This work presents the development of Lactobacillus plantarum as a candidate mucosal vaccine against tuberculosis. Tuberculosis remains one of the top infectious diseases worldwide, and the only available vaccine, bacille Calmette-Guérin (BCG), fails to protect adults and adolescents. Direct antigen delivery to mucosal sites is a promising strategy in tuberculosis vaccine development, and lactic acid bacteria potentially provide easy, safe, and low-cost delivery vehicles for mucosal immunization. We have engineered L. plantarum strains to produce a Mycobacterium tuberculosis fusion antigen and to anchor this antigen to the bacterial cell wall or to the cell membrane. The recombinant strains elicited proliferative antigen-specific T-cell responses in white blood cells from tuberculosis-positive humans and induced specific immune responses after nasal and oral administrations in mice.
Collapse
|