1
|
Chen Y, Liu N, Chen F, Liu M, Mu Y, Wang C, Xia L, Peng M, Zhou M. Alleviation effects of Lactobacillus plantarum in colitis aggravated by a high-salt diet depend on intestinal barrier protection, NF-κB pathway regulation, and oxidative stress improvement. Food Funct 2025; 16:2718-2736. [PMID: 40111251 DOI: 10.1039/d4fo06377e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
A high-salt diet (HSD) can result in numerous health issues, including exacerbation of intestinal inflammation. Therefore, there is an immediate necessity of developing dietary supplements that can mitigate colitis exacerbated by a HSD. This study examined the impact of Lactobacillus plantarum HGD228 on colitis exacerbated by a HSD and the mechanisms underlying its alleviation. HGD228 treatment significantly enhanced colonic goblet cells and MUC2, upregulated ZO-1 and occludin, inhibited epithelial cell apoptosis, and mitigated colitis exacerbated by a HSD. Moreover, HGD228 significantly regulated oxidative stress-related enzymes, including SOD, GSH-PX, and CAT. HGD228 treatment significantly suppressed the NF-κB pathway induced by a HSD and regulated the levels of cytokines, including TNF-α, IL-10, and IL-1β. Furthermore, HGD228 reestablished the gut microbiota altered by HSDDSS, increasing Bifidobacterium while decreasing Escherichia-Shigella and Clostridium sensu stricto 1. HGD228 treatment also enhanced the production of butyric acid and acetic acid, suppressed pro-inflammatory cytokines, and strengthened the intestinal mucosal barrier. Therefore, HGD228 enhanced the production of beneficial metabolites by regulating inflammatory cytokines and oxidative stress, preserving the mucosal barrier, and enhancing gut microbiota, and mitigated colitis aggravated by a HSD. These results will aid in clinical trials of probiotics and the development of dietary supplements for colitis, with promising application value.
Collapse
Affiliation(s)
- Yang Chen
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Nian Liu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Fangyi Chen
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Mengyuan Liu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Yang Mu
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Chao Wang
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Lusha Xia
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430068, China
| | - Mingye Peng
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| | - Mengzhou Zhou
- Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, Hubei, 430068, China.
| |
Collapse
|
2
|
Shang X, Guo J, Chen P. Effect of Food Matrix on Regulation of Intestinal Barrier and Microbiota Homeostasis by Polysaccharides Sulfated Carrageenan. Foods 2025; 14:635. [PMID: 40002079 PMCID: PMC11854102 DOI: 10.3390/foods14040635] [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: 01/04/2025] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Carrageenan (CGN) has side effects on the intestinal barrier. Damage to the intestinal barrier is associated with exposure to sulfate groups. Food matrix has significant influence on the exposure quantity of sulfate groups and conformation in κ-CGN, but the corresponding side effects are not reported specifically. This study aimed to explore the regulatory effect of κ-CGN dissolved in aqueous (κ-CGN) and in 3% casein (κ-carrageenan-casein, κ-CC) on the intestinal barrier and microbiota homeostasis. Research has shown that both κ-CGN and κ-CC can induce different extents of intestinal barrier damage through disrupting microbiota homeostasis. Importantly, κ-CGN in casein with lower sulfate groups content was found to repair the intestinal barrier injury induced by an equivalent dose of κ-CGN aqueous through increasing the abundance of Oscillibacter and decreasing Weissella. These alleviating effects were reflected in lower levels of tumor necrosis factor (TNF)-α and C-reaction protein (CRP), higher levels of interleukin (IL)-10, raised secretion of mucus and goblet cells, and improved expression of epithelial cell compact proteins zonula occluden (ZO)-1 and mucin protein 2 (MUC2). This study states that κ-CGN in casein has a positive regulatory effect on the intestinal barrier damage compared to in aqueous solution, which can provide guidance for processing and utilization of CGN.
Collapse
Affiliation(s)
- Xuke Shang
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Juanjuan Guo
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| | - Peilin Chen
- College of Oceanology and Food Sciences, Quanzhou Normal University, Quanzhou 362000, China; (X.S.); (P.C.)
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, Quanzhou 362000, China
- Key Laboratory of Inshore Resources Biotechnology, Quanzhou Normal University, Quanzhou 362000, China
| |
Collapse
|
3
|
Salvador-Erro J, Pastor Y, Gamazo C. Targeting Enterotoxins: Advancing Vaccine Development for Enterotoxigenic Escherichia coli ETEC. Toxins (Basel) 2025; 17:71. [PMID: 39998088 PMCID: PMC11860656 DOI: 10.3390/toxins17020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal disease worldwide, particularly in children in low- and middle-income countries. Its ability to rapidly colonize the intestinal tract through diverse colonization factors and toxins underpins its significant public health impact. Despite extensive research and several vaccine candidates reaching clinical trials, no licensed vaccine exists for ETEC. This review explores the temporal and spatial coordination of ETEC virulence factors, focusing on the interplay between adherence mechanisms and toxin production as critical targets for therapeutic intervention. Advancements in molecular biology and host-pathogen interaction studies have uncovered species-specific variations and cross-reactivity between human and animal strains. In particular, the heat-labile (LT) and heat-stable (ST) toxins have provided crucial insights into molecular mechanisms and intestinal disruption. Additional exotoxins, such as EAST-1 and hemolysins, further highlight the multifactorial nature of ETEC pathogenicity. Innovative vaccine strategies, including multiepitope fusion antigens (MEFAs), mRNA-based approaches, and glycoconjugates, aim to enhance broad-spectrum immunity. Novel delivery methods, like intradermal immunization, show promise in eliciting robust immune responses. Successful vaccination against ETEC will offer an effective and affordable solution with the potential to greatly reduce mortality and prevent stunting, representing a highly impactful and cost-efficient solution to a critical global health challenge.
Collapse
Affiliation(s)
| | | | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (J.S.-E.); (Y.P.)
| |
Collapse
|
4
|
Damianos J, Abdelnaem N, Camilleri M. Gut Goo: Physiology, Diet, and Therapy of Intestinal Mucus and Biofilms in Gastrointestinal Health and Disease. Clin Gastroenterol Hepatol 2025; 23:205-215. [PMID: 39426645 PMCID: PMC11761393 DOI: 10.1016/j.cgh.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The gastrointestinal tract has remarkable capacity to withstand considerable insults from exposure to abrasive food particles, chemicals, allergens, and pathogenic microbes. Maintaining a robust epithelial barrier sequesters these potentially harmful substances in the lumen, preventing absorption into the systemic circulation. Normal functioning of this barrier is central in diverse physiological processes including digestion, immunity, inflammation, and gut-brain signaling. One crucial component of the barrier is the mucus layer covering the epithelium. There is increased appreciation of the importance of mucus in maintenance of the gut barrier, and how dysregulation of the mucus layer contributes to several common gastrointestinal pathologies. This manuscript reviews the physical and chemical properties of mucus, its maintenance and turnover, and its role in maintaining gut barrier integrity. The dynamic interactions of the mucus layer within the gut ecosystem are illustrated by highlighting how a weakened mucus layer or defective mucus production facilitate pathogenic microbial colonization and mucosal biofilm formation. These may potentially contribute to the pathogenesis of gastrointestinal diseases such as inflammatory bowel diseases or result in secretion and mucosal damage and inflammation in bile acid diarrhea. A final goal is to review how certain dietary factors, especially low-fiber diets and emulsifiers common in Western diets, can harm the mucus layer. This report summarizes evidence from preclinical and human studies that document damage to the mucus layer, and reviews approaches, including diets and probiotics, that promote a healthy mucus layer and break down pathogenic biofilms, thereby potentially preventing and/or treating gastrointestinal diseases that impact mucosal integrity.
Collapse
Affiliation(s)
- John Damianos
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Nada Abdelnaem
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
5
|
Xu H, Zhang T, Zhou Z, Gao T, Zhao R, Chen L. Polysaccharides from Lactarius volemus Fr. ameliorate high-fat and high-fructose diet induced metabolic disorders and intestinal barrier dysfunction. Int J Biol Macromol 2025; 287:138341. [PMID: 39638176 DOI: 10.1016/j.ijbiomac.2024.138341] [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: 11/15/2023] [Revised: 11/06/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Our research was conducted to investigate the effects of Lactarius volemus Fr. polysaccharides (LVP) on metabolic disorders and intestinal barrier dysfunction in HFFD-induced obese mice. Our findings demonstrated that LVP supplementation significantly ameliorated hyperlipoidemia and hyperglycemia, insulin resistance and hepatic inflammation. Additionally, LVP alleviated hepatic steatosis and histological lesions, as well as hepatic function dysbiosis. The underlying mechanism may involve the regulation of hepatic insulin signaling transduction pathway such as IRS1/AKT pathway and the suppression of MAPKs signaling pathway. Furthermore, LVP intervention improved intestinal barrier function and reduced intestinal permeability by enhancing the expression of tight junction proteins and restoring intestinal microbiota composition. In summary, our results provided evidence that LVP exerted beneficial effects on HFFD-induced metabolic disorders along with restoration of intestinal barrier function and reduction in endotoxin levels. These outcomes are associated with maintenance of gut microbiota homeostasis and up-regulation of Short-Chain Fatty Acids (SCFAs). Furthermore, butyric acid was found to restrict lipid accumulation in OA-induced HepG2 hepatocytes while strengthening intestinal barrier integrity in LPS-induced Caco-2 cells. Thus, polysaccharides LVP may serve as a potential prebiotic or health supplement in the prevention and treatment of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Hui Xu
- Bengbu First People's Hospital, Bengbu 233000, China
| | - Tianyu Zhang
- Bengbu First People's Hospital, Bengbu 233000, China; Graduate School, Bengbu Medical University, Bengbu 233030, China
| | - Ziming Zhou
- Bengbu First People's Hospital, Bengbu 233000, China; Graduate School, Bengbu Medical University, Bengbu 233030, China
| | - Tian Gao
- Bengbu First People's Hospital, Bengbu 233000, China; Graduate School, Bengbu Medical University, Bengbu 233030, China
| | - Ranran Zhao
- Bengbu First People's Hospital, Bengbu 233000, China; Graduate School, Bengbu Medical University, Bengbu 233030, China
| | - Lei Chen
- Anhui Key Laboratory of Ecological Engineering and Biotechnology, School of Life Sciences, Anhui University, Hefei 230601, China.
| |
Collapse
|
6
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
7
|
Gao JW, Liu YD, Jin MX. Intestinal epithelial glycocalyx and intestinal disease. Shijie Huaren Xiaohua Zazhi 2024; 32:887-896. [DOI: 10.11569/wcjd.v32.i12.887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/08/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
With the continuous research on glycobiology, more and more diseases are found to be associated with the glycocalyx. Glycocalyx can be categorized as endothelial glycocalyx and epithelial glycocalyx. Past studies mostly target endothelial glycocalyx, and this review focuses on the structure and function of intestinal epithelial glycocalyx, its degradation mechanism and biological relevance to different diseases of the intestinal tract, as well as the targeted delivery of drugs to organs by nanoparticle libraries mimicking the glycocalyx, in order to provide a theoretical basis for the study of potential diagnostic markers and therapeutic targets of intestinal epithelial glycocalyx in intestinal diseases.
Collapse
Affiliation(s)
- Jian-Wei Gao
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yan-Di Liu
- Department of Gastroenterology, Tianjin People's Hospital, Tianjin 300071, China
| | - Ming-Xing Jin
- Department of Gastroenterology, Tianjin People's Hospital, Tianjin 300071, China
| |
Collapse
|
8
|
Zhong Y, Chang X, Zhao Z, Zheng L, Kuang G, Li P, Liu C, Fan Y, Liang Z, Zhuang K, Xie Q, Liu Y. Bacteroides fragilis capsular polysaccharide A ameliorates ulcerative colitis in rat by recovering intestinal barrier integrity and restoring gut microbiota. Front Pharmacol 2024; 15:1402465. [PMID: 39776580 PMCID: PMC11703662 DOI: 10.3389/fphar.2024.1402465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/02/2024] [Indexed: 01/11/2025] Open
Abstract
Bacteroides fragilis (B. fragilis) is a Gram-negative, obligate anaerobic, commensal bacterium residing in the human gut and holds therapeutic potential for ulcerative colitis (UC). Previous studies have indicated that capsular polysaccharide A (PSA) of B. fragilis is a crucial component for its effectiveness, possessing various biological activities such as anti-inflammatory, anti-tumor, and immune-modulating effects. We previously isolated and characterized the B. fragilis strain ZY-312 from the feces of a healthy breastfed infant, and extracted its PSA, named TP2. In this study, we explored the impact of TP2 on colonic inflammation and delved into its potential mechanisms. Initially, we used 2,4,6-trinitrobenzenesulfonic acid (TNBS) to induce colitis in rats and found that TP2 treatment significantly ameliorated TNBS-induced weight loss, increased clinical scores, extensive ulcers, and intestinal epithelial damage in UC rats. Further analysis revealed that TP2 effectively restored the intestinal barrier integrity in UC rats by regulating the expression of Muc-2, tight junction proteins (ZO-1, occludin, claudin-1, and claudin-2), as well as apoptosis-related proteins Bcl-2, BAX, and Cleaved-Caspase-3. Additionally, TP2 suppressed the expression of pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and IL23, while promoting the secretion of anti-inflammatory cytokines IL-10 and IL-22, thereby inhibiting the occurrence of inflammation. TP2 also downregulated the phosphorylation levels of AKT and PI3K, effectively inhibiting the abnormal activation of the PI3K/AKT signaling pathway. More interestingly, 16S rRNA sequencing results showed that TP2 restored the ecological imbalance of the rat intestinal microbiota, with an increase in beneficial bacteria such as Lactobacillus and Limosilactobacillus observed in the treatment group. In conclusion, TP2 through the regulation of intestinal barrier-related cells and proteins, inhibition of apoptosis, modulation of inflammation-related cytokine levels, and control of abnormal activation of the PI3K/AKT signaling pathway, restores intestinal barrier integrity. Additionally, by reshaping the ecological imbalance of the gut microbiota, TP2 ultimately alleviates ulcerative colitis in rats.
Collapse
Affiliation(s)
- Yijia Zhong
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiujuan Chang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Zihan Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Lijun Zheng
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Gaobo Kuang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Ping Li
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | | | - Yuqin Fan
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhixuan Liang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ke Zhuang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiuling Xie
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| |
Collapse
|
9
|
Li L, Lei B, Zhang W, Wang W, Shang C, Hu Y, Zhao K, Yuan W. The disturbance of intestinal microbiome caused by the novel duck reovirus infection in Cherry Valley ducklings can induce intestinal damage. Poult Sci 2024; 103:104428. [PMID: 39490133 PMCID: PMC11550084 DOI: 10.1016/j.psj.2024.104428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/17/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024] Open
Abstract
Novel duck reovirus disease is an infectious disease mainly caused by novel duck reovirus (NDRV), which is characterized by spleen necrosis and persistent diarrhea in ducks. However, the pathogenic mechanism of NDRV infection in Cherry Valley ducks remains unclear. To investigate the distribution of NDRV in the intestines of Cherry Valley ducks, intestinal morphogenesis, intestinal permeability, inflammatory cytokines, and the expression of tight junction proteins (TJPs), we introduced NDRV via intramuscular infection. The diversity and composition of ileum flora and content of short-chain fatty acids (SCFAs) were analyzed using Illumina MiSeq sequencing. The relationship between changes in the intestinal microbial community and intestinal damage in Cherry Valley ducks infected with NDRV was also assessed to offer new insights into the pathogenesis of NDRV and intestinal flora composition. The results showed that intestinal inflammation and barrier dysfunction occurred following NDRV infection. Additionally, a significant reduction in dominant bacterial species and a decrease in SCFA content within the intestinal microbiota led to weakened colonization resistance and the enrichment of opportunistic pathogens, exacerbating intestinal damage post-NDRV infection. Notably, TJPs and inflammatory cytokine disruptions were linked to a decline in SCFA-producing bacteria and an accumulation of pathogenic bacteria. In summary, changes in the ileum intestinal flora and disruptions to the intestinal barrier were associated with NDRV infection. Consequently, disturbances in intestinal flora caused by NDRV infection can lead to intestinal damage. These findings may offer us a new perspective, targeting the gut microbiota to better understand the progression of NDRV disease and investigate its underlying pathogenesis.
Collapse
Affiliation(s)
- Lijie Li
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China
| | - Baishi Lei
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China
| | - Weizhu Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China
| | | | - Yibin Hu
- Beijing Centrebio Biological Co., Ltd, Beijing 102629, China
| | - Kuan Zhao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China
| | - Wanzhe Yuan
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, NO.2596 Lekai South Street, Hebei 071000, China; Veterinary Biological Technology Innovation Centre of Hebei Province, Baoding, Hebei 071000, China.
| |
Collapse
|
10
|
Jung N, Schreiner J, Baur F, Vogel-Kindgen S, Windbergs M. Predicting nanocarrier permeation across the human intestine in vitro: model matters. Biomater Sci 2024; 12:5775-5788. [PMID: 39402906 DOI: 10.1039/d4bm01092b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
For clinical translation of oral nanocarriers, simulation of the intestinal microenvironment during in vitro testing is crucial to evaluate interactions with the intestinal mucosa. However, studies are often conducted using simplistic cell culture models, overlooking key physiological factors, and potentially leading to an overestimation of nanocarrier permeation. In this study, we systematically investigate different tissue models of the human intestine under static cultivation and dynamic flow conditions and analyze the impact of altered tissue characteristics on nanocarrier permeation. Our results reveal that the selection of cell types as well as the respective culture condition have a notable impact on the physiological characteristics of the resulting tissues. Tissue layer thickness, mucus secretion, and barrier impairment, all increase with increasing amounts of goblet cells and the application of dynamic flow conditions. Permeation studies with poly(lactic-co-glycolic acid) (PLGA) nanocarriers with and without polyethylene glycol (PEG) coating elucidate that the amount of mucus present in the respective model is the limiting factor for the permeation of PLGA nanocarriers, while tissue topography presents the key factor influencing PEG-PLGA nanocarrier permeation. Furthermore, both nanocarriers exhibit diametrically opposite permeation kinetics compared to soluble compounds. In summary, these findings reveal the critical role of the implemented test systems on permeation assessment and emphasize that, in the context of preclinical nanocarrier testing, the choice of in vitro model matters.
Collapse
Affiliation(s)
- Nathalie Jung
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Jonas Schreiner
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Florentin Baur
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Peng Z, Wang D, He Y, Wei Z, Xie M, Xiong T. Gut Distribution, Impact Factor, and Action Mechanism of Bacteriocin-Producing Beneficial Microbes as Promising Antimicrobial Agents in Gastrointestinal Infection. Probiotics Antimicrob Proteins 2024; 16:1516-1527. [PMID: 38319538 DOI: 10.1007/s12602-024-10222-6] [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] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Gastrointestinal (GI) infection by intestinal pathogens poses great threats to human health, and the therapeutic use of antibiotics has reached a bottleneck due to drug resistance. The developments of antimicrobial peptides produced by beneficial bacteria have drawn attention by virtue of effective, safe, and not prone to developing resistance. Though bacteriocin as antimicrobial agent in gut infection has been intensively investigated and reviewed, reviews on that of bacteriocin-producing beneficial microbes are very rare. It is important to explicitly state the prospect of bacteriocin-producing microbes in prevention of gastrointestinal infection towards their application in host. This review discusses the potential of gut as an appropriate resource for mining targeted bacteriocin-producing microbes. Then, host-related factors affecting the bacteriocin production and activity of bacteriocin-producing microbes in the gut are summarized. Accordingly, the multiple mechanisms (direct inhibition and indirect inhibition) behind the preventive effects of bacteriocin-producing microbes on gut infection are discussed. Finally, we propose several targeted strategies for the manipulation of bacteriocin-producing beneficial microbes to improve their performance in antimicrobial outcomes. We anticipate an upcoming emergence of developments and applications of bacteriocin-producing beneficial microbes as antimicrobial agent in gut infection induced by pathogenic bacteria.
Collapse
Affiliation(s)
- Zhen Peng
- School of Food Science and Technology, Nanchang University, Nanchang, China
- International Institute of Food Innovation Co., Ltd., Nanchang University, Nanchang, Jiangxi, China
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Donglin Wang
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Yuyan He
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ziqi Wei
- School of Food Science and Technology, Nanchang University, Nanchang, China
| | - Mingyong Xie
- School of Food Science and Technology, Nanchang University, Nanchang, China
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Tao Xiong
- School of Food Science and Technology, Nanchang University, Nanchang, China.
- International Institute of Food Innovation Co., Ltd., Nanchang University, Nanchang, Jiangxi, China.
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China.
| |
Collapse
|
12
|
Wang J, Gao J, Sheng X, Tang X, Xing J, Chi H, Zhan W. Teleost Muc2 and Muc5ac: Key guardians of mucosal immunity in flounder (Paralichthys olivaceus). Int J Biol Macromol 2024; 277:134127. [PMID: 39053833 DOI: 10.1016/j.ijbiomac.2024.134127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Mucins secreted by mucous cells constitute a core part of the defense line against the invasion of pathogens. However, mucins' structure and immunological functions remain largely unknown in teleost fish. In this study, two typical mucins, Muc2 and Muc5ac of flounder (Paralichthys olivaceus), were cloned and their physicochemical properties, structure and conservation were analyzed. Notably, specific antibodies against flounder Muc2 and Muc5ac were developed. It was verified at the gene and protein level that Muc2 was expressed in the hindgut and gills but not in the skin, while Muc5ac was expressed in the skin and gills but not in the hindgut. After flounders were immunized by immersion with inactivated Edwardsiella tarda, Muc2 and Muc5ac were significantly upregulated at both the gene expression and protein levels, and Muc2+/Muc5ac+ mucous cells proliferated and increased secretion of Muc2 and Muc5ac. Moreover, Muc2 and Muc5ac exerted retention and clearance effects on E. tarda in a short period (within 1 dpi). These results revealed the characterization of fish mucins Muc2 and Muc5ac at the protein level and clarified the role of mucins as key guardians to maintain the mucus barrier, which advanced our understanding of teleost mucosal barrier.
Collapse
Affiliation(s)
- Jincheng Wang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China
| | - Jianliang Gao
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China.
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong 266237, PR China
| |
Collapse
|
13
|
Macura B, Kiecka A, Szczepanik M. Intestinal permeability disturbances: causes, diseases and therapy. Clin Exp Med 2024; 24:232. [PMID: 39340718 PMCID: PMC11438725 DOI: 10.1007/s10238-024-01496-9] [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: 08/20/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024]
Abstract
Nowadays, a pathological increase in the permeability of the intestinal barrier (the so-called leaky gut) is increasingly being diagnosed. This condition can be caused by various factors, mainly from the external environment. Damage to the intestinal barrier entails a number of adverse phenomena: dysbiosis, translocation of microorganisms deep into the intestinal tissue, immune response, development of chronic inflammation. These phenomena can ultimately lead to a vicious cycle that promotes the development of inflammation and further damage to the barrier. Activated immune cells in mucosal tissues with broken barriers can migrate to other organs and negatively affect their functioning. Damaged intestinal barrier can facilitate the development of local diseases such as irritable bowel disease, inflammatory bowel disease or celiac disease, but also the development of systemic inflammatory diseases such as rheumatoid arthritis, ankylosing spondylitis, hepatitis, and lupus erythematosus, neurodegenerative or psychiatric conditions, or metabolic diseases such as diabetes or obesity. However, it must be emphasized that the causal links between a leaky gut barrier and the onset of certain diseases often remain unclear and require in-depth research. In light of recent research, it becomes crucial to prevent damage to the intestinal barrier, as well as to develop therapies for the barrier when it is damaged. This paper presents the current state of knowledge on the causes, health consequences and attempts to treat excessive permeability of the intestinal barrier.
Collapse
Affiliation(s)
- Barbara Macura
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland.
| | - Aneta Kiecka
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| | - Marian Szczepanik
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| |
Collapse
|
14
|
Niu MM, Li Y, Su Q, Chen SY, Li QH, Guo HX, Meng XC, Liu F. A mannose-rich exopolysaccharide-1 isolated from Bifidobacterium breve mitigates ovalbumin-induced intestinal damage in mice by modulation CD4 + T cell differentiation and inhibiting NF-κB signaling pathway. Int J Biol Macromol 2024; 280:135850. [PMID: 39326613 DOI: 10.1016/j.ijbiomac.2024.135850] [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: 05/22/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
Ovalbumin (OVA)-induced intestinal injury is a recurrent and potentially fatal condition. Previous studies have highlighted the roles of exopolysaccharides, particularly a mannose-rich (89.59 %) exopolysaccharide-1 (EPS-1) with a molecular weight of 39.9 kDa, isolated from Bifidobacterium breve H4-2, in repairing intestinal barriers and regulating immune responses. In this study, a mouse model of OVA-induced intestinal injury was used to investigate the effects of EPS-1 on intestinal barrier restoration. The results demonstrated that EPS-1 treatment (400 mg/kg. d) significantly reduced the allergic index (3.25 ± 0.43) in OVA-challenged mice (p < 0.05), improved the physical integrity of the intestinal barrier by increasing mucin content and goblet cell number in the ileum (p < 0.05). EPS-1 treatment (400 mg/kg. d) also maintained immune barrier integrity by restoring imbalanced CD4 + T/CD8 + T ratios from 0.86 ± 0.02 to 1.04 ± 0.06, regulating Th1/Th2 and Th17/Treg cells balance, as well as inhibited the NF-κB signaling pathway. Furthermore, EPS-1 maintained microbiota homeostasis by increasing the abundances of Ruminococcus, Butyricicoccus, and Muribaculaceae, while reducing Streptococcus and Candidatus arthromitus. This microbiota modulation enhanced the levels of metabolites such as tyrosine, methionine, tryptophan, triglycerides, and salidroside. In conclusion, EPS-1 shows promise as a functional polysaccharide for therapeutic use.
Collapse
Affiliation(s)
- Meng-Meng Niu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Yan Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Qian Su
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Si-Yuan Chen
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Qiao-Hui Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Huan-Xin Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiang-Chen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; Food College, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
15
|
Kramer C, Rulff H, Ziegler JF, Mönch PW, Alzain N, Addante A, Kuppe A, Timm S, Schrade P, Bischoff P, Glauben R, Dürr J, Ochs M, Mall MA, Gradzielski M, Siegmund B. Ileal mucus viscoelastic properties differ in Crohn's disease. Mucosal Immunol 2024; 17:713-722. [PMID: 38750968 DOI: 10.1016/j.mucimm.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/26/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024]
Abstract
Crohn's disease (CD) is an inflammatory bowel disease that can affect any part of the gastrointestinal tract, frequently involving the terminal ileum. While colonic mucus alterations in CD patients have been described, terminal ileal mucus and its mechanobiological properties have been neglected. Our study is the first of its kind to decipher the viscoelastic and network properties of ileal mucus. With that aim, oscillatory rheological shear measurements based on an airway mucus protocol that was thoroughly validated for ileal mucus were performed. Our pilot study analyzed terminal ileum mucus from controls (n = 14) and CD patients (n = 14). Mucus network structure was visualized by scanning electron microscopy. Interestingly, a statistically significant increase in viscoelasticity as well as a decrease in mesh size was observed in ileal mucus from CD patients compared to controls. Furthermore, rheological data were analyzed in relation to study participants' clinical characteristics, revealing a noteworthy trend between non-smokers and smokers. In conclusion, this study provides the first data on the viscoelastic properties and structure of human ileal mucus in the healthy state and Crohn's disease, demonstrating significant alterations between groups and highlighting the need for further research on mucus and its effect on the underlying epithelial barrier.
Collapse
Affiliation(s)
- Catharina Kramer
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hanna Rulff
- Institute of Chemistry, Technische Universität Berlin, Berlin, Germany
| | - Jörn Felix Ziegler
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Paul Wilhelm Mönch
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nadra Alzain
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Annalisa Addante
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Aditi Kuppe
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Sara Timm
- Core Facility Electron Microscopy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Petra Schrade
- Core Facility Electron Microscopy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philip Bischoff
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany; Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rainer Glauben
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Dürr
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Matthias Ochs
- Core Facility Electron Microscopy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute of Functional Anatomy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcus A Mall
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | | | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Nguyen Duy T, Le Huy H, Đao Thanh Q, Ngo Thi H, Ngo Thi Minh H, Nguyen Dang M, Le Huu S, Ngo Tat T. Association between Bacteroides fragilis and Fusobacterium nucleatum infection and colorectal cancer in Vietnamese patients. Anaerobe 2024; 88:102880. [PMID: 38942229 DOI: 10.1016/j.anaerobe.2024.102880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/24/2024] [Accepted: 06/20/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a significant global health concern, and understanding the role of specific bacterial infections in its development and progression is of increasing interest. This cross-sectional study investigated the associations between Bacteroides fragilis (B. fragilis) and Fusobacterium nucleatum (F. nucleatum) infections and Vietnamese CRC patients. METHODS 192 patients with either polyps or CRC at varying stages were recruited from May 2017 to December 2020. Real-time PCR assessed infection rates and bacterial loads in CRC tissues. RESULTS B. fragilis infection was notably higher in CRC tissues (51.6 %) than polyps (9.4 %), with a fivefold higher relative load. Positive associations were found in stages II and III, indicating a fivefold increase in CRC progression risk. F. nucleatum infection rates were significantly higher in CRC tissues (55.2 %) than in polyps (10.5 %). In stage II, the infection rate exceeded that in adjacent tissues. The relative load of F. nucleatum was higher in stage III than in stages I and II. Positive F. nucleatum patients had a 3.2 times higher risk of CRC progression. CONCLUSION These findings suggest associations between loading of F. nucleatum or/and B. fragilis with the advanced stages of CRC.
Collapse
Affiliation(s)
- Truong Nguyen Duy
- Institute of Clinical Infectious Diseases, 108 Military Central Hospital, Hanoi, 10000, Viet Nam
| | - Hoang Le Huy
- Department of Bacteriology, National of Hygiene and Epidemiology, Hanoi, 10000, Viet Nam
| | - Quyen Đao Thanh
- Vietnamese-German Center of Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, 100000, Viet Nam
| | - Hoai Ngo Thi
- Department of Gastroenterological Intensive Care, 108 Military Central Hospital, Hanoi, 10000, Viet Nam
| | - Hanh Ngo Thi Minh
- Department of Pathology, 108 Military Central Hospital, Hanoi, 10000, Viet Nam
| | - Manh Nguyen Dang
- Institute of Clinical Infectious Diseases, 108 Military Central Hospital, Hanoi, 10000, Viet Nam
| | - Song Le Huu
- Institute of Clinical Infectious Diseases, 108 Military Central Hospital, Hanoi, 10000, Viet Nam; Vietnamese-German Center of Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, 100000, Viet Nam.
| | - Trung Ngo Tat
- Vietnamese-German Center of Medical Research (VG-CARE), 108 Military Central Hospital, Hanoi, 100000, Viet Nam; Centre for Genetics Consultation and Cancer Screening, 108 Military Central Hospital, Hanoi, 100000, Viet Nam.
| |
Collapse
|
17
|
Yan Z, Yeo J. Competing mechanisms in bacterial invasion of human colon mucus probed with agent-based modeling. Biophys J 2024; 123:1838-1845. [PMID: 38824388 PMCID: PMC11630638 DOI: 10.1016/j.bpj.2024.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/03/2024] Open
Abstract
The gastrointestinal tract is inhabited by a vast community of microorganisms, termed the gut microbiota. Large colonies can pose a health threat, but the gastrointestinal mucus system protects epithelial cells from microbiota invasion. The human colon features a bilayer of mucus lining. Due to imbalances in intestinal homeostasis, bacteria may successfully penetrate the inner mucus layer, which can lead to severe gut diseases. However, it is hard to tease apart the competing mechanisms that lead to this penetration. To probe the conditions that permit bacteria penetration into the inner mucus layer, we develop an agent-based model consisting of bacteria and an inner mucus layer subject to a constant flux of nutrient fields feeding the bacteria. We find that there are three important variables that determine bacterial invasion: the bacterial reproduction rate, the contact energy between bacteria and mucus, and the rate of bacteria degrading the mucus. Under healthy conditions, all bacteria are naturally eliminated by the constant removal of mucus. In diseased states, imbalances between the rates of bacterial degradation and mucus secretion lead to bacterial invasion at certain junctures. We conduct uncertainty quantification and sensitivity analysis to compare the relative impact between these parameters. The contact energy has the strongest influence on bacterial penetration, which, in combination with bacterial degradation rate and growth rate, greatly accelerates bacterial invasion of the human gut mucus lining. Our findings will serve as predictive indicators for the etiology of intestinal diseases and highlight important considerations when developing gut therapeutics.
Collapse
Affiliation(s)
- Zhongyu Yan
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York
| | - Jingjie Yeo
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
18
|
Pan I, Umapathy S. Probiotics an emerging therapeutic approach towards gut-brain-axis oriented chronic health issues induced by microplastics: A comprehensive review. Heliyon 2024; 10:e32004. [PMID: 38882279 PMCID: PMC11176854 DOI: 10.1016/j.heliyon.2024.e32004] [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: 12/07/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024] Open
Abstract
Applications for plastic polymers can be found all around the world, often discarded without any prior care, exacerbating the environmental issue. When large waste materials are released into the environment, they undergo physical, biological, and photo-degradation processes that break them down into smaller polymer fragments known as microplastics (MPs). The time it takes for residual plastic to degrade depends on the type of polymer and environmental factors, with some taking as long as 600 years or more. Due to their small size, microplastics can contaminate food and enter the human body through food chains and webs, causing gastrointestinal (GI) tract pain that can range from local to systemic. Microplastics can also acquire hydrophobic organic pollutants and heavy metals on their surface, due to their large surface area and surface hydrophobicity. The levels of contamination on the microplastic surface are significantly higher than in the natural environment. The gut-brain axis (GB axis), through which organisms interact with their environment, regulate nutritional digestion and absorption, intestinal motility and secretion, complex polysaccharide breakdown, and maintain intestinal integrity, can be altered by microplastics acting alone or in combination with pollutants. Probiotics have shown significant therapeutic potential in managing various illnesses mediated by the gut-brain axis. They connect hormonal and biochemical pathways to promote gut and brain health, making them a promising therapy option for a variety of GB axis-mediated illnesses. Additionally, taking probiotics with or without food can reduce the production of pro-inflammatory cytokines, reactive oxygen species (ROS), neuro-inflammation, neurodegeneration, protein folding, and both motor and non-motor symptoms in individuals with Parkinson's disease. This study provides new insight into microplastic-induced gut dysbiosis, its associated health risks, and the benefits of using both traditional and next-generation probiotics to maintain gut homeostasis.
Collapse
Affiliation(s)
- Ieshita Pan
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| | - Suganiya Umapathy
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, 602105, Tamil Nadu, India
| |
Collapse
|
19
|
Ermolenko E, Baryshnikova N, Alekhina G, Zakharenko A, Ten O, Kashchenko V, Novikova N, Gushchina O, Ovchinnikov T, Morozova A, Ilina A, Karaseva A, Tsapieva A, Gladyshev N, Dmitriev A, Suvorov A. Autoprobiotics in the Treatment of Patients with Colorectal Cancer in the Early Postoperative Period. Microorganisms 2024; 12:980. [PMID: 38792809 PMCID: PMC11124500 DOI: 10.3390/microorganisms12050980] [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: 02/23/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Despite great advances in the treatment of oncological diseases, the development of medical technologies to prevent or reduce complications of therapy, in particular, those associated with surgery and the introduction of antibiotics, remains relevant. The aim of this study is to evaluate the effectiveness of the use of autoprobiotics based on indigenous non-pathogenic strains of Enterococcus faecium and Enterococcus hirae as a personalized functional food product (PFFP) in the complex therapy of colorectal cancer (CRC) in the early postoperative period. A total of 36 patients diagnosed with CRC were enrolled in the study. Study group A comprised 24 CRC patients who received autoprobiotic therapy in the early postoperative period, while the control group C included 12 CRC patients without autoprobiotic therapy. Prior to surgery and between days 14 and 16 post-surgery, comprehensive evaluations were conducted on all patients, encompassing the following: stool and gastroenterological complaints analysis, examination of the gut microbiota (bacteriological study, quantitative polymerase chain reaction, metagenome analysis), and analysis of interleukins in the serum. Results: The use of autoprobiotics led to a decrease in dyspeptic complaints after surgery. It was also associated with the absence of postoperative complications, did not cause any side effects, and led to a decrease in the level of pro-inflammatory cytokines (IL-6 and IL-18) in the blood serum. The use of autoprobiotics led to positive changes in the structure of escherichia and enterococci populations, the elimination of Parvomonas micra and Fusobacterium nucleatum, and a decrease in the quantitative content of Clostridium perfringens and Akkermansia muciniphila. Metagenomic analysis (16S rRNA) revealed an increase in alpha diversity. Conclusion: The introduction of autoprobiotics in the postoperative period is a highly effective and safe approach in the complex treatment of CRC. Future studies will allow the discovery of additional fine mechanisms of autoprobiotic therapy and its impact on the digestive, immune, endocrine, and neural systems.
Collapse
Affiliation(s)
- Elena Ermolenko
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Natalia Baryshnikova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Department of Internal Disease of Stomatology Faculty, Pavlov First St-Petersburg State Medical University, 197022 St-Petersburg, Russia
- Laboratory of Medico-Social Problems of Pediatry, St-Petersburg State Pediatric Medical University, 194100 St-Petersburg, Russia
| | - Galina Alekhina
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alexander Zakharenko
- Oncology Department, Pavlov First St-Petersburg State Medical University, 197022 St-Petersburg, Russia;
| | - Oleg Ten
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Victor Kashchenko
- Department of Faculty Surgery, St-Petersburg State University, 199034 St-Petersburg, Russia;
- Beloostrov High Technology Clinic (MMC VT LLC), 188652 Leningrad Region, Russia
| | - Nadezhda Novikova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Olga Gushchina
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Timofey Ovchinnikov
- North-Western District Scientific and Clinical Center Named after L. G. Sokolov, 194291 St-Petersburg, Russia (O.G.)
| | - Anastasia Morozova
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Anastasia Ilina
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alena Karaseva
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Microbiology Department, St-Petersburg State University, 199034 St-Petersburg, Russia
| | - Anna Tsapieva
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Nikita Gladyshev
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| | - Alexander Dmitriev
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
- Department of Molecular Biotechnology, Saint-Petersburg State Institute of Technology, 190013 St-Petersburg, Russia
| | - Alexander Suvorov
- Scientific and Educational Center “Molecular Bases of Interaction of Microorganisms and Human”, World-Class Research Center “Center for Personalized Medicine”, Institute of Experimental Medicine, 197376 St-Petersburg, Russia; (E.E.); (A.M.); (A.T.); (N.G.)
| |
Collapse
|
20
|
Zhang L, Zhou E, Liu C, Tian X, Xue B, Zhang K, Luo B. Avian influenza and gut microbiome in poultry and humans: A "One Health" perspective. FUNDAMENTAL RESEARCH 2024; 4:455-462. [PMID: 38933214 PMCID: PMC11197557 DOI: 10.1016/j.fmre.2023.10.016] [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: 12/29/2022] [Revised: 09/20/2023] [Accepted: 10/10/2023] [Indexed: 06/28/2024] Open
Abstract
A gradual increase in avian influenza outbreaks has been found in recent years. It is highly possible to trigger the next human pandemic due to the characteristics of antigenic drift and antigenic shift in avian influenza virus (AIV). Although great improvements in understanding influenza viruses and the associated diseases have been unraveled, our knowledge of how these viruses impact the gut microbiome of both poultry and humans, as well as the underlying mechanisms, is still improving. The "One Health" approach shows better vitality in monitoring and mitigating the risk of avian influenza, which requires a multi-sectoral effort and highlights the interconnection of human health with environmental sustainability and animal health. Therefore, monitoring the gut microbiome may serve as a sentinel for protecting the common health of the environment, animals, and humans. This review summarizes the interactions between AIV infection and the gut microbiome of poultry and humans and their potential mechanisms. With the presented suggestions, we hope to address the current major challenges in the surveillance and prevention of microbiome-related avian influenza with the "One Health" approach.
Collapse
Affiliation(s)
- Ling Zhang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Erkai Zhou
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ce Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xiaoyu Tian
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Baode Xue
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, NY 12144, USA
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, China
- Shanghai Key Laboratory of Meteorology and Health, Shanghai Meteorological Bureau, Shanghai 200030, China
- Shanghai Typhoon Institute, China Meteorological Administration, Shanghai 200030, China
| |
Collapse
|
21
|
Ding S, Li K, Han X, Lin W, Qin Y, Cao R, Ren Y. Long-term use of etomidate disrupts the intestinal homeostasis and nervous system in mice. Toxicology 2024; 504:153802. [PMID: 38604439 DOI: 10.1016/j.tox.2024.153802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024]
Abstract
Etomidate (ETO) is used as an anesthetic in surgery, but it is being abused in some populations. The damage caused by long-term intake of ETO to intestinal and brain functions is not yet clear, and it remains to be determined whether the drug affects the central nervous system through the gut-brain axis. This study aimed to investigate the neurotoxic and gastrointestinal effects of ETO at doses of 1 mg/kg and 3 mg/kg in mice over 14 consecutive days. The results showed that long-term injection of ETO led to drug resistance in mice, affecting their innate preference for darkness and possibly inducing dependence on ETO. The levels of 5-hydroxytryptamine in the brain, serum, and colon decreased by 37%, 51%, and 42% respectively, while the levels of γ-aminobutyric acid reduced by 38%, 52%, and 41% respectively. H&E staining revealed that ETO reduced goblet cells in the colon and damaged the intestinal barrier. The expression of tight junction-related genes Claudin4 and ZO-1 was downregulated. The intestinal flora changed, the abundance of Akkermansia and Lactobacillus decreased by 33% and 14%, respectively, while Klebsiella increased by 18%. TUNEL results showed that high-dose ETO increased apoptotic cells in the brain. The expression of Claudin1 in the brain was downregulated. Untargeted metabolomics analysis of the colon and brain indicated that ETO caused abnormalities in glycerophospholipid metabolism. Abnormal lipid metabolism might lead to the production or accumulation of lipotoxic metabolites, causing central nervous system diseases. ETO induced changes in the intestinal flora and metabolism, further affecting the central nervous system through the gut-brain axis. The study unveiled the detrimental effects on the brain and gastrointestinal system resulting from long-term intake of ETO, which holds significant implications for comprehending the adverse impact of ETO abuse on human health.
Collapse
Affiliation(s)
- Siming Ding
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Kan Li
- National Anti-Drug Laboratory Guangdong Regional Center, Guangzhou 510230, PR China; Anti-Drug Technology Center of Guangdong Province, Guangzhou 510230, PR China
| | - Xing Han
- National Anti-Drug Laboratory Guangdong Regional Center, Guangzhou 510230, PR China; Anti-Drug Technology Center of Guangdong Province, Guangzhou 510230, PR China
| | - Wenting Lin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Yingjun Qin
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Renjuan Cao
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China
| | - Yuan Ren
- School of Environment and Energy, South China University of Technology, Guangzhou 510006, PR China; The Key Lab of Pollution Control and Ecosystem Restoration in Industry Clusters, Ministry of Education, Guangzhou 510006, PR China; The Key Laboratory of Environmental Protection and Eco-Remediation of Guangdong Regular Higher Education Institutions, Guangzhou 510006, PR China.
| |
Collapse
|
22
|
Missong H, Joshi R, Khullar N, Thareja S, Navik U, Bhatti GK, Bhatti JS. Nutrient-epigenome interactions: Implications for personalized nutrition against aging-associated diseases. J Nutr Biochem 2024; 127:109592. [PMID: 38325612 DOI: 10.1016/j.jnutbio.2024.109592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Aging is a multifaceted process involving genetic and environmental interactions often resulting in epigenetic changes, potentially leading to aging-related diseases. Various strategies, like dietary interventions and calorie restrictions, have been employed to modify these epigenetic landscapes. A burgeoning field of interest focuses on the role of microbiota in human health, emphasizing system biology and computational approaches. These methods help decipher the intricate interplay between diet and gut microbiota, facilitating the creation of personalized nutrition strategies. In this review, we analysed the mechanisms related to nutritional interventions while highlighting the influence of dietary strategies, like calorie restriction and intermittent fasting, on microbial composition and function. We explore how gut microbiota affects the efficacy of interventions using tools like multi-omics data integration, network analysis, and machine learning. These tools enable us to pinpoint critical regulatory elements and generate individualized models for dietary responses. Lastly, we emphasize the need for a deeper comprehension of nutrient-epigenome interactions and the potential of personalized nutrition informed by individual genetic and epigenetic profiles. As knowledge and technology advance, dietary epigenetics stands on the cusp of reshaping our strategy against aging and related diseases.
Collapse
Affiliation(s)
- Hemi Missong
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Riya Joshi
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, Punjab, India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
23
|
Chen H, Yu Z, Qi Z, Huang X, Gao J. Tongfu Lifei Decoction Attenuated Sepsis-Related Intestinal Mucosal Injury Through Regulating Th17/Treg Balance and Modulating Gut Microbiota. J Interferon Cytokine Res 2024; 44:208-220. [PMID: 38691831 DOI: 10.1089/jir.2024.0001] [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] [Indexed: 05/03/2024] Open
Abstract
Intestinal damage and secondary bacterial translocation are caused by the inflammatory response induced by sepsis. Tongfu Lifei (TLF) decoction has a protective effect on sepsis-related gastrointestinal function injury. However, the relation between gut microbiota, immune barrier, and sepsis under the treatment of TLF have not been well clarified yet. Here, rats were subjected to cecal ligation and puncture (CLP) to create a sepsis model. Subsequently, the TLF decoction was given to CLP rats by gavage, fecal microbiota transplantation (FMT), and antibiotic were used as positive control. TLF suppressed the inflammatory response and improved the pathological changes in the intestines of CLP rats. Besides, TLF promoted the balance of the percentage of the Th17 and Treg cells. Intestinal barrier function was also improved by TLF through enhancing ZO-1, and Occludin and Claudin 1 expression, preventing the secondary translocation of other gut microbiota. TLF dramatically boosted the gut microbiota's alpha- and beta-diversity in CLP rats. Moreover, it increased the relative abundance of anti-inflammatory gut microbiota and changed the progress of the glucose metabolism. In short, TLF regulated the gut microbiota to balance the ratio of Th17/Treg cells, reducing the inflammation in serum and intestinal mucosal injury in rats.
Collapse
Affiliation(s)
- Huizhen Chen
- Department of Intensive Care Medicine, and Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, Hangzhou, China
| | - Zhenfei Yu
- Department of Intensive Care Medicine, and Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, Hangzhou, China
| | - Zeming Qi
- Department of Infectious Diseases, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, Hangzhou, China
| | - Xiaozhe Huang
- Department of Infectious Diseases, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, Hangzhou, China
| | - Jianting Gao
- Department of Intensive Care Medicine, and Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medicine University, Hangzhou, China
| |
Collapse
|
24
|
Windfelder AG, Steinbart J, Graser L, Scherberich J, Krombach GA, Vilcinskas A. An enteric ultrastructural surface atlas of the model insect Manducasexta. iScience 2024; 27:109410. [PMID: 38558941 PMCID: PMC10981077 DOI: 10.1016/j.isci.2024.109410] [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: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
The tobacco hornworm is a laboratory model that is particularly suitable for analyzing gut inflammation, but a physiological reference standard is currently unavailable. Here, we present a surface atlas of the healthy hornworm gut generated by scanning electron microscopy and nano-computed tomography. This comprehensive overview of the gut surface reveals morphological differences between the anterior, middle, and posterior midgut, allowing the screening of aberrant gut phenotypes while accommodating normal physiological variations. We estimated a total resorptive midgut surface of 0.42 m2 for L5d6 larvae, revealing its remarkable size. Our data will support allometric scaling and dose conversion from Manduca sexta to mammals in preclinical research, embracing the 3R principles. We also observed non-uniform gut colonization by enterococci, characterized by dense biofilms in the pyloric cone and downstream of the pylorus associated with pore and spine structures in the hindgut intima, indicating a putative immunosurveillance function in the lepidopteran hindgut.
Collapse
Affiliation(s)
- Anton G. Windfelder
- Branch for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- Experimental Radiology, Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Justus Liebig University Giessen, Giessen, Germany
| | - Jessica Steinbart
- Experimental Radiology, Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Justus Liebig University Giessen, Giessen, Germany
| | - Leonie Graser
- Branch for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
| | - Jan Scherberich
- Experimental Radiology, Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Justus Liebig University Giessen, Giessen, Germany
| | - Gabriele A. Krombach
- Experimental Radiology, Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Justus Liebig University Giessen, Giessen, Germany
- Department of Diagnostic and Interventional Radiology, University-Hospital Giessen, Giessen, Germany
| | - Andreas Vilcinskas
- Branch for Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Giessen, Germany
- Institute for Insect Biotechnology, Department of Applied Entomology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
25
|
Pereira QC, Fortunato IM, Oliveira FDS, Alvarez MC, dos Santos TW, Ribeiro ML. Polyphenolic Compounds: Orchestrating Intestinal Microbiota Harmony during Aging. Nutrients 2024; 16:1066. [PMID: 38613099 PMCID: PMC11013902 DOI: 10.3390/nu16071066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
In the aging process, physiological decline occurs, posing a substantial threat to the physical and mental well-being of the elderly and contributing to the onset of age-related diseases. While traditional perspectives considered the maintenance of life as influenced by a myriad of factors, including environmental, genetic, epigenetic, and lifestyle elements such as exercise and diet, the pivotal role of symbiotic microorganisms had been understated. Presently, it is acknowledged that the intestinal microbiota plays a profound role in overall health by signaling to both the central and peripheral nervous systems, as well as other distant organs. Disruption in this bidirectional communication between bacteria and the host results in dysbiosis, fostering the development of various diseases, including neurological disorders, cardiovascular diseases, and cancer. This review aims to delve into the intricate biological mechanisms underpinning dysbiosis associated with aging and the clinical ramifications of such dysregulation. Furthermore, we aspire to explore bioactive compounds endowed with functional properties capable of modulating and restoring balance in this aging-related dysbiotic process through epigenetics alterations.
Collapse
Affiliation(s)
- Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Fabricio de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| |
Collapse
|
26
|
van Gorp C, de Lange IH, Hütten MC, López-Iglesias C, Massy KRI, Kessels L, Knoops K, Cuijpers I, Sthijns MMJPE, Troost FJ, van Gemert WG, Spiller OB, Birchenough GMH, Zimmermann LJI, Wolfs TGAM. Antenatal Ureaplasma Infection Causes Colonic Mucus Barrier Defects: Implications for Intestinal Pathologies. Int J Mol Sci 2024; 25:4000. [PMID: 38612809 PMCID: PMC11011967 DOI: 10.3390/ijms25074000] [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: 02/29/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
Chorioamnionitis is a risk factor for necrotizing enterocolitis (NEC). Ureaplasma parvum (UP) is clinically the most isolated microorganism in chorioamnionitis, but its pathogenicity remains debated. Chorioamnionitis is associated with ileal barrier changes, but colonic barrier alterations, including those of the mucus barrier, remain under-investigated, despite their importance in NEC pathophysiology. Therefore, in this study, the hypothesis that antenatal UP exposure disturbs colonic mucus barrier integrity, thereby potentially contributing to NEC pathogenesis, was investigated. In an established ovine chorioamnionitis model, lambs were intra-amniotically exposed to UP or saline for 7 d from 122 to 129 d gestational age. Thereafter, colonic mucus layer thickness and functional integrity, underlying mechanisms, including endoplasmic reticulum (ER) stress and redox status, and cellular morphology by transmission electron microscopy were studied. The clinical significance of the experimental findings was verified by examining colon samples from NEC patients and controls. UP-exposed lambs have a thicker but dysfunctional colonic mucus layer in which bacteria-sized beads reach the intestinal epithelium, indicating undesired bacterial contact with the epithelium. This is paralleled by disturbed goblet cell MUC2 folding, pro-apoptotic ER stress and signs of mitochondrial dysfunction in the colonic epithelium. Importantly, the colonic epithelium from human NEC patients showed comparable mitochondrial aberrations, indicating that NEC-associated intestinal barrier injury already occurs during chorioamnionitis. This study underlines the pathogenic potential of UP during pregnancy; it demonstrates that antenatal UP infection leads to severe colonic mucus barrier deficits, providing a mechanistic link between antenatal infections and postnatal NEC development.
Collapse
Affiliation(s)
- Charlotte van Gorp
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Ilse H. de Lange
- Department of Pediatrics, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Matthias C. Hütten
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Neonatology, Department of Pediatrics, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Kimberly R. I. Massy
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Lilian Kessels
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Kèvin Knoops
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, 6211 LK Maastricht, The Netherlands; (C.L.-I.); (K.K.)
| | - Iris Cuijpers
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Mireille M. J. P. E. Sthijns
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Freddy J. Troost
- Food Innovation and Health, Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 5911 BV Venlo, The Netherlands; (I.C.); (M.M.J.P.E.S.); (F.J.T.)
| | - Wim G. van Gemert
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Owen B. Spiller
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff CF14 4XW, UK;
| | - George M. H. Birchenough
- Department of Medical Biochemistry, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Luc J. I. Zimmermann
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School for Oncology and Reproduction (GROW), Maastricht University, 6229 ER Maastricht, The Netherlands; (C.v.G.); (M.C.H.); (K.R.I.M.); (L.K.); (L.J.I.Z.)
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
27
|
Hajihashemi P, Haghighatdoost F, Kassaian N, Hoveida L, Tamizifar B, Nili H, Rahim Khorasani M, Adibi P. Bovine Colostrum in Increased Intestinal Permeability in Healthy Athletes and Patients: A Meta-Analysis of Randomized Clinical Trials. Dig Dis Sci 2024; 69:1345-1360. [PMID: 38361147 DOI: 10.1007/s10620-023-08219-2] [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: 06/29/2023] [Accepted: 11/27/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND Increasing intestinal permeability causes chronic inflammation, which is one of the etiological factors of many diseases that presently constitute global challenges. AIMS Considering the importance of developing therapies to eliminate the increased intestinal permeability, in this systematic review and meta-analysis, we analyze the impact of bovine colostrum (BC) on the gut barrier and its permeability. METHODS Online databases, including PubMed, ISI Web of Science, and Scopus, were searched to find pertinent articles up to March 2022. Weighted mean difference (WMD) and 95% confidence intervals (CI) were considered as effect sizes. The random-effects model was used to pool the study results. RESULTS A total of ten articles were included in the meta-analysis. The pooled effect revealed a significant reduction in the 5-h urinary lactulose/rhamnose ratio after BC consumption [mean difference (MD): -0.24; 95% CI -0.43 to -0.04; I2 = 99%] and urinary lactulose/mannitol ratio (MD: -0.01; 95% CI -0.02 to -0.001; I2 = 29.8%). No differences were observed in the plasma intestinal fatty acid-binding protein (I-FABP) between BC and control groups (MD: 2.30; 95% CI -293.9 to 298.5; I2 = 92%). CONCLUSIONS BC supplementation significantly reduced intestinal permeability; however, to confirm the results, more randomized clinical trials considering different quality, dose, and duration are needed.
Collapse
Affiliation(s)
- Parisa Hajihashemi
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fahimeh Haghighatdoost
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nazila Kassaian
- Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Laleh Hoveida
- Department of Microbiology, Falavarjan Branch, Islamic Azad University, PO Box: 84515/155, Isfahan, Iran.
| | - Babak Tamizifar
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Nili
- Zeitoon Vaccine Innovators Company, Isfahan Town of Science and Technology, Isfahan, Iran
| | - Marzieh Rahim Khorasani
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Adibi
- Isfahan Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
28
|
Riemann B, Antoine T, Béduneau A, Pellequer Y, Lamprecht A, Moulari B. Active nanoparticle targeting of MUC5AC ameliorates therapeutic outcome in experimental colitis. NANOSCALE 2024; 16:5715-5728. [PMID: 38407269 DOI: 10.1039/d3nr05681c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory diseases of the gastrointestinal tract and are characterized by chronic recurrent ulceration of the bowels. Colon-targeted drug delivery systems (DDS) have received significant attention for their potential to treat IBD by improving the inflamed tissue selectivity. Herein, antiMUC5AC-decorated drug loaded nanoparticles (NP) are suggested for active epithelial targeting and selective adhesion to the inflamed tissue in experimental colitis. NPs conjugated with antiMUC5AC (anti-MUC5) were tested for their degree of bioadhesion with HT29-MTX cells by comparison with non-targeted BSA-NP conjugates. In vivo, the selectivity of bioadhesion and the influence of ligand density in bioadhesion efficiency as well as the therapeutic benefit for glucocorticoid loaded anti-MUC5-NP were studied in a murine colitis model. Quantitative adhesion analyses showed that anti-MUC5-conjugated NP exhibited a much higher binding and selectivity to inflamed tissue compared to PNA-, IgG1- and BSA-NP conjugates used as controls. This bioadhesion efficiency was found to be dependent on the ligand density, present at the NP surface. The binding specificity between anti-MUC5 ligand and inflamed tissues was confirmed by fluorescence imaging. Both anti-MUC5-NP and all other glucocorticoid containing formulations led to a significant mitigation of the experimental colitis, as became evident from the substantial reduction of myeloperoxidase activity and pro-inflammatory cytokine concentrations (TNF-α, IL-1β). Targeted NP by using anti-MUC5 appears to be a very promising tool in future treatment of various types of local disorders affecting the gastro-intestinal tract but not limited to colitis.
Collapse
Affiliation(s)
- Bernadette Riemann
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Thomas Antoine
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Arnaud Béduneau
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Yann Pellequer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Alf Lamprecht
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Brice Moulari
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| |
Collapse
|
29
|
Lu J, Su D, Yang Y, Shu M, Wang Y, Zhou X, Yu Q, Li C, Xie J, Chen Y. Disruption of intestinal epithelial permeability in the Co-culture system of Caco-2/HT29-MTX cells exposed individually or simultaneously to acrylamide and ochratoxin A. Food Chem Toxicol 2024; 186:114582. [PMID: 38460668 DOI: 10.1016/j.fct.2024.114582] [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: 06/22/2023] [Revised: 12/13/2023] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Mycotoxins and thermal processing hazards are common contaminants in various foods and cause severe problems in terms of food safety and health. Combined use of acrylamide (AA) and ochratoxin A (OTA) would result in more significant intestinal toxicity than either toxin alone, but the underlying mechanisms behind this poor outcome remain unclear. Herein, we established the co-culture system of Caco-2/HT29-MTX cells for simulating a real intestinal environment that is more sensitive to AA and OTA, and showed that the combination of AA and OTA could up-regulate permeability of the intestine via increasing LY permeabilization, and decreasing TEER, then induce oxidative stress imbalance (GSH, SOD, MDA, and ROS) and inflammatory system disorder (TNF-α, IL-1β, IL-10, and IL-6), thereby leading a rapid decline in cell viability. Western blot, PAS- and AB-staining revealed that AA and OTA showed a synergistic effect on the intestine mainly through the disruption of tight junctions (TJs) and a mucus layer. Furthermore, based on correlation analysis, oxidative stress was more relevant to the mucus layer and TJs. Therefore, our findings provide a better evaluation model and a potential mechanism for further determining or preventing the combined toxicity caused by AA and OTA.
Collapse
Affiliation(s)
- Jiawen Lu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Dan Su
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Ying Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Mengni Shu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Yuting Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Qiang Yu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Chang Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Jianhua Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China
| | - Yi Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, China.
| |
Collapse
|
30
|
Acosta JE, Burns JL, Hillyer LM, Van K, Brendel EBK, Law C, Ma DWL, Monk JM. Effect of Lifelong Exposure to Dietary Plant and Marine Sources of n-3 Polyunsaturated Fatty Acids on Morphologic and Gene Expression Biomarkers of Intestinal Health in Early Life. Nutrients 2024; 16:719. [PMID: 38474847 DOI: 10.3390/nu16050719] [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: 02/05/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Altered intestinal health is also associated with the incidence and severity of many chronic inflammatory conditions, which could be attenuated via dietary n-3 PUFA interventions. However, little is known about the effect of lifelong exposure to n-3 PUFA from plant and marine sources (beginning in utero via the maternal diet) on early life biomarkers of intestinal health. Harems of C57Bl/6 mice were randomly assigned to one of three isocaloric AIN-93G modified diets differing in their fat sources consisting of the following: (i) 10% safflower oil (SO, enriched in n-6 PUFA), (ii) 3% flaxseed oil + 7% safflower oil (FX, plant-based n-3 PUFA-enriched diet), or (iii) 3% menhaden fish oil + 7% safflower oil (MO, marine-based n-3 PUFA-enriched diet). Mothers remained on these diets throughout pregnancy and offspring (n = 14/diet) continued on the same parental diet until termination at 3 weeks of age. In ileum, villi:crypt length ratios were increased in both the FX and MO dietary groups compared to SO (p < 0.05). Ileum mRNA expression of critical intestinal health biomarkers was increased by both n-3 PUFA-enriched diets including Relmβ and REG3γ compared to SO (p < 0.05), whereas only the FX diet increased mRNA expression of TFF3 and Muc2 (p < 0.05) and only the MO diet increased mRNA expression of ZO-1 (p < 0.05). In the proximal colon, both the FX and MO diets increased crypt lengths compared to SO (p < 0.05), whereas only the MO diet increased goblet cell numbers compared to SO (p < 0.05). Further, the MO diet increased proximal colon mRNA expression of Relmβ and REG3γ (p < 0.05) and both MO and FX increased mRNA expression of Muc2 compared to SO (p < 0.05). Collectively, these results demonstrate that lifelong exposure to dietary n-3 PUFA, beginning in utero, from both plant and marine sources, can support intestinal health development in early life. The differential effects between plant and marine sources warrants further investigation for optimizing health.
Collapse
Affiliation(s)
- Julianna E Acosta
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jessie L Burns
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Lyn M Hillyer
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Kelsey Van
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Elaina B K Brendel
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Camille Law
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - David W L Ma
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Jennifer M Monk
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
31
|
Gutiérrez L, Bartelt L. Current Understanding of Giardia lamblia and Pathogenesis of Stunting and Cognitive Deficits in Children from Low- and Middle-Income Countries. CURRENT TROPICAL MEDICINE REPORTS 2024; 11:28-39. [PMID: 38993355 PMCID: PMC11238937 DOI: 10.1007/s40475-024-00314-2] [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] [Accepted: 01/10/2024] [Indexed: 07/13/2024]
Abstract
Purpose of Review Giardia lamblia is a common intestinal parasite worldwide, mainly in children from low- and middle-income countries (LMIC). Also, it has been associated with increased intestinal permeability, stunting, and cognitive impairment. Nonetheless, the pathogenesis of long-term consequences is difficult to elucidate. Recent Findings Recent studies try to understand the long-term consequences of Giardia infections. First, well-characterized studies associate Giardia with intestinal damage and child growth. Second, infections appear not to be associated with inflammation, but "lack of inflammation" may not, however, entirely exclude a pro-inflammatory pathway. Finally, some important amino acids are lower and could contribute to prolongate stunting and cognitive deficit. Summary Giardia infections in LMIC used to be associated with child growth shortfalls, gut permeability, and cognitive deficits. Multifactorial effects could be associated with Giardia, including nutritional, altered microbiota, and generation of potentially toxic microbial metabolic byproducts, all together increasing risk of long-term outcomes.
Collapse
Affiliation(s)
- Lester Gutiérrez
- Centro de Investigación de Enfermedades Tropicales (CIET), Faculty of Microbiology, University of Costa Rica, San José, Costa Rica
| | - Luther Bartelt
- Departments of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
32
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
33
|
Abstract
Biogeography is the study of species distribution and diversity within an ecosystem and is at the core of how we understand ecosystem dynamics and interactions at the macroscale. In gut microbial communities, a historical reliance on bulk sequencing to probe community composition and dynamics has overlooked critical processes whereby microscale interactions affect systems-level microbiota function and the relationship with the host. In recent years, higher-resolution sequencing and novel single-cell level data have uncovered an incredible heterogeneity in microbial composition and have enabled a more nuanced spatial understanding of the gut microbiota. In an era when spatial transcriptomics and single-cell imaging and analysis have become key tools in mammalian cell and tissue biology, many of these techniques are now being applied to the microbiota. This fresh approach to intestinal biogeography has given important insights that span temporal and spatial scales, from the discovery of mucus encapsulation of the microbiota to the quantification of bacterial species throughout the gut. In this Review, we highlight emerging knowledge surrounding gut biogeography enabled by the observation and quantification of heterogeneity across multiple scales.
Collapse
Affiliation(s)
- Giselle McCallum
- Department of Biology, Concordia University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carolina Tropini
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
- Humans and the Microbiome Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario, Canada.
| |
Collapse
|
34
|
Keane DP, Constantine CJ, Mellor MD, Poling-Skutvik R. Nanoparticle transport in biomimetic polymer-linked emulsions. AIChE J 2024; 70:e18307. [PMID: 40017798 PMCID: PMC11867629 DOI: 10.1002/aic.18307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 10/31/2023] [Indexed: 03/01/2025]
Abstract
The ability of nanoparticles to penetrate and transport through soft tissues is essential to delivering therapeutics to treat diseases or signaling agents for advanced imaging and sensing. Nanoparticle transport in biological systems, however, is challenging to predict and control due to the physicochemical complexity of tissues and biological fluids. Here, we demonstrate that nanoparticles suspended in a novel class of soft matter-polymer-linked emulsions (PLEs)-exhibit characteristics essential for mimicking transport in biological systems, including subdiffusive dynamics, non-Gaussian displacement distributions, and decoupling of dynamics from material viscoelasticity. Using multiple particle tracking, we identify the physical mechanisms underlying this behavior, which we attribute to a coupling of nanoparticle dynamics to fluctuations in the local network of polymer-linked droplets. Our findings demonstrate the potential of PLEs to serve as fully synthetic mimics of biological transport.
Collapse
Affiliation(s)
- Daniel P Keane
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Colby J Constantine
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Matthew D Mellor
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| | - Ryan Poling-Skutvik
- Department of Chemical Engineering, University of Rhode Island, Kingstown, Rhode Island, USA
| |
Collapse
|
35
|
Donadio JLS, Fabi JP, Sztein MB, Salerno-Gonçalves R. Dietary fiber pectin: challenges and potential anti-inflammatory benefits for preterms and newborns. Front Nutr 2024; 10:1286138. [PMID: 38283907 PMCID: PMC10811139 DOI: 10.3389/fnut.2023.1286138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Pectins, a class of dietary fibers abundant in vegetables and fruits, have drawn considerable interest due to their potential anti-inflammatory properties. Numerous studies have indicated that incorporating pectins into infant formula could be a safe strategy for alleviating infant regurgitation and diarrhea. Moreover, pectins have been shown to modulate cytokine production, macrophage activity, and NF-kB expression, all contributing to their anti-inflammatory effects. Despite this promising evidence, the exact mechanisms through which pectins exert these functions and how their structural characteristics influence these processes remain largely unexplored. This knowledge is particularly significant in the context of gut inflammation in developing preterm babies, a critical aspect of necrotizing enterocolitis (NEC), and in children and adults dealing with inflammatory bowel disease (IBD). Our mini review aims to provide an up-to-date compilation of relevant research on the effects of pectin on gut immune responses, specifically focusing on preterms and newborns. By shedding light on the underlying mechanisms and implications of pectin-mediated anti-inflammatory properties, this review seeks to advance our knowledge in this area and pave the way for future research and potential therapeutic interventions.
Collapse
Affiliation(s)
- Janaina L. S. Donadio
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - João Paulo Fabi
- Department of Food Science and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Food Research Center (FoRC), CEPID-FAPESP (Research, Innovation and Dissemination Centers, São Paulo Research Foundation), São Paulo, Brazil
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
36
|
Liu W, Liu J, Li D, Han H, Yan H, Sun Y, Lei Q, Wang J, Zhou Y, Cao D, Li H, Li F. Effect of Lactobacillus salivarius SNK-6 on egg quality, intestinal morphology, and cecal microbial community of laying hens. Poult Sci 2024; 103:103224. [PMID: 37980753 PMCID: PMC10658386 DOI: 10.1016/j.psj.2023.103224] [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: 08/31/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/21/2023] Open
Abstract
The objective of this study was to investigate the effect of Lactobacillus salivarius (L. salivarius) SNK-6 supple-mentation on the laying performance, egg quality, blood parameters, intestinal morphology, and cecal microbial community of laying hens. A total of 432 healthy 30-wk-age laying hens were randomly divided into 3 groups with 6 replicates under the same husbandry and dietary regimes: control (CON); 2.0 × 108 CFU/kg L. salivarius supplementation (T1); 2.0 × 109 CFU/kg L. salivarius supplementation (T2). The experiment lasted for 10 wk. The results indicated that the supplementation resulted in a significant reduction in the broken egg and unqualified egg ratios, and a significant increase in the eggshell strength, eggshell relative weight, albumen height, and Haugh units (P < 0.05). The L. salivarius-treated hens exhibited significantly reduced serum malondialdehyde levels (P < 0.05); significantly increased total protein, phosphorus, calcitonin, and immunoglobulin M (P < 0.05); significantly increased cecal secretory immunoglobulin A concentration (P < 0.05); significantly improved villus height (VH) in the duodenum and VH to crypt depth ratio in the jejunum (P < 0.05). The serum globulin and interleukin-1β, immunoglobulin G concentrations, and catalase activity significantly increased in T2 (P < 0.05). Furthermore, the serum interferon-α level in T1 was significantly higher than that of the CON (P < 0.05). The intestinal barrier-related mRNA gene ZO-1, CLDN1, and MUC2 expression in the jejunum was significantly upregulated in the T1 and T2 groups (P < 0.05). The Firmicutes/Bacteroidetes ratio was higher and the relative abundances of Flavonifractor and Clostridiales_noname were significantly higher in the T1 group (P < 0.05). In conclusion, dietary supplementation with L. salivarius SNK-6 may improve hen egg quality, serum antioxidant capacity, immune function, and intestinal health.
Collapse
Affiliation(s)
- Wei Liu
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Jie Liu
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Dapeng Li
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Haixia Han
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Huaxiang Yan
- Shanghai Academy of Agricultural Sciences, Animal Husbandry and Veterinary Research Institute, 201106, Shanghai, China
| | - Yan Sun
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Qiuxia Lei
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Jie Wang
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Yan Zhou
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Dingguo Cao
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Huimin Li
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China
| | - Fuwei Li
- Shandong Academy of Agricultural Sciences, Poultry Institute, 250100, Jinan, China; Poultry Breeding Engineering Technology Center of Shandong Province, 250100, Jinan, China; Jinan Key Laboratory of Poultry Germplasm Resources Innovation and Healthy Breeding, 250100, Jinan, China.
| |
Collapse
|
37
|
Islam MM, Raikwar S. Enhancement of Oral Bioavailability of Protein and Peptide by Polysaccharide-based Nanoparticles. Protein Pept Lett 2024; 31:209-228. [PMID: 38509673 DOI: 10.2174/0109298665292469240228064739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/22/2024]
Abstract
Oral drug delivery is a prevalent and cost-effective method due to its advantages, such as increased drug absorption surface area and improved patient compliance. However, delivering proteins and peptides orally remains a challenge due to their vulnerability to degradation by digestive enzymes, stomach acids, and limited intestinal membrane permeability, resulting in poor bioavailability. The use of nanotechnology has emerged as a promising solution to enhance the bioavailability of these vital therapeutic agents. Polymeric NPs, made from natural or synthetic polymers, are commonly used. Natural polysaccharides, such as alginate, chitosan, dextran, starch, pectin, etc., have gained preference due to their biodegradability, biocompatibility, and versatility in encapsulating various drug types. Their hydrophobic-hydrophilic properties can be tailored to suit different drug molecules.
Collapse
Affiliation(s)
- Md Moidul Islam
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| |
Collapse
|
38
|
Abubaker S, Miri S, Mottawea W, Hammami R. Microbial Extracellular Vesicles in Host-Microbiota Interactions. Results Probl Cell Differ 2024; 73:475-520. [PMID: 39242390 DOI: 10.1007/978-3-031-62036-2_19] [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] [Indexed: 09/09/2024]
Abstract
Extracellular vesicles have emerged as key players in cellular communication, influencing various physiological processes and pathophysiological progression, including digestion, immune response, and tissue repairs. Recently, a class of EVs derived from microbial communities has gained significant attention due to their pivotal role in intercellular communication and their potential as biomarkers and biotherapeutic agents. Microbial EVs are membrane-bound molecules encapsulating bioactive metabolites that modulate host physiological and pathological processes. This chapter discusses the evolving history of microbiota-produced EVs, including their discovery, characterization, current research status, and their diverse mechanisms of interaction with other microbes and hosts. This review also highlights the importance of EVs in health and disease and discusses recent research that shows promising results for the therapeutic potential of EVs.
Collapse
Affiliation(s)
- Sarah Abubaker
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Saba Miri
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Walid Mottawea
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Riadh Hammami
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada.
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
39
|
Horikiri M, Taniguchi M, Yoshikawa HY, Okumura R, Matsuzaki T. Mechanical Characterization of Mucus on Intestinal Tissues by Atomic Force Microscopy. Methods Mol Biol 2024; 2763:403-414. [PMID: 38347430 DOI: 10.1007/978-1-0716-3670-1_35] [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] [Indexed: 02/15/2024]
Abstract
Mucus is part of the innate immune system that defends the mucosa against microbiota and other infectious threats. The mechanical characteristics of mucus, such as viscosity, elasticity, and lubricity, are critically involved in its barrier function. However, assessing the mechanical properties of mucus remains challenging because of technical limitations. Thus, a new approach that characterizes the mechanical properties of mucus on colonic tissues needs to be developed. Here, we describe a novel strategy to characterize the ex vivo mechanical properties of mucus on colonic tissues using atomic force microscopy. This description includes the preparation of the mouse colon sample, AFM calibration, and determining the elasticity (Young's modulus, E [kPa]) of the mucus layer in the colon.
Collapse
Affiliation(s)
- Momoka Horikiri
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Mugen Taniguchi
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Y Yoshikawa
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Ryu Okumura
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Takahisa Matsuzaki
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Osaka, Japan.
- Center for Future Innovation, Graduate School of Engineering, Osaka University, Osaka, Japan.
| |
Collapse
|
40
|
Jia J, Zheng W, Tang S, Song S, Ai C. Scytosiphon lomentaria fucoidan ameliorates DSS-induced colitis in dietary fiber-deficient mice via modulating the gut microbiota and inhibiting the TLR4/NF-κB/MLCK pathway. Int J Biol Macromol 2023; 253:127337. [PMID: 37820918 DOI: 10.1016/j.ijbiomac.2023.127337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/28/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The prevalence of ulcerative colitis (UC) poses a serious threat to human health. This study showed that fiber-deficient diet (FD) increased the susceptibility of mice to low dosage of DSS-induced UC, and a UC model was established by feeding mice with DSS and FD to evaluate the effect of Scytosiphon lomentaria fucoidan (SLF) on UC. SLF ameliorated the symptoms of UC, as evidenced by increases in colon length, goblet cells and glycoprotein and reduction in inflammatory cell infiltration and intestinal epithelial injury. SLF alleviated oxidative stress and inhibited colonic inflammation by reducing the levels of lipopolysaccharides and pro-inflammatory cytokines and suppressing the activation of nuclear factor kappa B pathway. SLF protected tight junction integrity by reducing the level of myosin light chain kinase and increasing the levels of claudin, zonula occludens-1 and occludin. SLF improved serum metabolites profile and affected multiple metabolic pathways that are crucial to human health, e.g. butanoate metabolism. The underlying mechanism can be associated with modulation of the gut microbiota and metabolites, including increases in short chain fatty acids and reduction in Proteobacteria, Bacteroides and Romboutsia. It suggests that SLF could be developed as a prebiotic polysaccharide to benefit human health by improving intestinal microecology.
Collapse
Affiliation(s)
- Jinhui Jia
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuangru Tang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
41
|
Zheng W, Jia J, Tang S, Song S, Ai C. Undaria pinnatifida fucoidan contributes to anti-inflammation activity of Bacteroides in fiber-deficient mice via modulation of gut microbiota and protection of intestinal barrier integrity. Int J Biol Macromol 2023; 252:126256. [PMID: 37572807 DOI: 10.1016/j.ijbiomac.2023.126256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/14/2023]
Abstract
Bacteroides as potential probiotics has several health benefits to the host, but its practical application faces many challenges due to its inherent properties. In this study, Bacteroides strains isolated from human feces alleviated colonic inflammation in mice, as evidenced by increased colon length and reduced tissue damage. Further study showed that anti-inflammation activity of Bacteroides strains was disturbed by dietary fiber deficiency (FD), which disrupted the balance between gut microbiota and colonic mucus layer, leading to a thinning of colonic mucus layer. A combination of Bacteroides strains and Undaria pinnatifida fucoidan (UPF) better alleviated colonic inflammation than either of them, including increases in the densities of goblet cells and glycoproteins and reduction in intestinal epithelial damage, pro-inflammatory cytokines and oxidative stress. The underlying mechanisms can be attributed to that UPF-induced alterations of mucosal microbiota cannot only directly benefit host health but also create an ecological condition that facilitates Bacteroides strains exert their healthy properties. In addition, both Bacteroides strains and UPF improved FD-induced lipid metabolism abnormality, mainly involving glycerophospholipid metabolism pathway. This study suggests that the application of Bacteroides has certain limitations, and UPF can be developed as a probiotic adjuvant for Bacteroides to enhance human health.
Collapse
Affiliation(s)
- Weiyun Zheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Jinhui Jia
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuangru Tang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
42
|
Cohen N, Orenbuch-Harroch E, Olshtain-Pops K, Lachish T, Korem M. Epidemiology, Clinical Characteristics and Risk Factors for Severity of Chronic Disseminated Candidiasis in Jerusalem, Israel. Mycopathologia 2023; 188:873-883. [PMID: 37326819 DOI: 10.1007/s11046-023-00755-9] [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: 04/14/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Chronic disseminated candidiasis (CDC) occurs mostly in patients with acute hematologic malignancy and its clinical manifestations derive from immune reconstitution following neutrophil recovery. The aim of this study was to describe epidemiological and clinical characteristics of CDC and define risk factors for disease severity. Demographic and clinical data were collected from medical files of patients with CDC hospitalized in two tertiary medical centers in Jerusalem between 2005 and 2020. Associations between different variables and disease severity were evaluated, as well as characterization of Candida species. The study included 35 patients. CDC incidence slightly increased during study years and the average number of involved organs and disease duration was 3 ± 1.26 and 178 ± 123 days, respectively. Candida grew in blood in less than third of cases and the most common isolated pathogen was Candida tropicalis (50%). Histopathological or microbiological workup in patients who underwent an organ biopsy demonstrated Candida in about half of the patients. Nine months after starting antifungals, 43% of the patients still didn't have resolution of organ lesions in imaging modalities. Factors associated with protracted and extensive disease were prolonged fever prior to CDC and absence of candidemia. A C- Reactive Protein (CRP) cutoff level of 7.18 mg/dL was found to predict extensive disease. In conclusion, CDC incidence is increasing and the number of involved organs is higher than previously described. Clinical factors such as fever duration prior to CDC and absence of candidemia can predict severe course of disease and assist in treatment decisions and follow-up planning.
Collapse
Affiliation(s)
- Nerel Cohen
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Medical Corps, Israel Defense Forces, Ramat-Gan, Israel
| | - Efrat Orenbuch-Harroch
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Medical Intensive Care Unit, Hadassah Medical Center, Jerusalem, Israel
| | - Karen Olshtain-Pops
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, P.O. Box 12000, 9112102, Jerusalem, Israel
| | - Tamar Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Infectious Diseases Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Maya Korem
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- The Department of Clinical Microbiology and Infectious Diseases, Hadassah Medical Center, P.O. Box 12000, 9112102, Jerusalem, Israel.
| |
Collapse
|
43
|
Ma Q, Zhang X, Xu X, Lu Y, Chen Q, Chen Y, Liu C, Chen K. Long-term oral administration of burdock fructooligosaccharide alleviates DSS-induced colitis in mice by mediating anti-inflammatory effects and protection of intestinal barrier function. Immun Inflamm Dis 2023; 11:e1092. [PMID: 38018589 PMCID: PMC10664397 DOI: 10.1002/iid3.1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Ulcerative colitis, a typical subtype of inflammatory bowel disease, can cause many serious complications. Burdock fructooligosaccharide (BFO), a linear inulin with a purity of 99.439% and a molecular weight of 2345 Da, demonstrates anti-inflammatory and immunomodulatory properties. METHODS The Kunming mice were divided into two experimental models: a normal pretreatment model and a colitis experimental model. During the experimental treatment period, we assessed changes in weight and disease activity index (DAI), quantified the intestinal index, and determined myeloperoxidase (MPO) activity and reactive oxide species (ROS) levels in colitis mice. We also photographed colon morphology to investigate alterations in the integrity of the intestinal barrier function. Finally, we performed ELISA and qRT-PCR to evaluate the anti-inflammatory effect of BFO treatment on colitis mice. RESULT The long-term oral administration of BFO alone exhibited protective effects by preventing disruption of the intestinal functional structure and increasing the colon index in mice. However, in a dextran sodium sulfate (DSS)-induced colitis mouse model, BFO administration facilitated quick recovery of body weight and effectively reduced the DAI, especially in the BFO-H group (500 mg/kg/day). BFO treatment maintained the integrity of the intestinal barrier by attenuating the crypt distortion and increasing the goblet cells count It restored the DSS-induced colon shortening and reduced the symptoms of colitis. These effects may be attributed to the appropriate concentrations of BFO effectively inhibiting MPO activity, clearing excessive ROS, and relieving spleen abnormalitie. BFO also attenuated the overexpression and excessive secretion of inflammatory cytokines (TNF-α, IL-1β, IL-6, and MCP-1) induced by DSS, reduced intestinal inflammation, and consequently protected the intestinal barrier function. CONCLUSION BFO effectively alleviated the symptoms of DSS-induced colitis by mediating anti-inflammatory effects and protecting the intestinal barrier integrity, thereby potentially facilitating the utilization of safer and more efficacious polysaccharides for managing chronic inflammatory diseases.
Collapse
Affiliation(s)
- Qunfei Ma
- School of Life ScienceShandong UniversityQingdaoChina
- Department of PhysiologyNaval Medical UniversityShanghaiChina
| | - Xiujuan Zhang
- School of Life ScienceShandong UniversityQingdaoChina
| | - Xuan Xu
- School of Life ScienceShandong UniversityQingdaoChina
| | - Yan Lu
- School of Life ScienceShandong UniversityQingdaoChina
- Clinical Laboratory Medicine DepartmentJining No. 1 People's HospitalJiningChina
| | - Qiang Chen
- Burdock Biotechnology (Dezhou) Co., LtdDezhouChina
| | - Yiru Chen
- Burdock Biotechnology (Dezhou) Co., LtdDezhouChina
| | - Chunyan Liu
- Provincial Engineering Laboratory for Screening and Re‐Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of PharmacyDrug Research & Development Center, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical CollegeWuhuChina
| | - Kaoshan Chen
- School of Life ScienceShandong UniversityQingdaoChina
- Provincial Engineering Laboratory for Screening and Re‐Evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of PharmacyDrug Research & Development Center, Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Wannan Medical CollegeWuhuChina
| |
Collapse
|
44
|
Jha SK, Imran M, Jha LA, Hasan N, Panthi VK, Paudel KR, Almalki WH, Mohammed Y, Kesharwani P. A Comprehensive review on Pharmacokinetic Studies of Vaccines: Impact of delivery route, carrier-and its modulation on immune response. ENVIRONMENTAL RESEARCH 2023; 236:116823. [PMID: 37543130 DOI: 10.1016/j.envres.2023.116823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The lack of knowledge about the absorption, distribution, metabolism, and excretion (ADME) of vaccines makes former biopharmaceutical optimization difficult. This was shown during the COVID-19 immunization campaign, where gradual booster doses were introduced.. Thus, understanding vaccine ADME and its effects on immunization effectiveness could result in a more logical vaccine design in terms of formulation, method of administration, and dosing regimens. Herein, we will cover the information available on vaccine pharmacokinetics, impacts of delivery routes and carriers on ADME, utilization and efficiency of nanoparticulate delivery vehicles, impact of dose level and dosing schedule on the therapeutic efficacy of vaccines, intracellular and endosomal trafficking and in vivo fate, perspective on DNA and mRNA vaccines, new generation sequencing and mathematical models to improve cancer vaccination and pharmacology, and the reported toxicological study of COVID-19 vaccines. Altogether, this review will enhance the reader's understanding of the pharmacokinetics of vaccines and methods that can be implied in delivery vehicle design to improve the absorption and distribution of immunizing agents and estimate the appropriate dose to achieve better immunogenic responses and prevent toxicities.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biomedicine, Health & Life Convergence Sciences, Mokpo National University, Muan-gun, Jeonnam, 58554, Republic of Korea; Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, 208016, Uttar Pradesh, India.
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Laxmi Akhileshwar Jha
- H. K. College of Pharmacy, Mumbai University, Pratiksha Nagar, Jogeshwari, West Mumbai, 400102, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vijay Kumar Panthi
- Department of Pharmacy, College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney, 2007, Australia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 24381, Saudi Arabia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
45
|
Kraski A, Mousavi S, Heimesaat MM, Bereswill S, Einspanier R, Alter T, Gölz G, Sharbati S. miR-125a-5p regulates the sialyltransferase ST3GAL1 in murine model of human intestinal campylobacteriosis. Gut Pathog 2023; 15:48. [PMID: 37848994 PMCID: PMC10583435 DOI: 10.1186/s13099-023-00577-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/08/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Zoonotic microorganisms are increasingly impacting human health worldwide. Due to the development of the global population, humans and animals live in shared and progressively crowded ecosystems, which enhances the risk of zoonoses. Although Campylobacter species are among the most important bacterial zoonotic agents worldwide, the molecular mechanisms of many host and pathogen factors involved in colonisation and infection are poorly understood. Campylobacter jejuni colonises the crypts of the human colon and causes acute inflammatory processes. The mucus and associated proteins play a central host-protective role in this process. The aim of this study was to explore the regulation of specific glycosyltransferase genes relevant to differential mucin-type O-glycosylation that could influence host colonisation and infection by C. jejuni. RESULTS Since microRNAs are known to be important regulators of the mammalian host cell response to bacterial infections, we focussed on the role of miR-125a-5p in C. jejuni infection. Combining in vitro and in vivo approaches, we show that miR-125a-5p regulates the expression of the sialyltransferase ST3GAL1 in an infection-dependent manner. The protein ST3GAL1 shows markedly increased intestinal levels in infected mice, with enhanced distribution in the mucosal epithelial layer in contrast to naïve mice. CONCLUSION From our previous studies and the data presented here, we conclude that miR-125a-5p and the previously reported miR-615-3p are involved in regulating the glycosylation patterns of relevant host cell response proteins during C. jejuni infection. The miRNA-dependent modulation of mucin-type O-glycosylation could be part of the mucosal immune response, but also a pathogen-driven modification that allows colonisation and infection of the mammalian host.
Collapse
Affiliation(s)
- Angelina Kraski
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Berlin, Germany
| | - Ralf Einspanier
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Thomas Alter
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Greta Gölz
- Institute of Food Safety and Food Hygiene, Freie Universität Berlin, Berlin, Germany
| | - Soroush Sharbati
- Institute of Veterinary Biochemistry, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
46
|
Craig CF, Finkelstein DI, McQuade RM, Diwakarla S. Understanding the potential causes of gastrointestinal dysfunctions in multiple system atrophy. Neurobiol Dis 2023; 187:106296. [PMID: 37714308 DOI: 10.1016/j.nbd.2023.106296] [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: 08/03/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
Multiple system atrophy (MSA) is a rare, progressive neurodegenerative disorder characterised by autonomic, pyramidal, parkinsonian and/or cerebellar dysfunction. Autonomic symptoms of MSA include deficits associated with the gastrointestinal (GI) system, such as difficulty swallowing, abdominal pain and bloating, nausea, delayed gastric emptying, and constipation. To date, studies assessing GI dysfunctions in MSA have primarily focused on alterations of the gut microbiome, however growing evidence indicates other structural components of the GI tract, such as the enteric nervous system, the intestinal barrier, GI hormones, and the GI-driven immune response may contribute to MSA-related GI symptoms. Here, we provide an in-depth exploration of the physiological, structural, and immunological changes theorised to underpin GI dysfunction in MSA patients and highlight areas for future research in order to identify more suitable pharmaceutical treatments for GI symptoms in patients with MSA.
Collapse
Affiliation(s)
- Colin F Craig
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David I Finkelstein
- Parkinson's Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Rachel M McQuade
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE), Sunshine Hospital, St Albans, VIC 3021, Australia
| | - Shanti Diwakarla
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE), Sunshine Hospital, St Albans, VIC 3021, Australia.
| |
Collapse
|
47
|
Brasil VP, Siqueira RM, Campos FG, Yoshitani MM, Pereira GP, Mendonça RLDS, Kanno DT, Pereira JA, Martinez CAR. Mucin levels in glands of the colonic mucosa of rats with diversion colitis subjected to enemas containing sucralfate and n-acetylcysteine alone or in combination. Acta Cir Bras 2023; 38:e384023. [PMID: 37851785 PMCID: PMC10578094 DOI: 10.1590/acb384023] [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: 06/06/2023] [Accepted: 07/17/2023] [Indexed: 10/20/2023] Open
Abstract
PURPOSE To evaluate the tissue content of neutral and acidic mucins, sulfomucins and sialomucins in colonic glands devoid of intestinal transit after enemas containing sucralfate and n-acetylcysteine alone or in combination. METHODS Sixty-four rats underwent intestinal transit bypass. A colonic segment was collected to compose the white group (without intervention). After derivation, the animals were divided into two groups according to whether enemas were performed daily for two or four weeks. Each group was subdivided into four subgroups according to the substance used: control group: saline 0.9%; sucralfate group (SCF): SCF 2 g/kg/day; n-acetylcysteine group (NAC): NAC 100 mg/kg/day; and SCF+NAC group: SCF 2 g/kg/day + NAC 100 mg/kg/day.Neutral and acidic mucins were stained by periodic acid-Schiff and alcian-blue techniques, respectively. The distinction between sulfomucins and sialomucin was made by the high alcian-blue iron diamine technique. The content of mucins in the colonic glands was measured by computerized morphometry. The inflammatory score was assessed using a validated scale. The results between the groups were compared by the Mann-Whitney's test, while the variation according to time by the Kruskal-Wallis' test (Dunn's post-test). A significance level of 5% was adopted. RESULTS There was reduction in the inflammatory score regardless of the application of isolated or associated substances. Intervention with SCF+NAC increased the content of all mucin subtypes regardless of intervention time. CONCLUSIONS The application of SCF+NAC reduced the inflammatory process of the colonic mucosa and increased the content of different types of mucins in the colonic glands of segments excluded from fecal transit.
Collapse
Affiliation(s)
- Verena Palmeiras Brasil
- Universidade Estadual de Campinas – Postgraduate Program in Surgical Sciences – Campinas (São Paulo) – Brazil
| | - Rayama Moreira Siqueira
- Universidade Estadual de Campinas – Postgraduate Program in Surgical Sciences – Campinas (São Paulo) – Brazil
| | - Fabio Guilherme Campos
- Universidade de São Paulo – Department of Gastroenterology – Faculty of Medicine – São Paulo (São Paulo) – Brazil
| | - Mateus Magami Yoshitani
- Universidade São Francisco – Faculty of Medicine – Medical School – Bragança Paulista (São Paulo) – Brazil
| | - Geovanna Pacciulli Pereira
- Universidade São Francisco – Faculty of Medicine – Medical School – Bragança Paulista (São Paulo) – Brazil
| | | | - Danilo Toshio Kanno
- Universidade São Francisco – Faculty of Medicine – Medical School – Bragança Paulista (São Paulo) – Brazil
| | - José Aires Pereira
- Universidade São Francisco – Faculty of Medicine – Medical School – Bragança Paulista (São Paulo) – Brazil
| | - Carlos Augusto Real Martinez
- Universidade Estadual de Campinas – Postgraduate Program in Surgical Sciences – Campinas (São Paulo) – Brazil
- Universidade São Francisco – Faculty of Medicine – Medical School – Bragança Paulista (São Paulo) – Brazil
| |
Collapse
|
48
|
Wang Y, Li Z, Bao Y, Cui H, Li J, Song B, Wang M, Li H, Cui X, Chen Y, Chen W, Yang S, Yang Y, Jin Z, Si X, Li B. Colon-targeted delivery of polyphenols: construction principles, targeting mechanisms and evaluation methods. Crit Rev Food Sci Nutr 2023; 65:64-86. [PMID: 37823723 DOI: 10.1080/10408398.2023.2266842] [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] [Indexed: 10/13/2023]
Abstract
Polyphenols have received considerable attention for their promotive effects on colonic health. However, polyphenols are mostly sensitive to harsh gastrointestinal environments, thus, must be protected. It is necessary to design and develop a colon-targeted delivery system to improve the stability, colon-targeting and bioavailability of polyphenols. This paper mainly introduces research on colon-targeted controlled release of polyphenols. The physiological features affecting the dissolution, release and absorption of polyphenol-loaded delivery systems in the colon are first discussed. Simultaneously, the types of colon-targeted carriers with different release mechanisms are described, and colon-targeting assessment models that have been studied so far and their advantages and limitations are summarized. Based on the current research on polyphenols colon-targeting, outlook and reflections are proposed, with the goal of inspiring strategic development of new colon-targeted therapeutics to ensure that the polyphenols reach the colon with complete bioactivity.
Collapse
Affiliation(s)
- Yidi Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Zhiying Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yiwen Bao
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Jiaxin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Baoge Song
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Mengzhu Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Haikun Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xingyue Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yi Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Chen
- Faculty of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Shufang Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Yiyun Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Zhufeng Jin
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| |
Collapse
|
49
|
Kordahi MC, Delaroque C, Bredèche MF, Gewirtz AT, Chassaing B. Vaccination against microbiota motility protects mice from the detrimental impact of dietary emulsifier consumption. PLoS Biol 2023; 21:e3002289. [PMID: 37725584 PMCID: PMC10508614 DOI: 10.1371/journal.pbio.3002289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/07/2023] [Indexed: 09/21/2023] Open
Abstract
Dietary emulsifiers, including carboxymethylcellulose (CMC) and polysorbate 80 (P80), perturb gut microbiota composition and gene expression, resulting in a microbiota with enhanced capacity to activate host pro-inflammatory gene expression and invade the intestine's inner mucus layer. Such microbiota alterations promote intestinal inflammation, which can have a variety of phenotypic consequences including increased adiposity. Bacterial flagellin is a key mediator of emulsifiers' impact in that this molecule enables motility and is itself a pro-inflammatory agonist. Hence, we reasoned that training the adaptive mucosal immune system to exclude microbes that express flagellin might protect against emulsifiers. Investigating this notion found that immunizing mice with flagellin elicited an increase in mucosal anti-flagellin IgA and IgA-coated microbiota that would have otherwise developed in response to CMC and P80 consumption. Yet, eliciting these responses in advance via flagellin immunization prevented CMC/P80-induced increases in microbiota expression of pro-inflammatory agonists including LPS and flagellin. Furthermore, such immunization prevented CMC/P80-induced microbiota encroachment and deleterious pro-inflammatory consequences associated therewith, including colon shortening and increased adiposity. Hence, eliciting mucosal immune responses to pathobiont surface components, including flagellin, may be a means of combatting the array of inflammatory diseases that are promoted by emulsifiers and perhaps other modern microbiota stressors.
Collapse
Affiliation(s)
- Melissa C. Kordahi
- INSERM U1016, Team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Clara Delaroque
- INSERM U1016, Team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Marie-Florence Bredèche
- INSERM U1016, Team “Robustness and evolvability of life”, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Andrew T. Gewirtz
- Institute for Biomedical Sciences, Centre for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, Georgia, United States of America
| | - Benoit Chassaing
- INSERM U1016, Team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
| |
Collapse
|
50
|
Bai Y, Wang Y, Kang M, Gabe CM, Srirangapatanam S, Edwards A, Stoller M, Green SJ, Aloni S, Tamura N, Beniash E, Hardt M, Ho SP. Organic Matrix Derived from Host-Microbe Interplay Contributes to Pathological Renal Biomineralization. ACS NANOSCIENCE AU 2023; 3:335-346. [PMID: 37601921 PMCID: PMC10436370 DOI: 10.1021/acsnanoscienceau.2c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 08/22/2023]
Abstract
Matrix stones are a rare form of kidney stones. They feature a high percentage of hydrogel-like organic matter, and their formation is closely associated with urinary tract infections. Herein, comprehensive materials and biochemical approaches were taken to map the organic-inorganic interface and gather insights into the host-microbe interplay in pathological renal biomineralization. Surgically extracted soft and slimy matrix stones were examined using micro-X-ray computed tomography and various microspectroscopy techniques. Higher-mineral-density laminae were positive for calcium-bound Alizarin red. Lower-mineral-density laminae revealed periodic acid-Schiff-positive organic filamentous networks of varied thickness. These organic filamentous networks, which featured a high polysaccharide content, were enriched with zinc, carbon, and sulfur elements. Neutrophil extracellular traps (NETs) along with immune response-related proteins, including calprotectin, myeloperoxidase, CD63, and CD86, also were identified in the filamentous networks. Expressions of NETs and upregulation of polysaccharide-rich mucin secretion are proposed as a part of the host immune defense to "trap" pathogens. These host-microbe derived organic matrices can facilitate heterogeneous nucleation and precipitation of inorganic particulates, resulting in macroscale aggregates known as "matrix stones". These insights into the plausible aggregation of constituents through host-microbe interplay underscore the unique "double-edged sword" effect of the host immune response to pathogens and the resulting renal biominerals.
Collapse
Affiliation(s)
- Yushi Bai
- Department
of Preventive and Restorative Dental Sciences, School of Dentistry, University of California San Francisco, San Francisco, California 94143, United States
| | - Yongmei Wang
- Department
of Preventive and Restorative Dental Sciences, School of Dentistry, University of California San Francisco, San Francisco, California 94143, United States
| | - Misun Kang
- Department
of Preventive and Restorative Dental Sciences, School of Dentistry, University of California San Francisco, San Francisco, California 94143, United States
| | - Claire M. Gabe
- Department
of Oral and Craniofacial Sciences, School of Dentistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Sudarshan Srirangapatanam
- Department
of Preventive and Restorative Dental Sciences, School of Dentistry, University of California San Francisco, San Francisco, California 94143, United States
- College
of Medicine, University of Central Florida, Orlando, Florida 32827, United States
| | - Austin Edwards
- Biological
Imaging Development Center, University of
California San Francisco, San Francisco, California 94143, United States
| | - Marshall Stoller
- Department
of Urology, School of Medicine, University
of California San Francisco, San Francisco, California 94143, United States
| | - Stefan J. Green
- Department
of Internal Medicine, Division of Infectious Diseases, Rush Medical
College, Rush University, Chicago, Illinois 60612, United States
| | - Shaul Aloni
- The
Molecular Foundry, Lawrence Berkeley National
Laboratory, Berkeley, California 94720, United States
| | - Nobumichi Tamura
- Advanced
Light Source, Lawrence Berkeley National
Laboratory, Berkeley, California 94720, United States
| | - Elia Beniash
- Department
of Oral and Craniofacial Sciences, School of Dentistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Markus Hardt
- Center
for Salivary Diagnostics, The Forsyth Institute, Cambridge, Massachusetts 02142, United States
- Department
of Developmental Biology, Harvard School
of Dental Medicine, Boston, Massachusetts 02115, United States
| | - Sunita P. Ho
- Department
of Preventive and Restorative Dental Sciences, School of Dentistry, University of California San Francisco, San Francisco, California 94143, United States
- Department
of Urology, School of Medicine, University
of California San Francisco, San Francisco, California 94143, United States
| |
Collapse
|