1
|
Millman JF, Kondrashina A, Walsh C, Busca K, Karawugodage A, Park J, Sirisena S, Martin FP, Felice VD, Lane JA. Biotics as novel therapeutics in targeting signs of skin ageing via the gut-skin axis. Ageing Res Rev 2024:102518. [PMID: 39389239 DOI: 10.1016/j.arr.2024.102518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024]
Abstract
Skin ageing is a phenomenon resulting from the aggregative changes to skin structure and function and is clinically manifested by physical features such as wrinkles, hyperpigmentation, elastosis, telangiectasia, and deterioration of skin barrier integrity. One of the main drivers of skin ageing, UV radiation, negatively influences the homeostasis of cells and tissues comprising the skin by triggering production of immune-mediated reactive oxygen species (ROS) and pro-inflammatory cytokines, as well as a various hormones and neuropeptides. Interestingly, an established link between the gut and the skin coined the 'gut-skin axis' has been demonstrated, with dysbiosis and gut barrier dysfunction frequently observed in certain inflammatory skin conditions and more recently, implicated in skin ageing. Therapeutic use of 'biotics' including prebiotics, probiotics, postbiotics, and synbiotics, which modulate the gut microbiota and production of microbially associated metabolites, influence the activity of the gut mucosal and immune systems and are showing promise as key candidates in addressing signs of skin ageing. In this review we aim to focus on the structure and function of the gut-skin axis and showcase the recent in-vitro and clinical evidence demonstrating the beneficial effects of select biotics in targeting signs of skin ageing and discuss the proposed mechanisms mediated via the gut-skin axis underpinning these effects.
Collapse
Affiliation(s)
- Jasmine F Millman
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood VIC, Australia, 3066.
| | - Alina Kondrashina
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork, Ireland, P61K202
| | - Clodagh Walsh
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork, Ireland, P61K202
| | - Kizkitza Busca
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork, Ireland, P61K202
| | - Aneesha Karawugodage
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood VIC, Australia, 3066
| | - Julia Park
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood VIC, Australia, 3066
| | - Sameera Sirisena
- Health and Happiness (H&H) Group, H&H Research, Level 6 & 7, 88 Langridge St, Collingwood VIC, Australia, 3066
| | - Francois-Pierre Martin
- Health and Happiness (H&H) Group, H&H Research, Avenue Sécheron 15, Bat F2/F3 1202, Geneva, Switzerland
| | - Valeria D Felice
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork, Ireland, P61K202
| | - Jonathan A Lane
- Health and Happiness (H&H) Group, H&H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co. Cork, Ireland, P61K202.
| |
Collapse
|
2
|
Chen Y, Chen H, Han C, Ou H, Zhan X. The structure and proteomic analysis of byssus in Pteria penguin: Insights into byssus evolution and formation. J Proteomics 2024; 307:105267. [PMID: 39089615 DOI: 10.1016/j.jprot.2024.105267] [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: 04/01/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Byssus is a unique external structure in sessile bivalves and is critical for settlement and metamorphosis. However, little is known about the stout byssus in Pteria penguin. We explored the byssus structure and proteins using scanning electron microscopy and proteomics, respectively. The results revealed that P. penguin byssus has a dense and highly aligned fiber inner core, and the outer cuticle contains protein granules embedded in the protein matrix. Proteomic analysis revealed 31 proteins in the byssus, among which 15 differentially expressed proteins were mainly enriched in the EGF/EGF-like and laminin EGF-like domains. Foot proteins were enriched in the EF-hand, immunoglobulin, and fibronectin domains. All these domains can participate in protein-protein and/or protein-metal interactions in the extracellular matrix (ECM), which, together with the seven types of ECM proteins detected in the byssus, supports the hypothesis that the byssus is derived from the ECM. We also found that in vitro acellular structures of the byssus and the shell shared commonalities in their formation processes. These results are useful for further understanding byssus evolution and the characterization of byssus-related proteins. SIGNIFICANCE: This manuscript investigates the structure and the origin of Pteria penguin byssus, given that byssus is vital to provide critical protection for reproduction and even against environmental stresses that affect survival. However, there is rare research on byssus protein composition. Hence, though scanning electron microscopy and proteomic analysis, we discovered that P. penguin byssus possesses the dense and highly aligned fiber inner core, and the outer cuticle has protein granules embedded in the protein matrix. Proteomic analysis showed that there were 31 proteins in the byssus, among which 15 proteins were mainly enriched in the EGF/EGF-like and laminin EGF-like domains. Foot proteins closely related to byssus formation were enriched in EF hand, immunoglobulin, and fibronectin domains. These domains are able to participate in protein-protein and/or protein-metal interactions in the extracellular matrix (ECM), which together with the seven types of ECM proteins detected in byssus support the hypothesis that byssus derive from the ECM. We also found in vitro acellular structures the byssus and the shell share commonalities in their formation processes. These results were useful for further understanding the byssus evolution and the characterization of the byssus-related proteins.
Collapse
Affiliation(s)
- Yi Chen
- School of Ecology, Hainan University, Haikou 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan University, Haikou 570228, China
| | - Hengda Chen
- School of Marine Biology and Fisheries, Hainan University, Haikou 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan University, Haikou 570228, China
| | - Changqing Han
- School of Marine Biology and Fisheries, Hainan University, Haikou 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan University, Haikou 570228, China
| | - Huilong Ou
- School of Marine Biology and Fisheries, Hainan University, Haikou 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan University, Haikou 570228, China
| | - Xin Zhan
- School of Marine Biology and Fisheries, Hainan University, Haikou 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China; Key Laboratory of Tropical Hydrobiology and Biotechnology of Hainan Province, Hainan University, Haikou 570228, China.
| |
Collapse
|
3
|
Demirturk M, Cinar MS, Avci FY. The immune interactions of gut glycans and microbiota in health and disease. Mol Microbiol 2024; 122:313-330. [PMID: 38703041 DOI: 10.1111/mmi.15267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
The human digestive system harbors a vast diversity of commensal bacteria and maintains a symbiotic relationship with them. However, imbalances in the gut microbiota accompany various diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancers (CRCs), which significantly impact the well-being of populations globally. Glycosylation of the mucus layer is a crucial factor that plays a critical role in maintaining the homeostatic environment in the gut. This review delves into how the gut microbiota, immune cells, and gut mucus layer work together to establish a balanced gut environment. Specifically, the role of glycosylation in regulating immune cell responses and mucus metabolism in this process is examined.
Collapse
Affiliation(s)
- Mahmut Demirturk
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mukaddes Sena Cinar
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fikri Y Avci
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
4
|
Einhorn V, Haase H, Maares M. Interaction and competition for intestinal absorption by zinc, iron, copper, and manganese at the intestinal mucus layer. J Trace Elem Med Biol 2024; 84:127459. [PMID: 38640745 DOI: 10.1016/j.jtemb.2024.127459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Trace elements such as zinc, manganese, copper, or iron are essential for a wide range of physiological functions. It is therefore crucial to ensure an adequate supply of these elements to the body. Many previous investigations have dealt with the role of transport proteins, in particular their selectivity for, and competition between, different ions. Another so far less well investigated major factor influencing the absorption of trace elements seems to be the intestinal mucus layer. This gel-like substance covers the entire gastrointestinal tract and its physiochemical properties can be mainly assigned to the glycoproteins it contains, so-called mucins. Interaction with mucins has already been demonstrated for some metals. However, knowledge about the impact on the respective bioavailability and competition between those metals is still sketchy. This review therefore aims to summarize the findings and knowledge gaps about potential effects regarding the interaction between gastrointestinal mucins and the trace elements iron, zinc, manganese, and copper. Mucins play an indispensable role in the absorption of these trace elements in the neutral to slightly alkaline environment of the intestine, by keeping them in a soluble form that can be absorbed by enterocytes. Furthermore, the studies so far indicate that the competition between these trace elements for uptake already starts at the intestinal mucus layer, yet further research is required to completely understand this interaction.
Collapse
Affiliation(s)
- Vincent Einhorn
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Hajo Haase
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany
| | - Maria Maares
- Technische Universität Berlin, Department of Food Chemistry and Toxicology, Straße des 17. Juni 135, Berlin 10623, Germany; Trace Age-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly, Potsdam-Jena-Wuppertal, Berlin, Germany; Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, Nuthetal 14558, Germany.
| |
Collapse
|
5
|
Fan Y, Wang Y, Xiao H, Sun H. Advancements in understanding the role of intestinal dysbacteriosis mediated mucosal immunity in IgA nephropathy. BMC Nephrol 2024; 25:203. [PMID: 38907188 PMCID: PMC11191200 DOI: 10.1186/s12882-024-03646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
IgA nephropathy, presently recognized as the foremost primary glomerular disorder, emerges as a principal contributor to renal failure globally, with its pathogenesis yet to be fully elucidated. Extensive research has highlighted the critical role of gut microbiome in the onset and progression of IgA nephropathy, underscoring its importance in accurately delineating the disease's etiology. For example, gut microbiome dysbacteriosis can lead to the production of nephritogenic IgA1 antibodies, which form immune complexes that deposit in the kidneys, causing inflammation and damage. The gut microbiome, a source of numerous bioactive compounds, interacts with the host and plays a regulatory role in gut-immune axis modulation, earning it the moniker of the "second brain." Recent investigations have particularly emphasized a significant correlation between IgA nephropathy and gut microbiome dysbacteriosis. This article offers a detailed overview of the pathogenic mechanisms of IgA nephropathy, specifically focusing on elucidating how alterations in the gut microbiome are associated with anomalies in the intestinal mucosal system in IgA nephropathy. Additionally, it describes the possible influence of gut microbiome on recurrent IgA nephropathy following kidney transplantation. Furthermore, it compiles potential therapeutic interventions, offering both theoretical and practical foundations for the management of IgA nephropathy. Lastly, the challenges currently faced in the therapeutic approaches to IgA nephropathy are discussed.
Collapse
Affiliation(s)
- Yitao Fan
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yan Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Han Xiao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Hui Sun
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730030, Gansu, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
6
|
Almhanna H, AL-Mahmodi AMM, Kadhim AB, Kumar AAHS. Network and structural analysis of quail mucins with expression pattern of mucin 1 and mucin 4 in the intestines of the Iraqi common quail ( Coturnix coturnix). Vet World 2024; 17:1227-1237. [PMID: 39077459 PMCID: PMC11283604 DOI: 10.14202/vetworld.2024.1227-1237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 07/31/2024] Open
Abstract
Background and Aim In avian and other species, mucins (MUCs) play a crucial role in the gastrointestinal tract (GIT), and constitute a large group of O-glycosylated glycoproteins, are glycoconjugate proteins. MUCs present in two forms: (1) membrane-attached on cell surfaces to repel external threats and (2) detachable, gel-forming proteins in the soluble form. In quail GIT, the specific types of MUCs that are expressed remain largely unknown. We investigated the expression of MUC1 and MUC4 MUCs in the GIT of Iraqi common quails and conducted network and structural analyses of all known MUC types across quail breeds. Materials and Methods Histological and gene expression analyses of MUC1 and MUC4 were conducted using fresh small intestine and large intestine samples from 10 quails. Using the STRING Database, Chimera software, and PrankWeb-ligand binding site prediction tool, network and structural analyses of all reported types of quail MUCs were conducted. Results Most intestinal MUCs in quails were acidic, with few neutral MUCs detectable through Alcian blue and periodic acid-schiff stains. Acidic MUCs were more expressed in the duodenum, ileum, cecum, and colon, whereas neutral MUCs were more expressed in the jejunum. MUC1 and MUC4 messenger RNA expression was significantly higher in the jejunum and colon than in the duodenum and ileum. The analysis of the network revealed that MUC 1, 15, 16, and 24 formed homologous networks, while MUC 2, 4, 5, and 6 formed heterologous networks. Specific MUC combinations, including MUC5A-MUC6, MUC5A-MUC5B, and MUC5B-MUC6, show higher intermolecular hydrogen bond formation affinity. MUC15, MUC16, and MUC24 showed minimal interaction with other MUC types. Among the analyzed MUCs, MUC5B, and MUC6 had the highest probability for binding, while MUC2, MUC4, and MUC5A showed lower probabilities despite greater numbers of binding sites. Conclusion This study's results offer significant insights into quails' MUCs' composition, expression, network interactions, and binding sites, advancing knowledge of MUC-related processes in gastrointestinal physiology and their potential connection to gastrointestinal diseases.
Collapse
Affiliation(s)
- Hazem Almhanna
- Department of Anatomy and Histology, College of Veterinary Medicine, University of Al-Qadisiyah, Al Diwaniyah, Iraq
| | | | - Abdulrazzaq B Kadhim
- Department of Anatomy and Histology, College of Veterinary Medicine, University of Al-Qadisiyah, Al Diwaniyah, Iraq
| | - aArun H. S. Kumar
- Department of Veterinary Biosciences, Stemcology, School of Veterinary Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
7
|
Wei X, Tsai MS, Liang L, Jiang L, Hung CJ, Jelliffe-Pawlowski L, Rand L, Snyder M, Jiang C. Vaginal microbiomes show ethnic evolutionary dynamics and positive selection of Lactobacillus adhesins driven by a long-term niche-specific process. Cell Rep 2024; 43:114078. [PMID: 38598334 DOI: 10.1016/j.celrep.2024.114078] [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: 09/15/2023] [Revised: 03/01/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
The vaginal microbiome's composition varies among ethnicities. However, the evolutionary landscape of the vaginal microbiome in the multi-ethnic context remains understudied. We perform a systematic evolutionary analysis of 351 vaginal microbiome samples from 35 multi-ethnic pregnant women, in addition to two validation cohorts, totaling 462 samples from 90 women. Microbiome alpha diversity and community state dynamics show strong ethnic signatures. Lactobacillaceae have a higher ratio of non-synonymous to synonymous polymorphism and lower nucleotide diversity than non-Lactobacillaceae in all ethnicities, with a large repertoire of positively selected genes, including the mucin-binding and cell wall anchor genes. These evolutionary dynamics are driven by the long-term evolutionary process unique to the human vaginal niche. Finally, we propose an evolutionary model reflecting the environmental niches of microbes. Our study reveals the extensive ethnic signatures in vaginal microbial ecology and evolution, highlighting the importance of studying the host-microbiome ecosystem from an evolutionary perspective.
Collapse
Affiliation(s)
- Xin Wei
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Ming-Shian Tsai
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liang Liang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Liuyiqi Jiang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China
| | - Chia-Jui Hung
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Informatics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Laura Jelliffe-Pawlowski
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Larry Rand
- Department of Obstetrics, Gynecology & Reproductive Sciences, School of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Chao Jiang
- MOE Key Laboratory of Biosystems Homeostasis & Protection, and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310030, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
8
|
Wang Z, Shen J. The role of goblet cells in Crohn' s disease. Cell Biosci 2024; 14:43. [PMID: 38561835 PMCID: PMC10985922 DOI: 10.1186/s13578-024-01220-w] [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: 06/01/2023] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of Crohn's disease (CD), a subtype of inflammatory bowel disease (IBD), is increasing worldwide. The pathogenesis of CD is hypothesized to be related to environmental, genetic, immunological, and bacterial factors. Current studies have indicated that intestinal epithelial cells, including columnar, Paneth, M, tuft, and goblet cells dysfunctions, are strongly associated with these pathogenic factors. In particular, goblet cells dysfunctions have been shown to be related to CD pathogenesis by direct or indirect ways, according to the emerging studies. The mucus barrier was established with the help of mucins secreted by goblet cells. Not only do the mucins mediate the mucus barrier permeability and bacterium selection, but also, they are closely linked with the endothelial reticulum stress during the synthesis process. Goblet cells also play a vital role in immune response. It was indicated that goblet cells take part in the antigen presentation and cytokines secretion process. Disrupted goblet cells related immune process were widely discovered in CD patients. Meanwhile, dysbiosis of commensal and pathogenic microbiota can induce myriad immune responses through mucus and goblet cell-associated antigen passage. Microbiome dysbiosis lead to inflammatory reaction against pathogenic bacteria and abnormal tolerogenic response. All these three pathways, including the loss of mucus barrier function, abnormal immune reaction, and microbiome dysbiosis, may have independent or cooperative effect on the CD pathogenesis. However, many of the specific mechanisms underlying these pathways remain unclear. Based on the current understandings of goblet cell's role in CD pathogenesis, substances including butyrate, PPARγagonist, Farnesoid X receptor agonist, nuclear factor-Kappa B, nitrate, cytokines mediators, dietary and nutrient therapies were all found to have potential therapeutic effects on CD by regulating the goblet cells mediated pathways. Several monoclonal antibodies already in use for the treatment of CD in the clinical settings were also found to have some goblet cells related therapeutic targets. In this review, we introduce the disease-related functions of goblet cells, their relationship with CD, their possible mechanisms, and current CD treatments targeting goblet cells.
Collapse
Affiliation(s)
- Zichen Wang
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China.
| |
Collapse
|
9
|
Koh YC, Chang YC, Lin WS, Leung SY, Chen WJ, Wu SH, Wei YS, Gung CL, Chou YC, Pan MH. Efficacy and Mechanism of the Action of Live and Heat-Killed Bacillus coagulans BC198 as Potential Probiotic in Ameliorating Dextran Sulfate Sodium-Induced Colitis in Mice. ACS OMEGA 2024; 9:10253-10266. [PMID: 38463297 PMCID: PMC10918820 DOI: 10.1021/acsomega.3c07529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/01/2024] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
Inflammatory bowel disease alters the gut microbiota, causes defects in mucosal barrier function, and leads to dysregulation of the immune response to microbial stimulation. This study investigated and compared the efficacy of a candidate probiotic strain, Bacillus coagulans BC198, and its heat-killed form in treating dextran sulfate sodium-induced colitis. Both live and heat-killed B. coagulans BC198 increased gut barrier-associated protein expression, reduced neutrophil and M1 macrophage infiltration of colon tissue, and corrected gut microbial dysbiosis induced by colitis. However, only live B. coagulans BC198 could alleviate the general symptoms of colitis, prevent colon shortening, and suppress inflammation and tissue damage. At the molecular level, live B. coagulans BC198 was able to inhibit Th17 cells while promoting Treg cells in mice with colitis, reduce pro-inflammatory MCP-1 production, and increase anti-inflammatory IL-10 expression in the colonic mucosa. The live form of B. coagulans BC198 functioned more effectively than the heat-killed form in ameliorating colitis by enhancing the anti-inflammatory response and promoting Treg cell accumulation in the colon.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Ya-Chu Chang
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Sheng Lin
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Food Science, National Quemoy University, Quemoy 892, Taiwan
| | - Siu-Yi Leung
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wei-Jen Chen
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Shiuan-Huei Wu
- Biotech
Department, Syngen Biotech Co., Ltd., Tainan 744094, Taiwan
| | - Yu-Shan Wei
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Chiau-Ling Gung
- Research
and Development Department, Syngen Biotech
Co., Ltd., Tainan 744094, Taiwan
| | - Ya-Chun Chou
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung City 40402, Taiwan
- Department
of Health and Nutrition Biotechnology, Asia
University, Taichung City 41354, Taiwan
| |
Collapse
|
10
|
Niu Y, Zhang R, Yang C, He J, Wang T. Dietary supplementation with dihydroartemisinin improves intestinal barrier function in weaned piglets with intrauterine growth retardation by modulating the gut microbiota. J Anim Sci 2024; 102:skae140. [PMID: 38813622 PMCID: PMC11222986 DOI: 10.1093/jas/skae140] [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: 11/20/2023] [Accepted: 05/29/2024] [Indexed: 05/31/2024] Open
Abstract
The aim of this study was to investigate whether dietary dihydroartemisinin (DHA) supplementation could improve intestinal barrier function and microbiota composition in intrauterine growth restriction (IUGR) weaned piglets. Twelve normal birth weight (NBW) piglets and 24 IUGR piglets at 21 d of age were divided into three groups, which were fed a basal diet (NBW-CON and IUCR-CON groups) and an 80 mg/kg DHA diet (IUGR-DHA group). At 49 d of age, eight piglets of each group with similar body weights within groups were slaughtered, and serum and small intestine samples were collected. The results showed that IUGR piglets reduced growth performance, impaired the markers of intestinal permeability, induced intestinal inflammation, decreased intestinal immunity, and disturbed the intestinal microflora. Dietary DHA supplementation increased average daily gain, average daily feed intake, and body weight at 49 d of age in IUGR-weaned piglets (P < 0.05). DHA treatment decreased serum diamine oxidase activity and increased the numbers of intestinal goblet cells and intraepithelial lymphocytes, concentrations of jejunal mucin-2 and ileal trefoil factor 3, and intestinal secretory immunoglobin A and immunoglobin G (IgG) concentrations of IUGR piglets (P < 0.05). Diet supplemented with DHA also upregulated mRNA abundances of jejunal IgG, the cluster of differentiation 8 (CD8), major histocompatibility complex-I (MHC-I), and interleukin 6 (IL-6) and ileal IgG, Fc receptor for IgG (FcRn), cluster of differentiation 8 (CD4), CD8, MHC-I, IL-6 and tumor necrosis factor α (TNF-α), and enhanced mRNA abundance and protein expression of intestinal occludin and ileal claudin-1 in IUGR piglets (P < 0.05). In addition, DHA supplementation in the diet improved the microbial diversity of the small intestine of IUGR piglets and significantly increased the relative abundance of Actinobacteriota, Streptococcus, Blautia and Streptococcus in the jejunum, and Clostridium sensu_ stricto_in the ileum (P < 0.05). The intestinal microbiota was correlated with the mRNA abundance of tight junction proteins and inflammatory response-related genes. These data suggested that DHA could improve the markers of intestinal barrier function in IUGR-weaned piglets by modulating gut microbiota. DHA may be a novel nutritional candidate for preventing intestinal dysfunction in IUGR pigs.
Collapse
Affiliation(s)
- Yu Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A and F University, Hangzhou, 311300, China
| | - Ruiqiang Zhang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A and F University, Hangzhou, 311300, China
| | - Caimei Yang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A and F University, Hangzhou, 311300, China
| | - Jintian He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
11
|
Wagner A, Kapounková K, Struhár I. The relationship between the gut microbiome and resistance training: a rapid review. BMC Sports Sci Med Rehabil 2024; 16:4. [PMID: 38166998 PMCID: PMC10763211 DOI: 10.1186/s13102-023-00791-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
The human gut microbiome is attracting increasing attention because of its overall effect on health. Several reviews have investigated the impact of physical activity on the gut microbiome; however, these predominantly concentrate on either endurance or a combination of physical activities. This study aims to describe the effect of resistance or strength training on the gut microbiome of a human population. This rapid review follows the guidelines of the Cochrane Rapid Reviews Guidance along with PRISMA. A review of the literature was carried out using articles indexed by PubMed, Scopus, and Web of Science published in the last 12 years. None of the seven studies included find significant change in the gut microbiome in terms of bacterial taxa composition or overall diversity, though the results show that resistance training might decrease the zonulin level and increase mucin production and thereby reduce inflammation in the gut. Interestingly, two studies point to a gut-muscle axis connection and this is discussed in our paper. However, due to the small number of existing studies and certain methodological disagreements, it was hard to find a consensus on the relationship between the gut microbiome and resistance training.
Collapse
Affiliation(s)
- Adam Wagner
- Department of Sport Performance and Exercise Testing Promotion, Faculty of Sport Studies, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| | - Kateřina Kapounková
- Department of Physical Activities and Health Sciences, Faculty of Sport Studies, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Ivan Struhár
- Department of Physical Activities and Health Sciences, Faculty of Sport Studies, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| |
Collapse
|
12
|
Miller A, Cutroneo G, Lombardo GP, D'Angelo R, Pallio S, Migliorato A, Fumia A, Favaloro A, Lauriano ER, Pergolizzi S. Association between neuropeptides and mucins in Crohn's disease mucous cells. Acta Histochem 2023; 125:152115. [PMID: 37979446 DOI: 10.1016/j.acthis.2023.152115] [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: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are both inflammatory bowel diseases (IBD). Unlike UC, which is limited to the mucosa of the colon, CD inflammation is characterized by chronic mucosal ulcerations affecting the entire gastrointestinal tract. Goblet cells (GCs) can be found in some lining epithelia, particularly in the respiratory and digestive tracts. GCs represent the main source of mucin that are the significant components of the mucus layer; hypertrophy of GCs and an increase in mucin production are observed in many enteric infections. The cytoplasm of goblet cells may also contain neuropeptides, such as serotonin, that can be altered in inflammatory bowel disease (IBD). The defense system of the gut is represented by the intestinal mucosal barrier, its protective function is strictly connected to the regulation of the mucus layer and the coordination of the neuro-immune response. Paraformaldehyde-fixed intestinal tissues, obtained from fifteen patients with Crohn's disease, were analyzed by immunostaining for MUC2, MUC4, 5-HT, and VAChT. This study aims to define the link between neuropeptides and mucins in mucous cells and their involvement in the inflammation process. Our results showed in mucous cells of Crohn's disease (CD) patients a high expression of MUC4 and a decrease in the expression of vesicular acetylcholine transporter (VAChT) demonstrating the presence of an inflammatory state.
Collapse
Affiliation(s)
- Anthea Miller
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell'Annunziata, 98168 Messina, Italy
| | - Giuseppina Cutroneo
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98125 Messina, Italy
| | - Giorgia Pia Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Roberta D'Angelo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Socrate Pallio
- Department of Clinical and Experimental Medicine, University of Messina, 98147 Messina, Italy
| | - Alba Migliorato
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98125 Messina, Italy
| | - Angelo Fumia
- Department of Clinical and Experimental Medicine, University of Messina, 98147 Messina, Italy.
| | - Angelo Favaloro
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98125 Messina, Italy
| | - Eugenia Rita Lauriano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Simona Pergolizzi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| |
Collapse
|
13
|
Chen X, Qiao T, Mao Z, Jia G, Zhao H, Liu G, Huang Z. Caffeic acid improves intestinal barrier functions by regulating colonic bacteria and tight junction protein expression and alleviating inflammation in weaning piglets. Anim Biotechnol 2023; 34:3693-3699. [PMID: 37067399 DOI: 10.1080/10495398.2023.2200441] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The experiment investigated the effect of caffeic acid on bacteria, short-chain fatty acids (SCFA), and the expression of tight junction protein and inflammation related genes in the colon of weaning piglets. Thirty-six weaning piglets were allocated to three treatment groups, which were fed with a basal diet, a basal diet supplemented with 250 mg/kg or 500 mg/kg caffeic acid for 28 days. The results showed that caffeic acid treatment increased the contents of acetate acid, propionate acid and total SCFA. Moreover, real-time quantitative PCR showed that the number of Bifidobacterium (p < 0.05) and Lactobacillus (p < 0.05) were increased and the number of Escherichia coli (p < 0.05) was decreased by caffeic acid in colonic mucosa. Real-time quantitative PCR also showed that the mRNA levels of zonula occludens-1 (p < 0.01), claudin-1 (p < 0.01), occludin (p < 0.01), mucin 1 (MUC1) (p < 0.01), MUC2 (p < 0.01), interleukin 4 (IL-4) (p < 0.01) and IL-10 (p < 0.05) were increased, while the mRNA expression levels of histone deacetylases (p < 0.01), IL-1 (p < 0.01), IL-6 (p < 0.01) and tumor necrosis factor-α (TNF-α) (p < 0.01) were decreased, by caffeic acid in colonic mucosa. These results suggested that caffeic acid could improve intestinal barrier function in weaned pigs, which might be mediated by regulating colonic bacteria and tight junction protein expression and alleviating inflammation.
Collapse
Affiliation(s)
- Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Tianlei Qiao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Zhengyu Mao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| |
Collapse
|
14
|
Raev SA, Raque M, Kick MK, Saif LJ, Vlasova AN. Differential transcriptome response following infection of porcine ileal enteroids with species A and C rotaviruses. Virol J 2023; 20:238. [PMID: 37848925 PMCID: PMC10580564 DOI: 10.1186/s12985-023-02207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/06/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Rotavirus C (RVC) is the major causative agent of acute gastroenteritis in suckling piglets, while most RVAs mostly affect weaned animals. Besides, while most RVA strains can be propagated in MA-104 and other continuous cell lines, attempts to isolate and culture RVC strains remain largely unsuccessful. The host factors associated with these unique RVC characteristics remain unknown. METHODS In this study, we have comparatively evaluated transcriptome responses of porcine ileal enteroids infected with RVC G1P[1] and two RVA strains (G9P[13] and G5P[7]) with a focus on innate immunity and virus-host receptor interactions. RESULTS The analysis of differentially expressed genes regulating antiviral immune response indicated that in contrast to RVA, RVC infection resulted in robust upregulation of expression of the genes encoding pattern recognition receptors including RIG1-like receptors and melanoma differentiation-associated gene-5. RVC infection was associated with a prominent upregulation of the most of glycosyltransferase-encoding genes except for the sialyltransferase-encoding genes which were downregulated similar to the effects observed for G9P[13]. CONCLUSIONS Our results provide novel data highlighting the unique aspects of the RVC-associated host cellular signalling and suggest that increased upregulation of the key antiviral factors maybe one of the mechanisms responsible for RVC age-specific characteristics and its inability to replicate in most cell cultures.
Collapse
Affiliation(s)
- Sergei A Raev
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, 44677, USA.
| | - Molly Raque
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, 44677, USA
| | - Maryssa K Kick
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, 44677, USA
| | - Linda J Saif
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, 44677, USA
| | - Anastasia N Vlasova
- Center for Food Animal Health Research Program, Department of Veterinary Preventive Medicine, College of Veterinary Medicine, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH, 44677, USA.
| |
Collapse
|
15
|
Zhang C, Wang L, Liu X, Wang G, Guo X, Liu X, Zhao J, Chen W. The Different Ways Multi-Strain Probiotics with Different Ratios of Bifidobacterium and Lactobacillus Relieve Constipation Induced by Loperamide in Mice. Nutrients 2023; 15:4230. [PMID: 37836514 PMCID: PMC10574055 DOI: 10.3390/nu15194230] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Constipation is currently one of the most common gastrointestinal disorders, and its causes are diverse. Multi-strain probiotics are often considered a more effective treatment than single-strain probiotics. In this study, a constipation model was constructed using loperamide hydrochloride to evaluate the ability of a multi-strain probiotic combination of four different ratios of Bifidobacterium and Lactobacillus to regulate intestinal flora, relieve constipation, and explore the initial mechanism in mice. After four weeks of probiotic intervention, BM1, BM2, and PB2 effectively relieved constipation; however, the pathways involved were different. The Bifidobacteria-dominated formulations BM1 and BM2 mainly changed the composition and structure of the intestinal flora and significantly decreased the relative abundance of Tyzzerella, Enterorhabdus, Faecalibaculum, Gordonibacter, and Mucispirillum in stool; increased the relative abundance of Parabacteroides and the content of short-chain fatty acids (SCFAs) in stool; restored motilin (MTL) and vasoactive intestinal peptide (VIP) levels; and downregulated interleukin 6 (IL-6) and IL-8 levels in serum. This repaired the inflammatory response caused by constipation. Finally, it promoted peristalsis of the gastrointestinal tract, increasing stool water content, and relieving constipation. While Lactobacillus-dominated formula PB2 mainly restored the levels of serum neurotransmitters (MTL, SP (substance P), VIP and PYY (Peptide YY)) and inflammatory factors (IL-1, IL-6 and IL-8), it significantly decreased the relative abundance of Tyzzerella, Enterorhabdus, Faecalibaculum, Gordonibacter and Mucispirillum in stool; it then increased acetic acid content, thereby reducing the level of inflammation and changing stool properties and gastrointestinal motility.
Collapse
Affiliation(s)
- Chenyue Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Linlin Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Xinmei Guo
- JinQiao Biotechnology Co., Ltd., Huai’an 223010, China; (X.G.); (X.L.)
| | - Xuecong Liu
- JinQiao Biotechnology Co., Ltd., Huai’an 223010, China; (X.G.); (X.L.)
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (C.Z.); (X.L.); (G.W.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
16
|
Calvigioni M, Panattoni A, Biagini F, Donati L, Mazzantini D, Massimino M, Daddi C, Celandroni F, Vozzi G, Ghelardi E. Development of an In Vitro Model of the Gut Microbiota Enriched in Mucus-Adhering Bacteria. Microbiol Spectr 2023; 11:e0033623. [PMID: 37289064 PMCID: PMC10433972 DOI: 10.1128/spectrum.00336-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023] Open
Abstract
Culturing the gut microbiota in in vitro models that mimic the intestinal environment is increasingly becoming a promising alternative approach to study microbial dynamics and the effect of perturbations on the gut community. Since the mucus-associated microbial populations in the human intestine differ in composition and functions from their luminal counterpart, we attempted to reproduce in vitro the microbial consortia adhering to mucus using an already established three-dimensional model of the human gut microbiota. Electrospun gelatin structures supplemented or not with mucins were inoculated with fecal samples and compared for their ability to support microbial adhesion and growth over time, as well as to shape the composition of the colonizing communities. Both scaffolds allowed the establishment of long-term stable biofilms with comparable total bacterial loads and biodiversity. However, mucin-coated structures harbored microbial consortia especially enriched in Akkermansia, Lactobacillus, and Faecalibacterium, being therefore able to select for microorganisms commonly considered mucosa-associated in vivo. IMPORTANCE These findings highlight the importance of mucins in shaping intestinal microbial communities, even those in artificial gut microbiota systems. We propose our in vitro model based on mucin-coated electrospun gelatin structures as a valid device for studies evaluating the effects of exogenous factors (nutrients, probiotics, infectious agents, and drugs) on mucus-adhering microbial communities.
Collapse
Affiliation(s)
- Marco Calvigioni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Adelaide Panattoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Francesco Biagini
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Leonardo Donati
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Diletta Mazzantini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mariacristina Massimino
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Costanza Daddi
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Francesco Celandroni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Vozzi
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Center “Enrico Piaggio”, University of Pisa, Pisa, Italy
| | - Emilia Ghelardi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Research Center “Nutraceuticals and Food for Health – Nutrafood”, University of Pisa, Pisa, Italy
| |
Collapse
|
17
|
Kochetkov N, Smorodinskaya S, Vatlin A, Nikiforov-Nikishin D, Nikiforov-Nikishin A, Danilenko V, Anastasia K, Reznikova D, Grishina Y, Antipov S, Marsova M. Ability of Lactobacillus brevis 47f to Alleviate the Toxic Effects of Imidacloprid Low Concentration on the Histological Parameters and Cytokine Profile of Zebrafish ( Danio rerio). Int J Mol Sci 2023; 24:12290. [PMID: 37569666 PMCID: PMC10418720 DOI: 10.3390/ijms241512290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
In the present article, the possible mitigation of the toxic effect of imidacloprid low-concentration chronic exposure on Danio rerio by the probiotic strain Lactobacillus brevis 47f (1 × 108 CFU/g) was examined. It was found that even sublethal concentration (2500 µg/L) could lead to the death of some fish during the 60-day chronic experiment. However, the use of Lactobacillus brevis 47f partially reduced the toxic effects, resulting in an increased survival rate and a significant reduction of morphohistological lesions in the intestines and kidneys of Danio rerio. The kidneys were found to be the most susceptible organ to toxic exposure, showing significant disturbances. Calculation of the histopathological index, measurement of morphometric parameters, and analysis of principal components revealed the most significant parameters affected by the combined action of imidacloprid and Lactobacillus brevis 47f. This effect of imidacloprid and the probiotic strain had a multidirectional influence on various pro/anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-8). Therefore, the results suggest the possibility of further studying the probiotic strain Lactobacillus brevis 47f as a strain that reduces the toxic effects of xenobiotics. Additionally, the study established the possibility of using imidacloprid as a model toxicant to assess the detoxification ability of probiotics on the kidney and gastrointestinal tract of fish.
Collapse
Affiliation(s)
- Nikita Kochetkov
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Svetlana Smorodinskaya
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Aleksey Vatlin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Dmitry Nikiforov-Nikishin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Alexei Nikiforov-Nikishin
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Valery Danilenko
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Klimuk Anastasia
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Faculty of Biotechnology and Fisheries, Moscow State University of Technologies and Management (FCU), 73, Zemlyanoy Val Str., 109004 Moscow, Russia;
| | - Diana Reznikova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Institutsky Lane 9, 141700 Dolgoprudny, Russia
| | - Yelena Grishina
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| | - Sergei Antipov
- Department of Biophysics and Biotechnology, Voronezh State University, University Square, 1, 394063 Voronezh, Russia;
| | - Maria Marsova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119333 Moscow, Russia; (S.S.); (A.V.); (D.N.-N.); (V.D.); (K.A.); (D.R.); (Y.G.)
| |
Collapse
|
18
|
Padoan A, Musso G, Contran N, Basso D. Inflammation, Autoinflammation and Autoimmunity in Inflammatory Bowel Diseases. Curr Issues Mol Biol 2023; 45:5534-5557. [PMID: 37504266 PMCID: PMC10378236 DOI: 10.3390/cimb45070350] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
In this review, the role of innate and adaptive immunity in the pathogenesis of inflammatory bowel diseases (IBD) is reported. In IBD, an altered innate immunity is often found, with increased Th17 and decreased Treg cells infiltrating the intestinal mucosa. An associated increase in inflammatory cytokines, such as IL-1 and TNF-α, and a decrease in anti-inflammatory cytokines, such as IL-10, concur in favoring the persistent inflammation of the gut mucosa. Autoinflammation is highlighted with insights in the role of inflammasomes, which activation by exogenous or endogenous triggers might be favored by mutations of NOD and NLRP proteins. Autoimmunity mechanisms also take place in IBD pathogenesis and in this context of a persistent immune stimulation by bacterial antigens and antigens derived from intestinal cells degradation, the adaptive immune response takes place and results in antibodies and autoantibodies production, a frequent finding in these diseases. Inflammation, autoinflammation and autoimmunity concur in altering the mucus layer and enhancing intestinal permeability, which sustains the vicious cycle of further mucosal inflammation.
Collapse
Affiliation(s)
- Andrea Padoan
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Giulia Musso
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Nicole Contran
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Daniela Basso
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
19
|
Zhang S, Zhang S, Hou Y, Huang Y, Cai J, Wang G, Cao Y, Chen Z, Fang X, Bao W. Porcine Deltacoronavirus Infection Disrupts the Intestinal Mucosal Barrier and Inhibits Intestinal Stem Cell Differentiation to Goblet Cells via the Notch Signaling Pathway. J Virol 2023; 97:e0068923. [PMID: 37289083 PMCID: PMC10308910 DOI: 10.1128/jvi.00689-23] [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: 05/09/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023] Open
Abstract
Goblet cells and their secreted mucus are important elements of the intestinal mucosal barrier, which allows host cells to resist invasion by intestinal pathogens. Porcine deltacoronavirus (PDCoV) is an emerging swine enteric virus that causes severe diarrhea in pigs and causes large economic losses to pork producers worldwide. To date, the molecular mechanisms by which PDCoV regulates the function and differentiation of goblet cells and disrupts the intestinal mucosal barrier remain to be determined. Here, we report that in newborn piglets, PDCoV infection disrupts the intestinal barrier: specifically, there is intestinal villus atrophy, crypt depth increases, and tight junctions are disrupted. There is also a significant reduction in the number of goblet cells and the expression of MUC-2. In vitro, using intestinal monolayer organoids, we found that PDCoV infection activates the Notch signaling pathway, resulting in upregulated expression of HES-1 and downregulated expression of ATOH-1 and thereby inhibiting the differentiation of intestinal stem cells into goblet cells. Our study shows that PDCoV infection activates the Notch signaling pathway to inhibit the differentiation of goblet cells and their mucus secretion, resulting in disruption of the intestinal mucosal barrier. IMPORTANCE The intestinal mucosal barrier, mainly secreted by the intestinal goblet cells, is a crucial first line of defense against pathogenic microorganisms. PDCoV regulates the function and differentiation of goblet cells, thereby disrupting the mucosal barrier; however, the mechanism by which PDCoV disrupts the barrier is not known. Here, we report that in vivo, PDCoV infection decreases villus length, increases crypt depth, and disrupts tight junctions. Moreover, PDCoV activates the Notch signaling pathway, inhibiting goblet cell differentiation and mucus secretion in vivo and in vitro. Thus, our results provide a novel insight into the mechanism underlying intestinal mucosal barrier dysfunction caused by coronavirus infection.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shuoshuo Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yuchen Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jiajia Cai
- Joint International Research Laboratory of Agriculture & Agri-Product Safety of MOE, Yangzhou University, Yangzhou, China
| | - Guangzheng Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiaomin Fang
- Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
20
|
Wojnacki J, Lujan AL, Brouwers N, Aranda-Vallejo C, Bigliani G, Rodriguez MP, Foresti O, Malhotra V. Tetraspanin-8 sequesters syntaxin-2 to control biphasic release propensity of mucin granules. Nat Commun 2023; 14:3710. [PMID: 37349283 PMCID: PMC10287693 DOI: 10.1038/s41467-023-39277-9] [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: 09/24/2022] [Accepted: 06/06/2023] [Indexed: 06/24/2023] Open
Abstract
Agonist-mediated stimulated pathway of mucin and insulin release are biphasic in which rapid fusion of pre-docked granules is followed by slow docking and fusion of granules from the reserve pool. Here, based on a cell-culture system, we show that plasma membrane-located tetraspanin-8 sequesters syntaxin-2 to control mucin release. Tetraspanin-8 affects fusion of granules during the second phase of stimulated mucin release. The tetraspanin-8/syntaxin-2 complex does not contain VAMP-8, which functions with syntaxin-2 to mediate granule fusion. We suggest that by sequestering syntaxin-2, tetraspanin-8 prevents docking of granules from the reserve pool. In the absence of tetraspanin-8, more syntaxin-2 is available for docking and fusion of granules and thus doubles the quantities of mucins secreted. This principle also applies to insulin release and we suggest a cell type specific Tetraspanin/Syntaxin combination is a general mechanism regulating the fusion of dense core granules.
Collapse
Affiliation(s)
- José Wojnacki
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Agustin Leonardo Lujan
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Nathalie Brouwers
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Carla Aranda-Vallejo
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Gonzalo Bigliani
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Maria Pena Rodriguez
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Ombretta Foresti
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Vivek Malhotra
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- ICREA, Barcelona, Spain.
| |
Collapse
|
21
|
Gorman H, Moreau F, Dufour A, Chadee K. IgGFc-binding protein and MUC2 mucin produced by colonic goblet-like cells spatially interact non-covalently and regulate wound healing. Front Immunol 2023; 14:1211336. [PMID: 37359538 PMCID: PMC10285406 DOI: 10.3389/fimmu.2023.1211336] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The colonic mucus bilayer is the first line of innate host defense that at the same time houses and nourishes the commensal microbiota. The major components of mucus secreted by goblet cells are MUC2 mucin and the mucus-associated protein, FCGBP (IgGFc-binding protein). In this study, we determine if FCGBP and MUC2 mucin were biosynthesized and interacted together to spatially enhance the structural integrity of secreted mucus and its role in epithelial barrier function. MUC2 and FCGBP were coordinately regulated temporally in goblet-like cells and in response to a mucus secretagogue but not in CRISPR-Cas9 gene-edited MUC2 KO cells. Whereas ~85% of MUC2 was colocalized with FCGBP in mucin granules, ~50% of FCGBP was diffusely distributed in the cytoplasm of goblet-like cells. STRING-db v11 analysis of the mucin granule proteome revealed no protein-protein interaction between MUC2 and FCGBP. However, FCGBP interacted with other mucus-associated proteins. FCGBP and MUC2 interacted via N-linked glycans and were non-covalently bound in secreted mucus with cleaved low molecular weight FCGBP fragments. In MUC2 KO, cytoplasmic FCGBP was significantly increased and diffusely distributed in wounded cells that healed by enhanced proliferation and migration within 2 days, whereas, in WT cells, MUC2 and FCGBP were highly polarized at the wound margin which impeded wound closure by 6 days. In DSS colitis, restitution and healed lesions in Muc2+/+ but not Muc2-/- littermates, were accompanied by a rapid increase in Fcgbp mRNA and delayed protein expression at 12- and 15-days post DSS, implicating a potential novel endogenous protective role for FCGBP in wound healing to maintain epithelial barrier function.
Collapse
Affiliation(s)
- Hayley Gorman
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - France Moreau
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Department of Microbiology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Department of Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
22
|
Overstreet AMC, Anderson B, Burge M, Zhu X, Tao Y, Cham CM, Michaud B, Horam S, Sangwan N, Dwidar M, Liu X, Santos A, Finney C, Dai Z, Leone VA, Messer JS. HMGB1 acts as an agent of host defense at the gut mucosal barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542477. [PMID: 37398239 PMCID: PMC10312563 DOI: 10.1101/2023.05.30.542477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Mucosal barriers provide the first line of defense between internal body surfaces and microbial threats from the outside world. 1 In the colon, the barrier consists of two layers of mucus and a single layer of tightly interconnected epithelial cells supported by connective tissue and immune cells. 2 Microbes colonize the loose, outer layer of colonic mucus, but are essentially excluded from the tight, epithelial-associated layer by host defenses. 3 The amount and composition of the mucus is calibrated based on microbial signals and loss of even a single component of this mixture can destabilize microbial biogeography and increase the risk of disease. 4-7 However, the specific components of mucus, their molecular microbial targets, and how they work to contain the gut microbiota are still largely unknown. Here we show that high mobility group box 1 (HMGB1), the prototypical damage-associated molecular pattern molecule (DAMP), acts as an agent of host mucosal defense in the colon. HMGB1 in colonic mucus targets an evolutionarily conserved amino acid sequence found in bacterial adhesins, including the well-characterized Enterobacteriaceae adhesin FimH. HMGB1 aggregates bacteria and blocks adhesin-carbohydrate interactions, inhibiting invasion through colonic mucus and adhesion to host cells. Exposure to HMGB1 also suppresses bacterial expression of FimH. In ulcerative colitis, HMGB1 mucosal defense is compromised, leading to tissue-adherent bacteria expressing FimH. Our results demonstrate a new, physiologic role for extracellular HMGB1 that refines its functions as a DAMP to include direct, virulence limiting effects on bacteria. The amino acid sequence targeted by HMGB1 appears to be broadly utilized by bacterial adhesins, critical for virulence, and differentially expressed by bacteria in commensal versus pathogenic states. These characteristics suggest that this amino acid sequence is a novel microbial virulence determinant and could be used to develop new approaches to diagnosis and treatment of bacterial disease that precisely identify and target virulent microbes.
Collapse
|
23
|
Masloh S, Culot M, Gosselet F, Chevrel A, Scapozza L, Zeisser Labouebe M. Challenges and Opportunities in the Oral Delivery of Recombinant Biologics. Pharmaceutics 2023; 15:pharmaceutics15051415. [PMID: 37242657 DOI: 10.3390/pharmaceutics15051415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant biological molecules are at the cutting-edge of biomedical research thanks to the significant progress made in biotechnology and a better understanding of subcellular processes implicated in several diseases. Given their ability to induce a potent response, these molecules are becoming the drugs of choice for multiple pathologies. However, unlike conventional drugs which are mostly ingested, the majority of biologics are currently administered parenterally. Therefore, to improve their limited bioavailability when delivered orally, the scientific community has devoted tremendous efforts to develop accurate cell- and tissue-based models that allow for the determination of their capacity to cross the intestinal mucosa. Furthermore, several promising approaches have been imagined to enhance the intestinal permeability and stability of recombinant biological molecules. This review summarizes the main physiological barriers to the oral delivery of biologics. Several preclinical in vitro and ex vivo models currently used to assess permeability are also presented. Finally, the multiple strategies explored to address the challenges of administering biotherapeutics orally are described.
Collapse
Affiliation(s)
- Solene Masloh
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Maxime Culot
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
24
|
View from the Biological Property: Insight into the Functional Diversity and Complexity of the Gut Mucus. Int J Mol Sci 2023; 24:ijms24044227. [PMID: 36835646 PMCID: PMC9960128 DOI: 10.3390/ijms24044227] [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: 11/29/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
Due to mucin's important protective effect on epithelial tissue, it has garnered extensive attention. The role played by mucus in the digestive tract is undeniable. On the one hand, mucus forms "biofilm" structures that insulate harmful substances from direct contact with epithelial cells. On the other hand, a variety of immune molecules in mucus play a crucial role in the immune regulation of the digestive tract. Due to the enormous number of microorganisms in the gut, the biological properties of mucus and its protective actions are more complicated. Numerous pieces of research have hinted that the aberrant expression of intestinal mucus is closely related to impaired intestinal function. Therefore, this purposeful review aims to provide the highlights of the biological characteristics and functional categorization of mucus synthesis and secretion. In addition, we highlight a variety of the regulatory factors for mucus. Most importantly, we also summarize some of the changes and possible molecular mechanisms of mucus during certain disease processes. All these are beneficial to clinical practice, diagnosis, and treatment and can provide some potential theoretical bases. Admittedly, there are still some deficiencies or contradictory results in the current research on mucus, but none of this diminishes the importance of mucus in protective impacts.
Collapse
|
25
|
Xi Y, Li Y, Ying S, Yan J, Shi Z. Bacterial lipopolysaccharide with different administration routes affects intestinal mucosal morphological, immunological, and microbial barrier functions in goslings. Poult Sci 2023; 102:102599. [PMID: 36940655 PMCID: PMC10033283 DOI: 10.1016/j.psj.2023.102599] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
The current study was conducted to evaluate the effects of different administration routes of bacterial lipopolysaccharide (LPS) on intestinal mucosal morphological, immunological, and microbial barrier functions in goslings. First, we compared intestinal villi morphology of goslings under intraperitoneal or oral LPS treatment through hematoxylin and eosin staining. Then, we determined the signatures of the microbiome in the ileum mucosa of goslings subjected to oral LPS treatment at 0, 2, 4, and 8 mg/kg BW by 16S sequencing, and analyzed the changes in intestinal barrier functions and permeability, levels of LPS in the ileum mucosa, plasma, and liver tissue, and the induced inflammatory response of Toll-like receptor 4 (TLR4). As a result, intraperitoneal LPS injection resulted in a thicker intestinal wall in the ileum within a short time, whereas villus height was less affected; in contrast, oral LPS treatment exerted a stronger influence on villus height but not on intestinal wall thickness. We also found that oral LPS treatment affected the structure of the intestinal microbiome, reflected by changes in the clustering of intestinal microbiota. The average abundance of Muribaculaceae showed an increasing trend with increasing LPS levels, and that of the genus Bacteroides decreased, compared with the control group. In addition, oral LPS treatment with 8 mg/kg BW affected the intestinal epithelial morphology, damage the mucosal immune barrier, downregulated the expression of tight junction proteins, increased circulating D-lactate levels, and stimulated the secretion of various inflammatory mediators and activation of the TLR4/MyD88/NFκB pathway. This study presented the injuries of intestinal mucosal barrier function induced by LPS challenges in goslings and provided a scientific model for searching the novel strategies to attenuate the immunological stress and gut injury caused by LPS.
Collapse
Affiliation(s)
- Yumeng Xi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yue Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Shijia Ying
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Junshu Yan
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China.
| | - Zhendan Shi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base, Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| |
Collapse
|
26
|
Nabi F, Arain MA, Fazlani SA, Khalid M, Bugti F, Ali S, Fareed SK, Liu J. Effect of In Ovo Trace Element Supplementation on Immune-Related Cells of the Small Intestine of Post-hatched Broiler Chicken. Biol Trace Elem Res 2022:10.1007/s12011-022-03492-0. [PMID: 36402885 DOI: 10.1007/s12011-022-03492-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/14/2022] [Indexed: 11/20/2022]
Abstract
Pathological conditions and harmful drugs cause many gastrointestinal diseases in broiler chicken. The current study was conducted to investigate the effect of trace elements zinc (Zn) and selenium (Se) supplementation on histomorphology, immunological role, and functional activity of goblet cells (GCs) of the small intestine. The Alcian blue-periodic acid-Schiff (AB-PAS) was performed to assess the histomorphological changes in GCs, which revealed the regular dispersion with high electron density of GCs throughout the mucosal surface in the supplemented group. However, irregular dispersion with low electron density of GCs was present in the control group. The immunological functional role of GCs within the small intestine was examined by mucicarmine staining, immunohistochemistry, and immunofluorescence. The results showed a high mucin glycol protein secretion in the supplemented group, whereas limited mucin glycol protein secretion in the control group. Furthermore, the biological significance showed a high and low immunoreactivity of Muc2 and Muc13 in the supplemented and control groups, respectively. Immunofluorescence was used to confirm the immunosignaling of Muc2. Results revealed high immunosignaling of Muc2 at the apical part of the small intestine in the supplementation group, while low immunosignaling of Muc2 in the control group. Results suggest that trace element supplementation had significant effect on morphology and immunological role of GCs, which might be essential for immune function and health status of broiler chicken.
Collapse
Affiliation(s)
- Fazul Nabi
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Balochistan, Pakistan.
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, 402460, China.
| | - Muhammad Asif Arain
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Balochistan, Pakistan
| | - Sarfaraz Ali Fazlani
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Balochistan, Pakistan
| | - Mariyam Khalid
- Department of Livestock Management, Sindh Agriculture University, Tandojam, Pakistan
| | - Firdous Bugti
- Center of Advanced Studies in Vaccinology and Biotechnology (CASVAB), University of Balochistan, Quetta, Pakistan
| | - Sikandar Ali
- Dow Institute for Advanced Biological and Animal Research, Dow University of Health Sciences, Ojha Campus, Karachi, Pakistan
| | - Syed Khurram Fareed
- Dow Institute for Advanced Biological and Animal Research, Dow University of Health Sciences, Ojha Campus, Karachi, Pakistan
| | - Juan Liu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, 402460, China.
| |
Collapse
|
27
|
Kim MY, Lee SJ, Randolph G, Han YH. Lubiprostone significantly represses fatty liver diseases via induction of mucin and HDL release in mice. Life Sci 2022; 311:121176. [DOI: 10.1016/j.lfs.2022.121176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
|
28
|
Yao W, Zhang Y, Zhang W, Wen Y, Yang R, Dong J, Zhang X, Hua Y, Ji P, Wei Y. Pathological mechanism of intestinal mucosal barrier injury of large intestine dampness-heat syndrome rats and the protective effect of Yujin powder. Res Vet Sci 2022; 152:485-496. [PMID: 36156378 DOI: 10.1016/j.rvsc.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/12/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Large intestine dampness-heat syndrome (LIDHS) is frequently-occurring in the inflammatory intestinal disease of animals and human. Yujin powder (YJP) is a classical prescription for treating LIDHS. To explore the pathological mechanism of intestinal mucosal barrier injury of LIDHS and the protection of YJP, the LIDHS rat model was established through imitating the inducing conditions of LIDHS and treated with YJP. The integrity of ileal and colonic mucosa was detected through histopathological examination. The serum DAO, D-LA and ET levels were detected by ELISA. The mRNA and protein expression levels of Occludin, ZO-1 and MUC2 in ileum and colon were detected using RT-PCR and immunohistochemistry methods, respectively. The results showed that the ileal and colonic epithelium of LIDHS rats were destroyed; the serum DAO, D-LA and ET levels were significantly increased; the mRNA and protein expression levels of Occludin, ZO-1 and MUC2 in ileum and colon were all abnormally expressed. After treatment with YJP, the mucosal integrity was restored; the levels of serum DAO, D-LA and ET, mRNA and protein levels of Occludin and ZO-1 in ileum and colon and MUC2 in ileum were back-regulated; however, MUC2 level in colon was further increased. The results demonstrated that the intestinal mucosal barrier was damaged in LIDHS rats and Occludin, ZO-1 and MUC2 were abnormally expressed, and YJP could repair the intestinal mucosal barrier through up-regulating the expression of Occludin and ZO-1 in ileum and colon as well as MUC2 in colon and down-regulating MUC2 in ileum.
Collapse
Affiliation(s)
- Wanling Yao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Yahui Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Wangdong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Yanqiao Wen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Rong Yang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Jiaqi Dong
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Xiaosong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Yongli Hua
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Peng Ji
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China
| | - Yanming Wei
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu Province 730070, China.
| |
Collapse
|
29
|
Yu S, Sun Y, Shao X, Zhou Y, Yu Y, Kuai X, Zhou C. Leaky Gut in IBD: Intestinal Barrier-Gut Microbiota Interaction. J Microbiol Biotechnol 2022; 32:825-834. [PMID: 35791076 PMCID: PMC9628915 DOI: 10.4014/jmb.2203.03022] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 12/15/2022]
Abstract
Inflammatory bowel disease (IBD) is a global disease that is in increasing incidence. The gut, which contains the largest amount of lymphoid tissue in the human body, as well as a wide range of nervous system components, is integral in ensuring intestinal homeostasis and function. By interacting with gut microbiota, immune cells, and the enteric nervous system, the intestinal barrier, which is a solid barrier, protects the intestinal tract from the external environment, thereby maintaining homeostasis throughout the body. Destruction of the intestinal barrier is referred to as developing a "leaky gut," which causes a series of changes relating to the occurrence of IBD. Changes in the interactions between the intestinal barrier and gut microbiota are particularly crucial in the development of IBD. Exploring the leaky gut and its interaction with the gut microbiota, immune cells, and the neuroimmune system may help further explain the pathogenesis of IBD and provide potential therapeutic methods for future use.
Collapse
Affiliation(s)
- Shunying Yu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China
| | - Yibin Sun
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China
| | - Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China
| | - Yuqing Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China
| | - Yang Yu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China
| | - Xiaoyi Kuai
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China,
X. Kuai Phone: +86-13776084279 E-mail:
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215001, Jiangsu, P.R. China,Corresponding authors C. Zhou Phone: +86-13962124345 E-mail:
| |
Collapse
|
30
|
Arora S, Puri S, Bhambri N. "A designer diet layout for astronauts using a microbiome mediated approach.". FEMS Microbiol Lett 2022; 369:6604380. [PMID: 35675219 DOI: 10.1093/femsle/fnac049] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/18/2022] Open
Abstract
Astronauts undergo space travel to bring scientific information to benefit humanity under various missions of space agencies such as NASA, European Space Agency, Indian Space Research Organization etc. During space missions, they encounter several stressors namely microgravity, fluid shifts, cosmic radiation, sleep deprivation and alteration in the circadian rhythm perturbing the quality of sleep. In addition, confined spaces makes pathogen interaction more likely if a pathobiont gets introduced into spacecraft. Microbiota is the first line оf resistаnсe tо vаriоus disorders and diseаses. It direсtly influenсes the biосhemiсаl, рhysiоlоgiсаl, аnd immunоlоgiсаl раthwаys. 'Gut microbiota' is essential for maintenance of healthy gut barrier functions. 'Dysbiosis' refers to perturbation of microbiota which is correlated with several metabolic and psychological disorders. Microbial metabolites are implicated in maintenance of human health. Investigations conducted on astronauts in international space missions and on analog terrestrial models have indicated a 'dysbiosis' of the gut microbiota associated with spaceflights. 'Dysbiosis' of the gut microbiome observed in astronauts has been implicated in immune dysregulation and a probiotic enriched diet is proposed to restore immune homeostasis. This article not just summarizes the state of art research on dysbiosis of the gut microbiome of astronauts, but also a diet mediated correction plan to restore their health especially during long term space missions. A characterization of microbial metabolites of the gut to enable administration of astronaut specific probiotic, postbiotic or synbiotic to alleviate space associated dysbiosis is proposed. It is also recommended that astronauts maintain a balanced nutritious diet throughout life to promote a resilient microbiota that is not perturbed by space missions. Further, a bioregenerative life support system wherein a probiotic may be produced in space station is proposed.
Collapse
Affiliation(s)
- Smriti Arora
- Department of Allied Health Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Energy Acres Building, Bidholi Dehradun, 248007 Uttarakhand, India
| | - Samikshha Puri
- Department of Allied Health Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Energy Acres Building, Bidholi Dehradun, 248007 Uttarakhand, India
| | - Nitika Bhambri
- Department of Allied Health Sciences, School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Energy Acres Building, Bidholi Dehradun, 248007 Uttarakhand, India
| |
Collapse
|
31
|
Candida Worsens Klebsiella pneumoniae Induced-Sepsis in a Mouse Model with Low Dose Dextran Sulfate Solution through Gut Dysbiosis and Enhanced Inflammation. Int J Mol Sci 2022; 23:ijms23137050. [PMID: 35806054 PMCID: PMC9266745 DOI: 10.3390/ijms23137050] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Klebsiella pneumoniae is an opportunistic pathogen and a commensal organism that is possibly enhanced in several conditions with gut dysbiosis, and frequently detectable together with Candida overgrowth. Here, K. pneumoniae with or without Candida albicans was daily orally administered for 3 months in 0.8% dextran sulfate solution-induced mucositis mice and also tested in vitro. As such, Candida worsened Klebsiella-DSS-colitis as demonstrated by mortality, leaky gut (FITC-dextran assay, bacteremia, endotoxemia, and serum beta-glucan), gut dysbiosis (increased Deferribacteres from fecal microbiome analysis), liver pathology (histopathology), liver apoptosis (activated caspase 3), and cytokines (in serum and in the internal organs) when compared with Klebsiella-administered DSS mice. The combination of heat-killed Candida plus Klebsiella mildly facilitated inflammation in enterocytes (Caco-2), hepatocytes (HepG2), and THP-1-derived macrophages as indicated by supernatant cytokines or the gene expression. The addition of heat-killed Candida into Klebsiella preparations upregulated TLR-2, reduced Occludin (an intestinal tight junction molecule), and worsened enterocyte integrity (transepithelial electrical resistance) in Caco-2 and enhanced casp8 and casp9 (apoptosis genes) in HepG2 when compared with heat-killed Klebsiella alone. In conclusion, Candida enhanced enterocyte inflammation (partly through TLR-2 upregulation and gut dysbiosis) that induced gut translocation of endotoxin and beta-glucan causing hyper-inflammatory responses, especially in hepatocytes and macrophages.
Collapse
|
32
|
Tian X, Wang G, Jin K, Ding Y, Cheng D. Rice hull insoluble dietary fiber alleviated experimental colitis induced by low dose of dextran sulfate sodium in cadmium-exposed mice. Food Funct 2022; 13:7215-7225. [PMID: 35713263 DOI: 10.1039/d2fo00891b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cadmium (Cd), an important toxic environmental pollutant, can invade the gastrointestinal tract and induce the occurrence of gastrointestinal diseases. This study aimed to investigate the protective effect of rice hull insoluble dietary fiber (RHF) on Cd-promoted colitis induced by low dose of dextran sulfate sodium. Administration of RHF attenuated inflammation by limiting Cd accumulation and regulating intestinal immune homeostasis in colitis mice with Cd exposure. RHF could maintain the structure of the gut barrier by increasing mucin secretion and intestinal tight connectivity in mice. Subsequently, RHF repressed the colonic inflammation mediated by the TLR4/MyD88/NF-κB pathway, and inhibited the transcription regulation of inflammatory cytokines. Furthermore, RHF showed an enhancement of a variety of probiotics, such as Eubacterium and Faecalibaculum. RHF also inhibited the growth of pathogenic bacteria, including Erysipelatoclostridium, Helicobacter and Bacteroides. The growth of beneficial bacteria was also accompanied by reversing the decline in short-chain fatty acids, supporting the initial potentiality of RHF as a prebiotic in cases of damage by Cd exposure in colitis mice. Importantly, RHF also remained resistant to Cd toxicity in colitis mice when the gut microbiota was depleted by antibiotics. We suggest that RHF could be used as a novel dietary supplement strategy against Cd-exacerbated colitis.
Collapse
Affiliation(s)
- Xuena Tian
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Guangliang Wang
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Kenan Jin
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Yixin Ding
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| | - Dai Cheng
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, 300457, PR China.
| |
Collapse
|
33
|
Fathima S, Shanmugasundaram R, Adams D, Selvaraj RK. Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens. Foods 2022; 11:1401. [PMID: 35626971 PMCID: PMC9140538 DOI: 10.3390/foods11101401] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 12/17/2022] Open
Abstract
The gut of warm-blooded animals is colonized by microbes possibly constituting at least 100 times more genetic material of microbial cells than that of the somatic cells of the host. These microbes have a profound effect on several physiological functions ranging from energy metabolism to the immune response of the host, particularly those associated with the gut immune system. The gut of a newly hatched chick is typically sterile but is rapidly colonized by microbes in the environment, undergoing cycles of development. Several factors such as diet, region of the gastrointestinal tract, housing, environment, and genetics can influence the microbial composition of an individual bird and can confer a distinctive microbiome signature to the individual bird. The microbial composition can be modified by the supplementation of probiotics, prebiotics, or synbiotics. Supplementing these additives can prevent dysbiosis caused by stress factors such as infection, heat stress, and toxins that cause dysbiosis. The mechanism of action and beneficial effects of probiotics vary depending on the strains used. However, it is difficult to establish a relationship between the gut microbiome and host health and productivity due to high variability between flocks due to environmental, nutritional, and host factors. This review compiles information on the gut microbiota, dysbiosis, and additives such as probiotics, postbiotics, prebiotics, and synbiotics, which are capable of modifying gut microbiota and elaborates on the interaction of these additives with chicken gut commensals, immune system, and their consequent effects on health and productivity. Factors to be considered and the unexplored potential of genetic engineering of poultry probiotics in addressing public health concerns and zoonosis associated with the poultry industry are discussed.
Collapse
Affiliation(s)
- Shahna Fathima
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Revathi Shanmugasundaram
- Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA
| | - Daniel Adams
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| | - Ramesh K. Selvaraj
- Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA; (S.F.); (D.A.); (R.K.S.)
| |
Collapse
|
34
|
Kang Y, Park H, Choe BH, Kang B. The Role and Function of Mucins and Its Relationship to Inflammatory Bowel Disease. Front Med (Lausanne) 2022; 9:848344. [PMID: 35602503 PMCID: PMC9120656 DOI: 10.3389/fmed.2022.848344] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/15/2022] [Indexed: 02/06/2023] Open
Abstract
Mucus is present throughout the gastrointestinal tract and is essential for regulating gut microbiota homeostasis and preventing disease by protecting the gastrointestinal barrier from microorganisms, pathogens and toxins or other irritants. Mucin (MUC)-2 is a secreted protein produced by epithelial goblet cells as the main component of mucus. Defects in the gastrointestinal tract, such as inflammation and ulcers, cause damage to the mucus barrier, which can worsen mucus quality and reduce mucus production. Therefore, we would like to review the characteristics of MUC2 and its role in intestinal disorders and highlight the importance of further studies. We also investigated whether the role of MUC2 differs between children and adults, ulcerative colitis (UC) and Crohn's disease (CD).
Collapse
Affiliation(s)
- Youra Kang
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hyeonjeong Park
- Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Byung-Ho Choe
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ben Kang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu, South Korea
- *Correspondence: Ben Kang
| |
Collapse
|
35
|
Solaymani-Mohammadi S. Mucosal Defense Against Giardia at the Intestinal Epithelial Cell Interface. Front Immunol 2022; 13:817468. [PMID: 35250996 PMCID: PMC8891505 DOI: 10.3389/fimmu.2022.817468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023] Open
Abstract
Human giardiasis, caused by the protozoan parasite Giardia duodenalis (syn. Giardia lamblia, Giardia intestinalis, Lamblia intestinalis), is one of the most commonly-identified parasitic diseases worldwide. Chronic G. duodenalis infections cause a malabsorption syndrome that may lead to failure to thrive and/or stunted growth, especially in children in developing countries. Understanding the parasite/epithelial cell crosstalk at the mucosal surfaces of the small intestine during human giardiasis may provide novel insights into the mechanisms underlying the parasite-induced immunopathology and epithelial tissue damage, leading to malnutrition. Efforts to identify new targets for intervening in the development of intestinal immunopathology and the progression to malnutrition are critical. Translating these findings into a clinical setting will require analysis of these pathways in cells and tissues from humans and clinical trials could be devised to determine whether interfering with unwanted mucosal immune responses developed during human giardiasis provide better therapeutic benefits and clinical outcomes for G. duodenalis infections in humans.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
36
|
Livzan MA, Bicbavova GR, Romanyuk AE. Ulcerative colitis: focus on colonic mucosal resistance. BULLETIN OF SIBERIAN MEDICINE 2022; 21:121-132. [DOI: 10.20538/1682-0363-2022-1-121-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
In recent decades, following cooperation between scientists in various specialties, new unique data on the pathogenesis of ulcerative colitis have been obtained. The role of an impaired immune response to antigens of gut microbiota in genetically predisposed individuals under the effect of certain environmental factors was proven. Assessing the interaction between the colonic mucosa and gut microbiota will help to understand the mechanisms of ulcerative colitis and develop new treatment strategies for the disease.This review presents modern views on the pathogenesis of ulcerative colitis with a focus on the imbalance between local protective and aggressive factors of the gastric and intestinal mucosa. The structure and role of the epithelial barrier both under normal conditions and in ulcerative colitis are considered in detail.The aim of this review was to summarize the data on resistance of the colonic mucosa and its damage in ulcerative colitis.
Collapse
|
37
|
Xu H, Lao L, Ji C, Lu Q, Guo Y, Pan D, Wu Z. Anti-inflammation and adhesion enhancement properties of the multifunctional LPxTG-motif surface protein derived from the Lactobacillus reuteri DSM 8533. Mol Immunol 2022; 146:38-45. [PMID: 35421739 DOI: 10.1016/j.molimm.2022.04.004] [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: 12/10/2021] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
LPxTG-motif protein (LMP) is one kind of a precursor protein that contains a conserved LPxTG-motif at the C-terminus, which can be recognized by sortase A (SrtA) and covalently bind to the bacterial peptidoglycan. In this study, LMP derived from Lactobacillus reuteri (L. reuteri) was heterologous expressed and the tolerance and intestinal colonization ability of the LMP on L. reuteri were analyzed in simulated gastrointestinal fluid. Meanwhile, the anti-inflammatory activity of LMP was also evaluated in the LPS-stimulated RAW 264.7 cell model. The results indicated that LMP can promote the intestinal survival rate and adhesion characteristics of L. reuteri and enhanced the autoinducer-2 (AI-2) signaling molecule of the Lactobacillus strains in quorum sensing. Furthermore, LMP can inhibit the expressions of inflammatory cytokine TNF-α and IL-1β via ERK-JNK related MAPK signaling cascades. These findings provide a better understanding of the multifunctional LPxTG-motif surface protein derived from L. reuteri in the gastrointestinal tract environment.
Collapse
Affiliation(s)
- Hai Xu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Lifeng Lao
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Chunyu Ji
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Qianqian Lu
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Yuxing Guo
- School of Food Science & Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Daodong Pan
- Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zhen Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd., Shanghai, China; Key Laboratory of Animal Protein Deep Processing Technology of Zhejiang, School of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
38
|
Latek U, Chłopecka M, Karlik W, Mendel M. Phytogenic Compounds for Enhancing Intestinal Barrier Function in Poultry-A Review. PLANTA MEDICA 2022; 88:218-236. [PMID: 34331305 DOI: 10.1055/a-1524-0358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
After the European Union ban of antibiotic growth promoters, works on different methods of improving gut health have intensified. The poultry industry is struggling with problems that were previously controlled by antibiotic growth promoters, therefore the search for optimal solutions continues. Simultaneously, there is also increasing social pressure to minimize the use of antibiotics and replace them with alternative feed additives. A variety of available alternatives is considered safe by consumers, among which phytogenics play a significant role. However, there are still some limitations that need to be considered. The most questionable are the issues related to bioavailability, metabolism of plant derivatives in birds, and the difficulty of standardizing commercial products. There is still a need for more evidence-based recommendations for the use of phytogenics in livestock. On the other hand, a positive influence of phytogenic compounds on the health of poultry has been previously described by many researchers and practical application of these compounds has auspicious perspectives in poultry production. Supplementation with phytogenic feed additives has been shown to protect birds from various environmental threats leading to impaired intestinal barrier function. Phytogenic feed additives have the potential to improve the overall structure of intestinal mucosa as well as gut barrier function on a molecular level. Recognition of the phytogenics' effect on the components of the intestinal barrier may enable the selection of the most suitable ones to alleviate negative effects of different agents. This review aims to summarize current knowledge of the influence of various phytogenic constituents on the intestinal barrier and health of poultry.
Collapse
Affiliation(s)
- Urszula Latek
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Magdalena Chłopecka
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Wojciech Karlik
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Marta Mendel
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| |
Collapse
|
39
|
Yao Q, Fan L, Zheng N, Blecker C, Delcenserie V, Li H, Wang J. 2'-Fucosyllactose Ameliorates Inflammatory Bowel Disease by Modulating Gut Microbiota and Promoting MUC2 Expression. Front Nutr 2022; 9:822020. [PMID: 35252301 PMCID: PMC8892212 DOI: 10.3389/fnut.2022.822020] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota dysbiosis, together with goblet cells dysfunction has been observed in ulcerative colitis cases. This study aims to evaluate the potential of 2'-fucosyllactose (2'-FL) supplementation in inhibiting intestinal inflammation through regulating gut microbiota, protecting goblet cells, and stimulating mucin secretion. 2'-FL was orally administered to C57BL/6J mice daily (400 mg/kg bw) for 21 days and 5% dextran sulfate sodium (DSS) was used to induce the colitis in the last 7 days. Meanwhile, fecal microbiota transplantation (FMT) was conducted to test the roles of gut microbiota in the remission of colitis by 2'-FL. Gut microbiota alteration was analyzed through 16S ribosomal RNA (16S rRNA) sequencing. Periodic acid-Schiff (PAS), immunofluorescence staining, as well as mucin 2 (MUC2) and NOD-like receptor family pyrin domain containing 6 (NLRP6) messenger RNA (mRNA) expression in colon fragments was performed and detected. The results showed that the DSS + 2'-FL mice were found to have a slower rate of weight loss, lower disease activity index (DAI) scores, and longer colon lengths than the DSS group (p < 0.05), so in the FMT recipient mice which received fecal microbiota from the DSS + 2'-FL group. In addition, the data revealed that 2'-FL relieved the disorder of DSS-induced gut microbiota, including decreasing the high abundance of mucin-utilizing bacteria in the DSS group, such as Bacteroides, Lachnospiraceae NK4A136, Lachnospiraceae, and Bacteroides vulgatus. PAS and immunofluorescence staining showed that 2'-FL treatment promoted the recovery of goblet cells and enhanced MUC2 and NLRP6 expression, which was also observed in the FM (DSS + 2'-FL) group. Moreover, NLRP6, which has been proved to be a negative regulator for Toll-like receptor 4/myeloid differential protein-8/nuclear factor-kappa B (TLR4/MyD88/NF-κB) pathway, was upregulated by 2'-FL in colon tissue. In conclusion, this study suggests that 2'-FL ameliorates colitis in a gut microbiota-dependent manner. The underlying protective mechanism associates with the recovery of goblet cells number and improves MUC2 secretion through TLR4-related pathway.
Collapse
Affiliation(s)
- Qianqian Yao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium,Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Linlin Fan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Nan Zheng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | - Véronique Delcenserie
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Huiying Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,*Correspondence: Huiying Li
| | - Jiaqi Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China,Jiaqi Wang
| |
Collapse
|
40
|
Mucins Dynamics in Physiological and Pathological Conditions. Int J Mol Sci 2021; 22:ijms222413642. [PMID: 34948435 PMCID: PMC8707880 DOI: 10.3390/ijms222413642] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Maintaining intestinal health requires clear segregation between epithelial cells and luminal microbes. The intestinal mucus layer, produced by goblet cells (GCs), is a key element in maintaining the functional protection of the epithelium. The importance of the gut mucus barrier is highlighted in mice lacking Muc2, the major form of secreted mucins. These mice show closer bacterial residence to epithelial cells, develop spontaneous colitis and became moribund when infected with the attaching and effacing pathogen, Citrobacter rodentium. Furthermore, numerous observations have associated GCs and mucus layer dysfunction to the pathogenesis of inflammatory bowel disease (IBD). However, the molecular mechanisms that regulate the physiology of GCs and the mucus layer remain obscured. In this review, we consider novel findings describing divergent functionality and expression profiles of GCs subtypes within intestinal crypts. We also discuss internal (host) and external (diets and bacteria) factors that modulate different aspects of the mucus layer as well as the contribution of an altered mucus barrier to the onset of IBD.
Collapse
|
41
|
Zhang P, Jing C, Liang M, Jiang S, Huang L, Jiao N, Li Y, Yang W. Zearalenone Exposure Triggered Cecal Physical Barrier Injury through the TGF-β1/Smads Signaling Pathway in Weaned Piglets. Toxins (Basel) 2021; 13:toxins13120902. [PMID: 34941739 PMCID: PMC8708673 DOI: 10.3390/toxins13120902] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
This study aims to investigate the effects of exposure to different dosages of zearalenone (ZEA) on cecal physical barrier functions and its mechanisms based on the TGF-β1/Smads signaling pathway in weaned piglets. Thirty-two weaned piglets were allotted to four groups and fed a basal diet supplemented with ZEA at 0, 0.15, 1.5, and 3.0 mg/kg, respectively. The results showed that 1.5 and 3.0 mg/kg ZEA damaged cecum morphology and microvilli, and changed distribution and shape of M cells. Moreover, 1.5 and 3.0 mg/kg ZEA decreased numbers of goblet cells, the expressions of TFF3 and tight junction proteins, and inhibited the TGF-β1/Smads signaling pathway. Interestingly, the 0.15 mg/kg ZEA had no significant effect on cecal physical barrier functions but decreased the expressions of Smad3, p-Smad3 and Smad7. Our study suggests that high-dose ZEA exposure impairs cecal physical barrier functions through inhibiting the TGF-β1/Smads signaling pathway, but low-dose ZEA had no significant effect on cecum morphology and integrity through inhibiting the expression of smad7. These findings provide a scientific basis for helping people explore how to reduce the toxicity of ZEA in feeds.
Collapse
Affiliation(s)
- Pengfei Zhang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Changwei Jing
- Technical Department, Shandong Chinwhiz Co., Ltd., Weifang 262400, China;
| | - Ming Liang
- Department of Feeding Microecology, Shandong Baolaililai Bioengineering Co., Ltd., Tai’an 271001, China;
| | - Shuzhen Jiang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Libo Huang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Ning Jiao
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
| | - Yang Li
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
- Correspondence: (Y.L.); (W.Y.)
| | - Weiren Yang
- Department of Animal Sciences and Technology, Shandong Agricultural University, Tai’an 271018, China; (P.Z.); (S.J.); (L.H.); (N.J.)
- Correspondence: (Y.L.); (W.Y.)
| |
Collapse
|
42
|
Panpetch W, Phuengmaung P, Cheibchalard T, Somboonna N, Leelahavanichkul A, Tumwasorn S. Lacticaseibacillus casei Strain T21 Attenuates Clostridioides difficile Infection in a Murine Model Through Reduction of Inflammation and Gut Dysbiosis With Decreased Toxin Lethality and Enhanced Mucin Production. Front Microbiol 2021; 12:745299. [PMID: 34925261 PMCID: PMC8672038 DOI: 10.3389/fmicb.2021.745299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Clostridioides difficile is a major cause of diarrhea in patients with antibiotic administration. Lacticaseibacillus casei T21, isolated from a human gastric biopsy, was tested in a murine C. difficile infection (CDI) model and colonic epithelial cells (Caco-2 and HT-29). Daily administration of L. casei T21 [1 × 108 colony forming units (CFU)/dose] for 4 days starting at 1 day before C. difficile challenge attenuated CDI as demonstrated by a reduction in mortality rate, weight loss, diarrhea, gut leakage, gut dysbiosis, intestinal pathology changes, and levels of pro-inflammatory cytokines [interleukin (IL)-1β, tumor necrosis factor (TNF)-α, macrophage inflammatory protein 2 (MIP-2), and keratinocyte chemoattractant (KC)] in the intestinal tissue and serum. Conditioned media from L. casei T21 exerted biological activities that fight against C. difficile as demonstrated in colonic epithelial cells by the following: (i) suppression of gene expression and production of IL-8, an important chemokine involved in C. difficile pathogenesis, (ii) reduction in the expression of SLC11A1 (solute carrier family 11 member 1) and HuR (human antigen R), important genes for the lethality of C. difficile toxin B, (iii) augmentation of intestinal integrity, and (iv) up-regulation of MUC2, a mucosal protective gene. These results supported the therapeutic potential of L. casei T21 for CDI and the need for further study on the intervention capabilities of CDI.
Collapse
Affiliation(s)
- Wimonrat Panpetch
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thanya Cheibchalard
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Naraporn Somboonna
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Immunology and Immune-Mediated Diseases, Bangkok, Thailand
- *Correspondence: Asada Leelahavanichkul,
| | - Somying Tumwasorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok, Thailand
- Somying Tumwasorn,
| |
Collapse
|
43
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Corrigendum: Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:790387. [PMID: 34790692 PMCID: PMC8591313 DOI: 10.3389/fnut.2021.790387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
44
|
Galosi L, Desantis S, Roncarati A, Robino P, Bellato A, Nebbia P, Ferrocino I, Santamaria N, Biagini L, Filoni L, Attili AR, Rossi G. Positive Influence of a Probiotic Mixture on the Intestinal Morphology and Microbiota of Farmed Guinea Fowls ( Numida meleagris). Front Vet Sci 2021; 8:743899. [PMID: 34778432 PMCID: PMC8586554 DOI: 10.3389/fvets.2021.743899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/28/2021] [Indexed: 11/15/2022] Open
Abstract
To understand the effectiveness of a probiotic mixture on intestinal morphology, mucus layer composition, and cecal microbiota diversity, 40 10-day-old Guinea fowls (Numida meleagris) were assigned to two groups: the control group (C), receiving drinking water, and the treated group (P), receiving water plus a commercial multi-strain probiotic (Slab51®, 2 × 1011 CFU/L). Birds were slaughtered after 4 months, and the intestines were collected. Samples from the duodenum, ileum, and cecum were processed for morphological and morphometric studies, and conventional glycohistochemistry. Cecal samples were also used to assess the microbiota by 16S metataxonomic approach. Group P showed significant increase in the villus height (p < 0.001 in the duodenum and p < 0.05 in the ileum and cecum), villus width (p < 0.05 in all investigated tracts), depth of crypts (p < 0.001 in the duodenum and cecum; p < 0.05 in the ileum), and goblet cells per villus (p < 0.001 in all investigated tracts) compared with group C. Cecal microbiota of the birds varied considerably and comparing the relative abundance of the main observational taxonomic units (OTUs), a positive enrichment of several beneficial taxa, such as Oscillospira, Eubacterium, Prevotella, and members of the Ruminococcaceae, was observed. The enrichment of those taxa can improve microbiota stability and resilience facing environmental stresses, enhancing its resistance against invading pathogens. Ruminococcaceae, which represent the most important taxon in both groups, and Prevotella have a key role in the gut physiology due to the production of short-chain fatty acids (SCFAs), which are a vital energy source for enterocytes, improve glucose metabolism, and exert an overall anti-inflammatory effect. Probiotic administration enriches the presence of Coprococcus, Oscillospira, and Eubacterium taxa that produce butyrate, which exerts a beneficial effect on growth performance, structure of villi, and pathogen control and has anti-inflammatory properties too. This study indicates that Slab51® supplementation positively affects the morphology and microbiota diversity of the guinea fowl intestine.
Collapse
Affiliation(s)
- Livio Galosi
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Salvatore Desantis
- Department of Emergency and Organ Trasplants (DETO), University of Bari Aldo Moro, Valenzano, Italy
| | - Alessandra Roncarati
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Patrizia Robino
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Alessandro Bellato
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Patrizia Nebbia
- Department of Veterinary Sciences, University of Torino, Grugliasco, Italy
| | - Ilario Ferrocino
- Department of Agriculture, Forestry and Food Science, University of Torino, Grugliasco, Italy
| | - Nicoletta Santamaria
- Department of Emergency and Organ Trasplants (DETO), University of Bari Aldo Moro, Valenzano, Italy
| | - Lucia Biagini
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Lorenzo Filoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Anna Rita Attili
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy
| |
Collapse
|
45
|
Driouich A, Gaudry A, Pawlak B, Moore JP. Root cap-derived cells and mucilage: a protective network at the root tip. PROTOPLASMA 2021; 258:1179-1185. [PMID: 34196784 DOI: 10.1007/s00709-021-01660-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/27/2021] [Indexed: 05/06/2023]
Abstract
Root cap-derived cells and mucilage provide the first line of defense of the plant against soil microbial pathogens. These cells form a mucilaginous root extracellular trap (RET), which also harbors a range of molecules including exDNA and defensive peptides and proteins much like the neutrophil extracellular trap (NET) of mammalians. Plant RETs resemble mucus structures found in mammalian systems and are rich in arabinogalactan proteins that have similarities to highly glycosylated human mucins. Human mucus and mucins regulate the intestinal flora microbiome through recruiting certain species of microbes and it is plausible that the arabinogalactan protein-rich mucilage found in plant roots fulfills a similar function by attracting specific microbes to the rhizosphere. The role of RETs in root defense functioning is highlighted.
Collapse
Affiliation(s)
- Azeddine Driouich
- UNIROUEN, Normandie Université, Laboratoire Glycobiologie Et Matrice Extracellulaire Végétale EA 4358, Université de Rouen Normandie, 76000, Rouen, France.
- UNIROUEN, Fédération de Recherche, Normandie Université, Normandie Végétal-FED 4277, Université de Rouen Normandie, 76000, Rouen, France.
| | - Alexia Gaudry
- UNIROUEN, Normandie Université, Laboratoire Glycobiologie Et Matrice Extracellulaire Végétale EA 4358, Université de Rouen Normandie, 76000, Rouen, France
- UNIROUEN, Fédération de Recherche, Normandie Université, Normandie Végétal-FED 4277, Université de Rouen Normandie, 76000, Rouen, France
| | - Barbara Pawlak
- UNIROUEN, Normandie Université, Laboratoire Glycobiologie Et Matrice Extracellulaire Végétale EA 4358, Université de Rouen Normandie, 76000, Rouen, France
- UNIROUEN, Fédération de Recherche, Normandie Université, Normandie Végétal-FED 4277, Université de Rouen Normandie, 76000, Rouen, France
| | - John P Moore
- Department of Viticulture and Oenology, Faculty of AgriSciences, South African Grape and Wine Research Institute, Stellenbosch University, Matieland, 7602, South Africa
| |
Collapse
|
46
|
Xu Y, Bai T, Xiong Y, Liu C, Liu Y, Hou X, Song J. Mechanical stimulation activates Piezo1 to promote mucin2 expression in goblet cells. J Gastroenterol Hepatol 2021; 36:3127-3139. [PMID: 34169583 DOI: 10.1111/jgh.15596] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/26/2021] [Accepted: 06/19/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIM Studies on the regulation of mucin2 expression in intestinal goblet cells by the endocrine system and the immune system have been comprehensive, but the effects of abundant mechanical factors in the intestinal microenvironment on goblet cells are not clear. METHODS We constructed mechanical stimulation models in vivo and in vitro to explore the effect of mechanical stimulation on intestinal goblet cells. Piezo1 expression and function were regulated through model mouse and drugs to explored whether Piezo1 mediated mechanical stimulation. RESULTS The results showed that hydrostatic pressure could promote mucus secretion in the mouse colon, and both traction force and shear force could promote the expression of mucin2 in the LS174T cell line. We further found that the Piezo1 protein, which was abundantly expressed in goblet cells, acted as a mechanoreceptor. Knockout of Piezo1 in the intestinal epithelial cells of mice could reduce the promotion of mucus secretion by pressure stimulation, and the specific downregulation of Piezo1 protein in LS174T cells or Piezo1 inhibitor treatment could significantly reduce the promotion of mucin2 expression in goblet cells by mechanical stimulation; however, treatment with a Piezo1 agonist had the opposite effect. Moreover, we found that Piezo1 regulated mucin2 expression through the downstream Erk/Ca2+ pathway. CONCLUSION In short, our study confirmed for the first time that goblet cells are mechanoreceptive cells that can directly sense mechanical stimulation in the intestinal tract and respond back through the Piezo1-Erk/Ca2+ -mucin2 pathway.
Collapse
Affiliation(s)
- Yan Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Bai
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Xiong
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caiyuan Liu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Liu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:718356. [PMID: 34589512 PMCID: PMC8475765 DOI: 10.3389/fnut.2021.718356] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The intestinal epithelial barrier (IEB) is one of the largest interfaces between the environment and the internal milieu of the body. It is essential to limit the passage of harmful antigens and microorganisms and, on the other side, to assure the absorption of nutrients and water. The maintenance of this delicate equilibrium is tightly regulated as it is essential for human homeostasis. Luminal solutes and ions can pass across the IEB via two main routes: the transcellular pathway or the paracellular pathway. Tight junctions (TJs) are a multi-protein complex responsible for the regulation of paracellular permeability. TJs control the passage of antigens through the IEB and have a key role in maintaining barrier integrity. Several factors, including cytokines, gut microbiota, and dietary components are known to regulate intestinal TJs. Gut microbiota participates in several human functions including the modulation of epithelial cells and immune system through the release of several metabolites, such as short-chain fatty acids (SCFAs). Mediators released by immune cells can induce epithelial cell damage and TJs dysfunction. The subsequent disruption of the IEB allows the passage of antigens into the mucosa leading to further inflammation. Growing evidence indicates that dysbiosis, immune activation, and IEB dysfunction have a role in several diseases, including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and gluten-related conditions. Here we summarize the interplay between the IEB and gut microbiota and mucosal immune system and their involvement in IBS, IBD, and gluten-related disorders.
Collapse
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
48
|
Gut Microbiota and Dietary Factors as Modulators of the Mucus Layer in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:ijms221910224. [PMID: 34638564 PMCID: PMC8508624 DOI: 10.3390/ijms221910224] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
The gastrointestinal tract is optimized to efficiently absorb nutrients and provide a competent barrier against a variety of lumen environmental compounds. Different regulatory mechanisms jointly collaborate to maintain intestinal homeostasis, but alterations in these mechanisms lead to a dysfunctional gastrointestinal barrier and are associated to several inflammatory conditions usually found in chronic pathologies such as inflammatory bowel disease (IBD). The gastrointestinal mucus, mostly composed of mucin glycoproteins, covers the epithelium and plays an essential role in digestive and barrier functions. However, its regulation is very dynamic and is still poorly understood. This review presents some aspects concerning the role of mucus in gut health and its alterations in IBD. In addition, the impact of gut microbiota and dietary compounds as environmental factors modulating the mucus layer is addressed. To date, studies have evidenced the impact of the three-way interplay between the microbiome, diet and the mucus layer on the gut barrier, host immune system and IBD. This review emphasizes the need to address current limitations on this topic, especially regarding the design of robust human trials and highlights the potential interest of improving our understanding of the regulation of the intestinal mucus barrier in IBD.
Collapse
|
49
|
Mucolytic bacteria: prevalence in various pathological diseases. World J Microbiol Biotechnol 2021; 37:176. [PMID: 34519941 DOI: 10.1007/s11274-021-03145-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023]
Abstract
All mucins are highly glycosylated and a key constituent of the mucus layer that is vigilant against pathogens in many organ systems of animals and humans. The viscous layer is organized in bilayers, i.e., an outer layer that is loosely arranged, variable in thickness, home to the commensal microbiota that grows in the complex environment, and an innermost layer that is stratified, non-aspirated, firmly adherent to the epithelial cells and devoid of any microorganisms. The O-glycosylation moiety represents the site of adhesion for pathogens and due to the increase of motility, mucolytic activity, and upregulation of virulence factors, some microorganisms can circumvent the component of the mucus layer and cause disruption in organ homeostasis. A dysbiotic microbiome, defective mucus barrier, and altered immune response often result in various diseases. In this review, paramount emphasis is given to the role played by the bacterial species directly or indirectly involved in mucin degradation, alteration in mucus secretion or its composition or mucin gene expression, which instigates many diseases in the digestive, respiratory, and other organ systems. A systematic view can help better understand the etiology of some complex disorders such as cystic fibrosis, ulcerative colitis and expand our knowledge about mucin degraders to develop new therapeutic approaches to correct ill effects caused by these mucin-dwelling pathogens.
Collapse
|
50
|
Morales-Ferré C, Azagra-Boronat I, Massot-Cladera M, Tims S, Knipping K, Garssen J, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Effects of a Postbiotic and Prebiotic Mixture on Suckling Rats' Microbiota and Immunity. Nutrients 2021; 13:2975. [PMID: 34578853 PMCID: PMC8469903 DOI: 10.3390/nu13092975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 01/04/2023] Open
Abstract
Human milk serves as a model for infant formula providing nutritional solutions for infants not able to receive enough mother's milk. Infant formulas aim to mimic the composition and functionality of human milk by providing ingredients reflecting those of the latest human milk insights, such as prebiotics, probiotics and postbiotics. The aim of this study was to examine the effects of the supplementation with a postbiotic (LactofidusTM) and its combination with the prebiotics short-chain galactooligosaccharides (scGOS) and long-chain fructooligosaccharides (lcFOS) in a preclinical model of healthy suckling rats. Pups were supplemented daily with LactofidusTM (POST group) and/or scGOS/lcFOS (P+P and PRE groups, respectively). Body weight and fecal consistency were analyzed. At the end of the study, immunoglobulin (Ig) profile, intestinal gene expression, microbiota composition and short chain fatty acid (SCFA) proportion were quantified. The supplementation with all nutritional interventions modulated the Ig profile, but the prebiotic mixture and the postbiotic induced differential effects: whereas scGOS/lcFOS induced softer feces and modulated microbiota composition and SCFA profile, Lactofidus™ upregulated Toll-like receptors gene expression. The use of the combination of scGOS/lcFOS and Lactofidus™ showed the effects observed for the oligosaccharides separately, as well as showing a synergistic impact on animal growth. Thus, the combined use of both products seems to be a good strategy to modulate immune and microbial features in early life.
Collapse
Affiliation(s)
- Carla Morales-Ferré
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Sebastian Tims
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (S.T.); (K.K.); (J.G.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 PB Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (C.M.-F.); (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|