1
|
Patel JJ, Barash M. The Gut in Critical Illness. Curr Gastroenterol Rep 2025; 27:11. [PMID: 39792234 DOI: 10.1007/s11894-024-00954-4] [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: 10/30/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE OF REVIEW The purpose of this narrative review is to describe the mechanisms for gut dysfunction during critical illness, outline hypotheses of gut-derived inflammation, and identify nutrition and non-nutritional therapies that have direct and indirect effects on preserving both epithelial barrier function and gut microbiota during critical illness. RECENT FINDINGS Clinical and animal model studies have demonstrated that critical illness pathophysiology and interventions breach epithelial barrier function and convert a normally commensal gut microbiome into a pathobiome. As a result, the gut has been postulated to be the "motor" of critical illness and numerous hypotheses have been put forward to explain how it contributes to systemic inflammation and drives multiple organ failure. Strategies to ameliorate gut dysfunction have focused on maintaining gut barrier function and promoting gut microbiota commensalism. The trajectory of critical illness may be closely related to gut epithelial barrier function, the gut microbiome and interventions that may contribute towards a deleterious pathobiome with immune dysregulation.
Collapse
Affiliation(s)
- Jayshil J Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Medical College of Wisconsin, 8701 West Watertown Plank Road, 8th Floor: HUB for Collaborative Medicine, Milwaukee, WI, 53226, USA.
| | - Mark Barash
- Division of Pulmonary, Critical Care, and Sleep Medicine, Medical College of Wisconsin, 8701 West Watertown Plank Road, 8th Floor: HUB for Collaborative Medicine, Milwaukee, WI, 53226, USA
| |
Collapse
|
2
|
Liu X, Mei L, Wang J, Liu X, Yang Y, Wu Z, Ji Y. Cutting-edge insights into the mechanistic understanding of plant-derived exosome-like nanoparticles: Implications for intestinal homeostasis. Food Res Int 2025; 208:116186. [PMID: 40263791 DOI: 10.1016/j.foodres.2025.116186] [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: 10/20/2024] [Revised: 01/11/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Plant-derived exosome-like nanoparticles (PDELNs) are extracted from plants such as ginger, garlic, broccoli, and others, attracting attention for their therapeutic potential due to their availability and capacity for large-scale production. Their unique physicochemical properties position PDELNs as ideal candidates for targeted gut delivery, improving intestinal health by modulating mucosal immunity, gut microbiota, and intestinal barrier integrity, all essential for maintaining intestinal homeostasis. PDELNs regulate intestinal barrier function through their bioactive components (e.g. microRNAs, lipids, and proteins). These vesicles enhance the expression of tight junction proteins and stimulate mucin production. Additionally, they promote intestinal stem cell proliferation and increase the secretion of antimicrobial peptides. PDELNs also modulate inflammatory cytokine levels and immune cell activity, fostering a balanced immune response. Further, they support the growth of beneficial gut microbiota and their metabolites, while suppressing the proliferation of pathogenic bacteria. This review summarizes recent advancements in understanding the roles of PDELNs in regulating intestinal homeostasis, focusing on their impact on mucosal immunity, intestinal barrier function, and gut microbiota composition, along with underlying molecular mechanisms and therapeutic implications. Overall, PDELNs show promise as a novel approach for treating and preventing intestinal diseases, paving the way for effective gut health interventions.
Collapse
Affiliation(s)
- Xiyuan Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Xuelian Liu
- State Key Laboratory of Direct-Fed Microbial Engineering, Beijing, 100192, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
3
|
He Z, Guo M, Zhang X, Wang S, Liu T, Lin Y, Ouyang Q, Hu S, He H, Li L, Liu H, Wang J. 16S rRNA and transcriptome analysis revealed the regulatory mechanism of Romboutsia lituseburensis on serum immunoglobulin levels in geese. Poult Sci 2025; 104:105018. [PMID: 40086254 PMCID: PMC11957522 DOI: 10.1016/j.psj.2025.105018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Romboutsia is a dominant genus in the goose intestine. Recent studies have suggested that Romboutsia lituseburensis might regulate serum immunoglobulin levels in female geese, although the underlying mechanisms remain unclear. In this study, we administered Romboutsia lituseburensis (R. lituseburensis) orally to female geese, leading to successful colonization of the ileum. Subsequent analysis showed that the levels of IgM, IgA, and IgG in the serum significantly decreased after colonization (P < 0.01). 16S rRNA sequencing revealed that R. lituseburensis significantly altered the microbial composition and increased the relative abundance of Jeotgalicoccus (P < 0.01), Turicibacter, and Bacillus (P < 0.05) in the ileum. Transcriptome sequencing further identified 263 differentially expressed genes (DEGs) in the ileum (146 upregulated, 117 downregulated) and 725 DEGs in the spleen (300 upregulated, 425 downregulated). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that these DEGs were enriched in 17 pathways in the ileum and 21 pathways in the spleen. Notably, the "Intestinal immune network for IgA production" pathway was significantly enriched in the spleen (P < 0.05). Further, Short Time-series Expression Miner (STEM) analysis grouped the DEGs in these 2 tissues into 49 clusters, with clusters 27 and 29 showing the highest significance and similar expression patterns. Pathway analysis confirmed that the "Intestinal immune network for IgA production" pathway was enriched in both clusters. Furthermore, a protein-protein interaction (PPI) network of these 2 clusters, along with correlation analysis between microbiota abundance and gene expression, highlighted KEL, SERPING1, CALR, and OSTN as key hub genes. Overall, R. lituseburensis significantly increased the abundance of Jeotgalicoccus, Turicibacter, and Bacillus in the ileum. Concurrently, it might downregulate the "Intestinal immune network for IgA production" pathway in the spleen (CCR9, TNFRSF13B, AICDA) via KEL, SERPING1, CALR, and OSTN, thereby contributing to the reduction of serum immunoglobulin levels. These findings offer new insights into how R. lituseburensis influences immune function in female geese and provide a theoretical basis for further research into its other physiological roles in geese.
Collapse
Affiliation(s)
- Zhiyu He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Mengge Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Xi Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Shangmin Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Tanze Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Yueyue Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China.; State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, PR China..
| |
Collapse
|
4
|
Gao Z, Shao S, Xu Z, Nie J, Li C, Du C. IDO1 induced macrophage M1 polarization via ER stress-associated GRP78-XBP1 pathway to promote ulcerative colitis progression. Front Med (Lausanne) 2025; 12:1524952. [PMID: 40370742 PMCID: PMC12075526 DOI: 10.3389/fmed.2025.1524952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disorder distinguished by alternating phases of remission and exacerbation. Restoring immune balance through the modulation of M1 macrophage polarization represents a potentially valuable therapeutic strategy for UC. Indoleamine 2,3-dioxygenase-1 (IDO1) has been shown to contribute to macrophage plasticity, but its role in the pathogenesis of UC via regulating M1 macrophage polarization has not been studied yet. For the clinical component, we analyzed IDO1 expression in UC using bioinformatics analysis of Gene Expression Omnibus (GEO) datasets and validated the result using western blotting of colonic tissues from new recruited UC patients. Colitis was induced in mice via dextran sulfate sodium (DSS) treatment and subsequently treated with oral administration of 1-methyl-DL-tryptophan (1-MT), an inhibitor of IDO1 pathway. The results indicated that IDO1 expression was significantly elevated in UC patients and correlated with M1 macrophage polarization observed in both human data and colitis mice. Furthermore, 1-MT markedly ameliorated DSS-induced weight loss, colonic shortening and disease severity via inhibiting IDO1 expression level, downregulating GRP78-XBP1 pathway and reducing M1 proportion. Notably, in vitro study revealed that overexpressing IDO1 in RAW264.7 cells induced macrophage M1 polarization with increased expression levels of GRP78 and XBP1, which was attenuated by 1-MT treatment. Additionally, the catalytic effect exerted by IDO1 overexpression on M1 polarization was neutralized by employing an inhibitor targeting the endoplasmic reticulum (ER) stress pathway. Thus, our findings suggest that IDO1 may promote UC progression by skewing macrophages towards M1 polarization through ER stress-associated GRP78-XBP1 pathway.
Collapse
Affiliation(s)
- Zijian Gao
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Shuai Shao
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Zhen Xu
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Jiao Nie
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chenglin Li
- Department of Oncology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chao Du
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Weihai Municipal Hospital, Shandong University, Weihai, China
| |
Collapse
|
5
|
Valencia S, Zuluaga M, Florian Pérez MC, Montoya-Quintero KF, Candamil-Cortés MS, Robledo S. Human Gut Microbiome: A Connecting Organ Between Nutrition, Metabolism, and Health. Int J Mol Sci 2025; 26:4112. [PMID: 40362352 PMCID: PMC12071897 DOI: 10.3390/ijms26094112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 05/15/2025] Open
Abstract
The gut microbiome plays a vital role in human health, functioning as a metabolic organ that influences nutrient absorption and overall well-being. With growing evidence that dietary interventions can modulate the microbiome and improve health, this review examines whether healthcare systems should prioritize personalized microbiome-targeted therapies, such as probiotics, prebiotics, and microbiota transplants, over traditional pharmaceutical treatments for chronic diseases like obesity, diabetes, cardiovascular risk, and inflammatory conditions. A systematic review using Web of Science and Scopus databases was conducted, followed by a scientometric analysis. Key metabolic pathways, such as dietary fiber fermentation and short-chain fatty acid production, were explored, focusing on their impact on lipid and glucose metabolism. The interactions between microbial metabolites and the immune system were also investigated. Dietary interventions, including increased fiber and probiotic intake, show potential for addressing dysbiosis linked to conditions, such as type 2 diabetes, obesity, and autoimmune diseases. The review emphasizes the need to incorporate microbiome modulation strategies into clinical practice and research, calling for a multidisciplinary approach that integrates nutrition, microbiology, and biochemistry to better understand the gut microbiome's complex role in health.
Collapse
Affiliation(s)
- Sandra Valencia
- Centro de Bioinformática y Biología Computacional de Colombia—BIOS, Grupo de Investigación—BIOS, Parque los Yarumos, Manizales 170002, Colombia;
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Martha Zuluaga
- Dirección Académica, Universidad Nacional de Colombia, Sede De La Paz, Km 9 Valledupar—La Paz, Cesar 202010, Colombia;
| | - María Cristina Florian Pérez
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Kevin Fernando Montoya-Quintero
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias para la Salud, Universidad de Caldas, Calle 65 # 26-10, Manizales 170004, Colombia; (M.C.F.P.); (K.F.M.-Q.)
| | - Mariana S. Candamil-Cortés
- Centro de Bioinformática y Biología Computacional de Colombia—BIOS, Grupo de Investigación—BIOS, Parque los Yarumos, Manizales 170002, Colombia;
| | - Sebastian Robledo
- Dirección Académica, Universidad Nacional de Colombia, Sede De La Paz, Km 9 Valledupar—La Paz, Cesar 202010, Colombia;
| |
Collapse
|
6
|
Yu Z, Swift KA, Hedges MA, Theiss AL, Andres SF. Microscopic messengers: Extracellular vesicles shaping gastrointestinal health and disease. Physiol Rep 2025; 13:e70292. [PMID: 40165585 PMCID: PMC11959161 DOI: 10.14814/phy2.70292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The field of extracellular vesicles (EVs) is advancing rapidly, and this review aims to synthesize the latest research connected to EVs and the gastrointestinal tract. We will address new and emerging roles for EVs derived from internal sources such as the pancreas and immune system and how these miniature messengers alter organismal health or the inflammatory response within the GI tract. We will examine what is known about external EVs from dietary and bacterial sources and the immense anti-inflammatory, immune-modulatory, and proliferative potential within these nano-sized information carriers. EV interactions with the intestinal and colonic epithelium and associated immune cells at homeostatic and disease states, such as necrotizing enterocolitis (NEC) and inflammatory bowel disease (IBD) will also be covered. We will discuss how EVs are being leveraged as therapeutics or for drug delivery and conclude with a series of unanswered questions in the field.
Collapse
Affiliation(s)
- Zhantao Yu
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Kevin A. Swift
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Madeline A. Hedges
- Department of Neonatology, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Arianne L. Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
- Rocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
| | - Sarah F. Andres
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| |
Collapse
|
7
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
8
|
He G, Liu P, Xuan X, Zhang M, Zhang H, Yang K, Luan Y, Yang Q, Yang J, Li Q, Zheng H, Wang P. Transcription factor ELF-1 protects against colitis by maintaining intestinal epithelium homeostasis. Commun Biol 2025; 8:395. [PMID: 40057592 PMCID: PMC11890729 DOI: 10.1038/s42003-025-07742-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 02/14/2025] [Indexed: 05/13/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, relapsing, and remitting disease characterized by chronic inflammation in the gastrointestinal tract. The exact etiology and pathogenesis of IBD remain elusive. Although ELF-1 has been known to be highly expressed in epithelial cells for past twenty years, little is known about its function in epithelial cells and epithelial-related IBD. Here, we demonstrated that ELF-1 deficiency in mouse lead to exacerbated DSS-induced colitis, marked by inflammation dominated by neutrophil infiltration and activation of IL-17 signaling pathways in various immune cells, including Th17, ILC3, γδT and NKT cells. Bone marrow transfer experiments confirmed ELF-1 deficiency in non-hematopoietic cells intrinsically worsened DSS-induced colitis. On one hand, ELF-1 deficiency enhanced the production of pro-inflammatory chemokines in colonic epithelial cells, leading to extensive infiltration of neutrophils and other immune cells into the colonic mucosal tissue. On the other hand, ELF-1 directly regulated the expression of the Rack1 gene in colonic epithelial tissue, which has been proved to play critical roles in maintaining intestinal homeostasis. Altogether, ELF-1 plays a protective role in colitis by maintaining intestinal epithelium homeostasis.
Collapse
Affiliation(s)
- Gege He
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Pingping Liu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoyan Xuan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Min Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongxia Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ka Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yusheng Luan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qian Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jingyuan Yang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qianru Li
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Huaixin Zheng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Peng Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
9
|
Yang F, Li X, Sun J, Pang X, Sun Q, Lu Y. Regulatory mechanisms of the probiotic-targeted gut-liver axis for the alleviation of alcohol-related liver disease: a review. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 39905925 DOI: 10.1080/10408398.2025.2455954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Alcohol abuse-triggered alcohol-related liver disease (ALD) has become as a global public health concern that substantially affects the well-being and clinical status of patients. Although modern medicine provides various treatments for ALD, their effectiveness is limited and can lead to adverse side effects. Probiotics have been employed to prevent, alleviate, and even treat ALD, with promising results. However, few comprehensive reviews are available on how they mitigate ALD by targeting the gut-liver axis. This review systematically clarifies the specific mediators of the gut-liver axis in healthy states. It also describes the alterations observed in ALD. Furthermore, this review thoroughly summarizes the underlying mechanisms through which probiotics act on the gut-liver axis to relieve ALD. It also discusses the current status and challenges faced in clinical research applications. Finally, we discuss the challenges and future prospects of using probiotics to treat ALD. This review improves our understanding of ALD and supports the development and application of probiotics that target the gut-liver axis for therapeutic use.
Collapse
Affiliation(s)
- Feiyu Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
10
|
Liu J, Zhao J, Zhang YL, Zhang C, Yang GD, Tian WS, Zhou BH, Wang HW. Underlying Mechanism of Fluoride Inhibits Colonic Gland Cells Proliferation by Inducing an Inflammation Response. Biol Trace Elem Res 2025; 203:973-985. [PMID: 38995434 DOI: 10.1007/s12011-024-04212-6] [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: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 07/13/2024]
Abstract
The integrity of colonic gland cells is a prerequisite for normal colonic function and maintenance. To evaluate the underlying injury mechanisms in colonic gland cells induced by excessive fluoride (F), forty-eight female Kunming mice were randomly allocated into four groups and treated with different concentrations of NaF (0, 25, 50, and 100 mg F-/L) for 70 days. As a result, the integrity of the colonic mucosa and the cell layer was seriously damaged after F treatment, as manifested by atrophy of the colonic glands, colonic cell surface collapse, breakage of microvilli, and mitochondrial vacuolization. Alcian blue and periodic acid Schiff staining revealed that F decreased the number of goblet cells and glycoprotein secretion. Furthermore, F increased the protein expression of TLR4, NF-κB, and ERK1/2 and decreased IL-6, interfered with NF-κB signaling, following induced colonic gland cells inflammation. The accumulation of F inhibited proliferation via the JAK/STAT signaling pathway, as characterized by decreased mRNA and protein expression of JAK, STAT3, STAT5, PCNA, and Ki67 in colon tissue. Additionally, the expression of CDK4 was up-regulated by increased F concentration. In conclusion, excessive F triggered colonic inflammation and inhibited colonic gland cell proliferation via regulation of the NF-κB and JAK/STAT signaling pathways, leading to histopathology and barrier damage in the colon. The results explain the damaging effect of the F-induced inflammatory response on the colon from the perspective of cell proliferation and provide a new idea for explaining the potential mechanism of F-induced intestinal damage.
Collapse
Affiliation(s)
- Jing Liu
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Jing Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Yu-Ling Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Cai Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Guo-Dong Yang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Wei-Shun Tian
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Bian-Hua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, Henan University of Science and Technology, Luoyang, Henan, 471000, People's Republic of China.
| |
Collapse
|
11
|
Haider K, Sufian M, Abbas D, Kabir K, Ali MS, Kausar Y, Ghafar MA. The Role of Gut Microbiota in Shaping Immune Responses in Tephritidae Fruit Fly and Prospective Implications for Management. NEOTROPICAL ENTOMOLOGY 2025; 54:34. [PMID: 39881025 DOI: 10.1007/s13744-025-01248-8] [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: 08/30/2024] [Accepted: 12/21/2024] [Indexed: 01/31/2025]
Abstract
The interaction of microbial communities with host immunity has become one of the most explored research areas with significant implications for pest control strategies. It has been found that the gut microbiota plays substantial roles in immune response regulation and host-gut microbiome symbiosis, as well as in pathogen resistance and overall fitness in Tephritidae fruit flies that are major pests of agricultural importance. In this review, we discuss the modulation of immune responses of Tephritidae fruit flies by the gut microbiota with particular emphasis on the general interactions between microbiota and the immune system. These interactions help to unravel new horizons of pest management. Regulating gut microbiota modifies the performance of biological control agents and SIT and allows the creation of microbial therapies that affect the vital physiological functions of fruit flies. Besides, deploying microbes that can modulate the immune response and using microbial-derived signals provide an eco-friendly and more sustainable way of eradicating chemical pesticides and making farming systems less susceptible to climatic variability. This paper reviews various aspects of the possibility of using gut microbiota for changing the approach to Integrated Pest Management (IPM) programs that would improve methods of controlling Tephritidae fruit fly populations more ecologically.
Collapse
Affiliation(s)
- Kamran Haider
- Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural Univ, Wuhan, People's Republic of China
| | - Muhammad Sufian
- Dept of Entomology, Univ of Agriculture, Faisalabad, Pakistan
| | - Dilawar Abbas
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Kamil Kabir
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Horticultural Plant Biology (MOE), China-Australia Joint Research Centre for Horticultural and Urban Pests, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural Univ, Wuhan, People's Republic of China
| | | | - Yasmin Kausar
- Department of Zoology Wildlife and Fisheries, Univ of Agriculture, Faisalabad, 38000, Pakistan
| | - Muhammad Adeel Ghafar
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Bio Pesticide and Chemical Biology, MOE, College of Plant Protection, Fujian Agriculture and Forestry Univ, Fuzhou, China.
| |
Collapse
|
12
|
Hada A, Xiao Z. Ligands for Intestinal Intraepithelial T Lymphocytes in Health and Disease. Pathogens 2025; 14:109. [PMID: 40005486 PMCID: PMC11858322 DOI: 10.3390/pathogens14020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The intestinal tract is constantly exposed to a diverse mixture of luminal antigens, such as those derived from commensals, dietary substances, and potential pathogens. It also serves as a primary route of entry for pathogens. At the forefront of this intestinal defense is a single layer of epithelial cells that forms a critical barrier between the gastrointestinal (GI) lumen and the underlying host tissue. The intestinal intraepithelial T lymphocytes (T-IELs), one of the most abundant lymphocyte populations in the body, play a crucial role in actively surveilling and maintaining the integrity of this barrier by tolerating non-harmful factors such as commensal microbiota and dietary components, promoting epithelial turnover and renewal while also defending against pathogens. This immune balance is maintained through interactions between ligands in the GI microenvironment and receptors on T-IELs. This review provides a detailed examination of the ligands present in the intestinal epithelia and the corresponding receptors expressed on T-IELs, including T cell receptors (TCRs) and non-TCRs, as well as how these ligand-receptor interactions influence T-IEL functions under both steady-state and pathological conditions. By understanding these engagements, we aim to shed light on the mechanisms that govern T-IEL activities within the GI microenvironment. This knowledge may help in developing strategies to target GI ligands and modulate T-IEL receptor expression, offering precise approaches for treating intestinal disorders.
Collapse
Affiliation(s)
| | - Zhengguo Xiao
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
13
|
Choi H, Garavito-Duarte Y, Gormley AR, Kim SW. Aflatoxin B1: Challenges and Strategies for the Intestinal Microbiota and Intestinal Health of Monogastric Animals. Toxins (Basel) 2025; 17:43. [PMID: 39852996 PMCID: PMC11768593 DOI: 10.3390/toxins17010043] [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: 12/26/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
The objective of this review is to investigate the impacts of aflatoxins, particularly aflatoxin B1 (AFB1), on intestinal microbiota, intestinal health, and growth performance in monogastric animals, primarily chickens and pigs, as well as dietary interventions to mitigate these effects. Aflatoxin B1 contamination in feeds disrupts intestinal microbiota, induces immune responses and oxidative damage, increases antioxidant activity, and impairs jejunal cell viability, barrier function, and morphology in the small intestine. These changes compromise nutrient digestion and reduce growth performance in animals. The negative impact of AFB1 on the % change in average daily gain (ΔADG) of chickens and pigs was estimated based on meta-analysis: ΔADG (%)chicken = -0.13 × AFB1 intake per body weight (ng/g·d) and ΔADG (%)pig = -0.74 × AFB1 intake per body weight (µg/kg·d), indicating that increasing AFB1 contamination linearly reduces the growth of animals. To mitigate the harmful impacts of AFB1, various dietary strategies have been effective. Mycotoxin-detoxifying agents include mycotoxin-adsorbing agents, such as clay and yeast cell wall compounds, binding to AFB1 and mycotoxin-biotransforming agents, such as specific strains of Bacillus subtilis and mycotoxin-degrading enzyme, degrading AFB1 into non-toxic metabolites such as aflatoxin D1. Multiple mycotoxin-detoxifying agents are often combined and used together to improve the intestinal health and growth of chickens and pigs fed AFB1-contaminated feeds. In summary, AFB1 negatively impacts intestinal microbiota, induces immune responses and oxidative stress, disrupts intestinal morphology, and impairs nutrient digestion in the small intestine, leading to reduced growth performance. Supplementing multi-component mycotoxin-detoxifying agents in feeds could effectively adsorb and degrade AFB1 co-contaminated with other mycotoxins prior to its absorption in the small intestine, preventing its negative impacts on the intestinal health and growth performance of chickens and pigs.
Collapse
Affiliation(s)
| | | | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (H.C.); (Y.G.-D.); (A.R.G.)
| |
Collapse
|
14
|
Li X, Li C, Wu P, Zhang L, Zhou P, Ma X. Recent status and trends of innate immunity and the gut-kidney aixs in IgAN: A systematic review and bibliometric analysis. Int Immunopharmacol 2024; 143:113335. [PMID: 39423662 DOI: 10.1016/j.intimp.2024.113335] [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: 05/26/2024] [Revised: 09/24/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND There is a significant global demand for precise diagnosis and effective treatment of IgA nephropathy (IgAN), with innate immunity, particularly the complement system, exerting a profound influence on its pathogenesis. Additionally, the gut-kidney axis pathway is vital in the emergence and development of IgAN. METHODS We conducted a comprehensive search in the Web of Science database, spanning from January 1, 2000 to December 18, 2023. The gathered literature underwent a visual examination through CiteSpace, VOSviewer, and Scimago Graphica to delve into authors, nations, organizations, key terms, and other pertinent elements. RESULT Between 2000 and 2023, a total of 720 publications were identified, out of which 436 publications underwent screening for highly relevant literature analysis. The average annual number of articles focusing on IgAN, innate immunity, and the gut-kidney axis is approximately 31, with an upward trend observed. In terms of research impact encompassing publication count and authorship, the United States emerged as the leading contributor. Prominent keywords included "complement", "activation", "microbe", "gut-kidney axis", "C4d deposition", "alternative pathway" and "B cells" along with other prospective hot topics. CONCLUSION The correlation between IgAN and innate immunity is a focal point in current scientific research. Recent literature underscores the significance of the gut-kidney axis, where intestinal microorganisms and metabolites may influence IgAN. The complement system, a key component of innate immunity, also has a crucial function.Advancements in prevention, diagnosis, and treatment hinge on unraveling this intricate relationship.
Collapse
Affiliation(s)
- Xun Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Chengni Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Peiwen Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Lifang Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Ping Zhou
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| | - Xin Ma
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China; Department of Nephrology, The First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China.
| |
Collapse
|
15
|
Yu LE, Yang WC, Liang YC. Crosstalk Within the Intestinal Epithelium: Aspects of Intestinal Absorption, Homeostasis, and Immunity. Biomedicines 2024; 12:2771. [PMID: 39767678 PMCID: PMC11673925 DOI: 10.3390/biomedicines12122771] [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: 10/31/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Gut health is crucial in many ways, such as in improving human health in general and enhancing production in agricultural animals. To maximize the effect of a healthy gastrointestinal tract (GIT), an understanding of the regulation of intestinal functions is needed. Proper intestinal functions depend on the activity, composition, and behavior of intestinal epithelial cells (IECs). There are various types of IECs, including enterocytes, Paneth cells, enteroendocrine cells (EECs), goblet cells, tuft cells, M cells, and intestinal epithelial stem cells (IESCs), each with unique 3D structures and IEC distributions. Although the communication between IECs and other cell types, such as immune cells and neurons, has been intensively reviewed, communication between different IECs has rarely been addressed. The present paper overviews the networks among IECs that influence intestinal functions. Intestinal absorption is regulated by incretins derived from EECs that induce nutrient transporter activity in enterocytes. EECs, Paneth cells, tuft cells, and enterocytes release signals to activate Notch signaling, which modulates IESC activity and intestinal homeostasis, including proliferation and differentiation. Intestinal immunity can be altered via EECs, goblet cells, tuft cells, and cytokines derived from IECs. Finally, tools for investigating IEC communication have been discussed, including the novel 3D intestinal cell model utilizing enteroids that can be considered a powerful tool for IEC communication research. Overall, the importance of IEC communication, especially EECs and Paneth cells, which cover most intestinal functional regulating pathways, are overviewed in this paper. Such a compilation will be helpful in developing strategies for maintaining gut health.
Collapse
Affiliation(s)
| | | | - Yu-Chaun Liang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115201, Taiwan; (L.-E.Y.); (W.-C.Y.)
| |
Collapse
|
16
|
Jaber Y, Sarusi-Portuguez A, Netanely Y, Naamneh R, Yacoub S, Saar O, Darawshi N, Eli-Berchoer L, Shapiro H, Elinav E, Wilensky A, Hovav AH. Gingival spatial analysis reveals geographic immunological variation in a microbiota-dependent and -independent manner. NPJ Biofilms Microbiomes 2024; 10:142. [PMID: 39627243 PMCID: PMC11615284 DOI: 10.1038/s41522-024-00625-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
In mucosal barriers, tissue cells and leukocytes collaborate to form specialized niches that support host-microbiome symbiosis. Understanding the spatial organization of these barriers is crucial for elucidating the mechanisms underlying health and disease. The gingiva, a unique mucosal barrier with significant health implications, exhibits intricate tissue architecture and likely contains specialized immunological regions. Through spatial transcriptomic analysis, this study reveals distinct immunological characteristics between the buccal and palate regions of the murine gingiva, impacting natural alveolar bone loss. The microbiota primarily affects gingival immunity in the buccal region. Additionally, a significant influence of the microbiota on the junctional epithelium facing the oral biofilm offers new insights into neutrophil recruitment. The microbiota also regulates the proliferation and barrier-sealing function of the gingival epithelium. This underscores the presence of immunological niches in the gingiva, with the microbiota differentially influencing them, highlighting the high complexity of this oral mucosal barrier.
Collapse
Affiliation(s)
- Yasmin Jaber
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | | | - Yasmin Netanely
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Reem Naamneh
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Shahd Yacoub
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Or Saar
- Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel; Department of Periodontology, Hadassah Medical Center, Jerusalem, Israel
| | - Nadeem Darawshi
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Luba Eli-Berchoer
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Hagit Shapiro
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Microbiome & Cancer Division, DKFZ, Heidelberg, Germany
| | - Asaf Wilensky
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
17
|
Qu K, Shi M, Chen L, Liu Y, Yao X, Li X, Tan B, Xie S. Residual levels of dietary deltamethrin interfere with growth and intestinal health in Litopenaeus vannamei. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117376. [PMID: 39612679 DOI: 10.1016/j.ecoenv.2024.117376] [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: 05/05/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
To date, few study explored the damage of chronic dietary exposure to the lipophilic pesticide deltamethrin (DM) in aquatic animals, and it remains unclear whether its toxicity and residue levels would be affected by dietary lipid levels. Therefore, the present study aimed to elucidate the interactions between dietary lipid levels and DM levels in the Pacific white shrimp, focusing on growth performance, antioxidant capacity, and intestinal microbiota. DM has excellent insecticidal activity and has been used worldwide. Previous research has shown that environmental DM poses toxicity risks to aquatic animals. Six different diets were formulated to feed shrimp for 6 weeks with two lipid levels (6.96 %, 10.88 %) and three DM levels (0.2 mg·kg-1, 1 mg·kg-1, 5 mg·kg-1), namely LF0.2, LF1, LF5, HF0.2, HF1, HF5, respectively. Each diet was assigned to three net cages with a total of 18 cages (40 shrimp per tank, average weight (0.382±0.001 g), of which 0.2 mg·kg-1, are grouped in environmental DM control groups. The growth of shrimp was reduced as the dietary DM levels increased. When shrimp were fed a diet containing a high dose of DM, a reduction in their antioxidant capacity was also observed. Enzyme activity and gene expression related to lipid metabolism in hepatopancreas and hemolymph indicated a significant interaction between dietary lipid levels and DM in the lipid metabolism of shrimp. The terms of detoxification-related genes (gst, sult, cyp1a1) were upregulated in shrimp fed the high-dose DM. Additionally, the presence of DM in the diet severely harmed the hepatopancreas and intestinal histological morphology. DM in the diet increased the susceptibility of shrimp to pathogens and induced intestine microbiota dysbiosis, disrupting the balance of inter-species interactions. DM was not detected in the muscle and hepatopancreas of the shrimp after six weeks of exposure. In conclusion, the presence of DM in feed reduced the growth performance and antioxidant capacity of shrimp, damaging intestinal health. DM was rapidly metabolized by shrimp.
Collapse
Affiliation(s)
- Kangyuan Qu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Menglin Shi
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Liutong Chen
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yucheng Liu
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xinzhou Yao
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaoyue Li
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Beiping Tan
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China
| | - Shiwei Xie
- College of Aquatic Sciences, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Province Research Center for Accurate Nutrition and High-Efficiency Feeding of Aquatic Animals, Zhanjiang 524088, China; Key Laboratory of Aquatic Feed Science and Technology for Livestock and Poultry in Southern China, under the Ministry of Agriculture, Zhanjiang 524088, China.
| |
Collapse
|
18
|
Wang Y, Ding P, Zhang K, Xu X, Li H. Correlation Between Regulation of Intestinal Flora by Danggui-Shaoyao-San and Improvement of Cognitive Impairment in Mice With Alzheimer's Disease. Brain Behav 2024; 14:e70110. [PMID: 39482855 PMCID: PMC11527834 DOI: 10.1002/brb3.70110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 11/03/2024] Open
Abstract
PURPOSE The abnormal central glucose metabolism in Alzheimer's disease (AD) is related to the brain-gut axis. This study aims to explore the target of Danggui-Shaoyao-San (DSS) in improving cognitive impairment. METHOD This study analyzed the differences in mice intestinal flora by 16S rRNA sequencing. The cognitive protective effects of DSS were observed through the Morris water maze and the new object recognition. The mitigation effects of DSS on Aβ and p-tau, regulatory effects on glucose metabolism targets, and intestinal structure effects were observed through brain and colon slices staining. The differences in neural ultrastructure were compared by transmission electron microscopy. FINDING The results showed that DSS affected the composition of intestinal dominant bacteria and bacteria genera and regulated the abundance of intestinal bacteria in AD mice. DSS improved the behavior of AD mice, alleviated the deposition of AD pathological products in the brain and colon, regulated the expression of glycometabolism-related proteins, and improved the colon barrier structure and neural ultrastructure in the brain of mice with AD. CONCLUSION Our findings suggest that DSS may affect AD central glucose metabolism and improve cognition by regulating the gut-brain axis.
Collapse
Affiliation(s)
- Ya‐Han Wang
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - Peng‐Li Ding
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Kai‐Xin Zhang
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| | - Xiang‐Qing Xu
- Department of NeurologyAffiliated Hospital of Shandong University of Traditional Chinese MedicineJinanChina
| | - He Li
- The First Clinical Medical CollegeShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
19
|
Ataollahi Eshkoor S, Fanijavadi S. Dysbiosis-epigenetics-immune system interaction and ageing health problems. J Med Microbiol 2024; 73. [PMID: 39606883 DOI: 10.1099/jmm.0.001921] [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: 11/29/2024] Open
Abstract
Background. The growing interest in microbiota-epigenetics-immune system research stems from the understanding that microbiota, a group of micro-organisms colonized in the human body, can influence the gene expression through epigenetic mechanisms and interaction with the immune system. Epigenetics refers to changes in gene activity that are not caused by the alteration in the DNA sequence itself.Discussion. The clinical significance of this research lies in the potential to develop new therapies for diseases linked to the imbalance of these microbial species (dysbiosis), such as cancer and neurodegenerative diseases. The intricate interaction between microbiota and epigenetics involves the production of metabolites and signalling molecules that can impact our health by influencing immune responses, metabolism and inflammation. Understanding these interactions could lead to novel therapeutic strategies targeting microbiota-epigenetic pathways to improve health outcomes.Conclusion. In this context, we aim to review and emphasize the current knowledge and key concepts that link the microbiota to epigenetics and immune system function, exploring their relevance to the development and maintenance of homeostasis and susceptibility to different diseases later in life. We aim to elucidate key concepts concerning the interactions and potential effects among the human gut microbiota, epigenetics, the immune system and ageing diseases linked to dysbiosis.
Collapse
|
20
|
MORALES-CANO KL, RIVERA-ALAMILLO YC, OLIART-ROS RM, PEÑA-MONTES C. Modulation of the gut microbiota by dietary intervention with Acanthocereus tetragonus improves the health status of Wistar rats with metabolic syndrome. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 44:100-109. [PMID: 40171391 PMCID: PMC11957756 DOI: 10.12938/bmfh.2024-041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/15/2024] [Indexed: 04/03/2025]
Abstract
The gastrointestinal tract is an ecosystem with heterogeneous patterns, distributions, and environments, resulting in different microbial compositions in each gut segment. The relationship between diet and microbiota determines this heterogeneity. Consumption of diets high in fat and carbohydrates (HLHC) is associated with gut dysbiosis, low microbial diversity, and metabolic syndrome (MetS). Functional fiber consumption improves the profile and diversity of the gut microbiota (GM); it stimulates the production of short-chain fatty acids (SCFAs), which act as signaling molecules that maintain the gut barrier integrity and induce hormone synthesis that regulates satiety and glucose metabolism, reducing some MetS parameters. The effect of a dietary intervention with Acanthocereus tetragonus (At), a cactus rich in fiber, antioxidants, amino acids, and minerals traditionally consumed by the Mexican population, is reported here. For this purpose, Wistar rats were randomly divided into three study groups: a control (C) group, a MetS group, and an At-supplemented group. In the MetS and At groups, an HLHC was administered for 12 weeks, inducing MetS. After 18 weeks, stool samples were collected for microbiota sequencing. HLHC administration favored Firmicutes and decreased the abundance of Bacteriodetes at the phylum level in the MetS group. At the genus level, the dietary intervention with At increased the presence of Roseburia, Ruminococcus, Blautia, Bacteroides, and Christensenella, reflecting the effect of A. tetragonus consumption on GM. At diet administration reduced body weight; the plasma glucose, insulin, and lipid levels; and insulin resistance.
Collapse
Affiliation(s)
- Karla Lizzeth MORALES-CANO
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Yokebed Cecilia RIVERA-ALAMILLO
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Rosa Maria OLIART-ROS
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| | - Carolina PEÑA-MONTES
- National Technological Institute of Mexico Campus Veracruz,
Miguel Ángel de Quevedo 2779, Formando Hogar, Veracruz, Ver. CP 91897, Mexico
| |
Collapse
|
21
|
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
|
22
|
Pieszka M, Szczepanik K, Łoniewski I. Utilizing pigs as a model for studying intestinal barrier function. ANNALS OF ANIMAL SCIENCE 2024. [DOI: 10.2478/aoas-2024-0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Intestinal permeability has been extensively studied, particularly in gastrointestinal diseases such as inflammatory bowel disease, food allergy, visceral disease, celiac disease, and Crohn’s disease. These studies have established that changes in intestinal permeability contribute to the pathogenesis of many gastrointestinal and systemic diseases. While numerous works in the 20th century focused on this topic, it remains relevant for several reasons. Despite the development of new research techniques, it is still unclear whether changes in intestinal permeability are the primary mechanism initiating the disease process or if they occur secondary to an ongoing chronic inflammatory process. Investigating the possibility of stabilizing the intestinal barrier, thereby reducing its permeability preemptively to prevent damage and after the damage has occurred, may offer new therapeutic approaches. Increased intestinal permeability is believed to lead to reduced nutrient absorption, resulting in decreased immunity and production of digestive enzymes.
Collapse
Affiliation(s)
- Marek Pieszka
- Department of Animal Nutrition and Feed Sciences , National Research Institute of Animal Production , Balice , Poland
| | - Kinga Szczepanik
- Department of Animal Nutrition and Feed Sciences , National Research Institute of Animal Production , Balice , Poland
| | - Igor Łoniewski
- Sanprobi sp. z o.o. sp. k ., Kurza Stopka 5/C , Szczecin , Poland
- Department of Biochemical Science , Pomeranian Medical University in Szczecin , Szczecin , Poland
| |
Collapse
|
23
|
Guo Y, Ren C, He Y, Wu Y, Yang X. Deciphering the spatiotemporal transcriptional landscape of intestinal diseases (Review). Mol Med Rep 2024; 30:157. [PMID: 38994768 PMCID: PMC11258600 DOI: 10.3892/mmr.2024.13281] [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: 11/21/2023] [Accepted: 04/19/2024] [Indexed: 07/13/2024] Open
Abstract
The intestines are the largest barrier organ in the human body. The intestinal barrier plays a crucial role in maintaining the balance of the intestinal environment and protecting the intestines from harmful bacterial invasion. Single‑cell RNA sequencing technology allows the detection of the different cell types in the intestine in two dimensions and the exploration of cell types that have not been fully characterized. The intestinal mucosa is highly complex in structure, and its proper functioning is linked to multiple structures in the proximal‑distal intestinal and luminal‑mucosal axes. Spatial localization is at the core of the efforts to explore the interactions between the complex structures. Spatial transcriptomics (ST) is a method that allows for comprehensive tissue analysis and the acquisition of spatially separated genetic information from individual cells, while preserving their spatial location and interactions. This approach also prevents the loss of fragile cells during tissue disaggregation. The emergence of ST technology allows us to spatially dissect enzymatic processes and interactions between multiple cells, genes, proteins and signals in the intestine. This includes the exchange of oxygen and nutrients in the intestine, different gradients of microbial populations and the role of extracellular matrix proteins. This regionally precise approach to tissue studies is gaining more acceptance and is increasingly applied in the investigation of disease mechanisms related to the gastrointestinal tract. Therefore, this review summarized the application of ST in gastrointestinal diseases.
Collapse
Affiliation(s)
- Yajing Guo
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Chao Ren
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yuxi He
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Yue Wu
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| | - Xiaojun Yang
- Department of Digestive Medicine, Chongqing City Hospital of Traditional Chinese Medicine, Chongqing 400021, P.R. China
| |
Collapse
|
24
|
Van Bockstal L, Prims S, Van Cruchten S, Ayuso M, Che L, Van Ginneken C. Cell migration and proliferation capacity of IPEC-J2 cells after short-chain fatty acid exposure. PLoS One 2024; 19:e0309742. [PMID: 39213333 PMCID: PMC11364292 DOI: 10.1371/journal.pone.0309742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Novel antimicrobial strategies are necessary to tackle using antibiotics during the suckling and weaning period of piglets, often characterized by E. coli-induced diarrhea. In the last decades, acetate, propionate, and butyrate, all short-chain fatty acids (SCFAs), have been proposed as an alternative to antibiotics. SCFAs are instrumental in promoting the proliferation of enterocytes, preserving intestinal integrity, and modulating the microbial community by suppressing the growth of pathogenic bacteria in pigs. The effect of individual SCFAs (proprionate, acetate and butyrate) on the regenerative capacity of intestinal cells was investigated via an optimized wound-healing assay in IPEC-J2 cells, a porcine jejunal epithelial cell line. IPEC-J2 cells proved a good model as they express the free fatty acid receptor 2 (FFAR2), an important SCFA receptor with a high affinity for proprionate. Our study demonstrated that propionate (p = 0.005) and acetate (p = 0.037) were more effective in closing the wound than butyrate (p = 0.190). This holds promise in using SCFA's per os as an alternative to antibiotics.
Collapse
Affiliation(s)
- Lieselotte Van Bockstal
- Comparative Perinatal Development, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sara Prims
- Comparative Perinatal Development, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Steven Van Cruchten
- Comparative Perinatal Development, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Miriam Ayuso
- Biogenesis Bagó, Development of Biotech Products, Madrid, Spain
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu City, Sichuan Province, China
| | - Chris Van Ginneken
- Comparative Perinatal Development, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
25
|
Ullah H, Arbab S, Tian Y, Chen Y, Liu CQ, Li Q, Li K. Crosstalk between gut microbiota and host immune system and its response to traumatic injury. Front Immunol 2024; 15:1413485. [PMID: 39144142 PMCID: PMC11321976 DOI: 10.3389/fimmu.2024.1413485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024] Open
Abstract
Millions of microorganisms make up the complex microbial ecosystem found in the human gut. The immune system's interaction with the gut microbiota is essential for preventing inflammation and maintaining intestinal homeostasis. Numerous metabolic products that can cross-talk between immune cells and the gut epithelium are metabolized by the gut microbiota. Traumatic injury elicits a great and multifaceted immune response in the minutes after the initial offense, containing simultaneous pro- and anti-inflammatory responses. The development of innovative therapies that improve patient outcomes depends on the gut microbiota and immunological responses to trauma. The altered makeup of gut microbes, or gut dysbiosis, can also dysregulate immunological responses, resulting in inflammation. Major human diseases may become more common as a result of chronic dysbiosis and the translocation of bacteria and the products of their metabolism beyond the mucosal barrier. In this review, we briefly summarize the interactions between the gut microbiota and the immune system and human disease and their therapeutic probiotic formulations. We also discuss the immune response to traumatic injury.
Collapse
Affiliation(s)
- Hanif Ullah
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Safia Arbab
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yali Tian
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Chang-qing Liu
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Qijie Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials/Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Haddad MJ, Zuluaga-Arango J, Mathieu H, Barbezier N, Anton PM. Intestinal Epithelial Co-Culture Sensitivity to Pro-Inflammatory Stimuli and Polyphenols Is Medium-Independent. Int J Mol Sci 2024; 25:7360. [PMID: 39000465 PMCID: PMC11242137 DOI: 10.3390/ijms25137360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/23/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
The complexification of in vitro models requires the compatibility of cells with the same medium. Since immune cells are the most sensitive to growth conditions, growing intestinal epithelial cells in their usual medium seems to be necessary. This work was aimed at comparing the sensitivity of these epithelial cells to pro-inflammatory stimuli but also to dietary polyphenols in both DMEM and RPMI-1640 media. Co-cultures of Caco-2 and HT29-MTX cells were grown for 21 days in the two media before their stimulation with a cocktail of TNF-α (20 ng/mL), IL-1β (1 ng/mL), and IFN-γ (10 ng/mL) or with LPS (10 ng/mL) from E. coli (O111:B4). The role of catechins (15 µM), a dietary polyphenol, was evaluated after its incubation with the cells before their stimulation for 6 h. The RPMI-1640 medium did not alter the intensity of the inflammatory response observed with the cytokines. By contrast, LPS failed to stimulate the co-culture in inserts regardless of the medium used. Lastly, catechins were unable to prevent the pro-inflammatory response observed with the cytokines in the two media. The preservation of the response of this model of intestinal epithelium in RPMI-1640 medium is promising when considering its complexification to evaluate the complex cellular crosstalk leading to intestinal homeostasis.
Collapse
Affiliation(s)
- Michelle J Haddad
- Transformations et Agroressources, ULR 7519, Institut Polytechnique UniLaSalle, Université d'Artois, 60000 Beauvais, France
- HCS Pharma, 59120 Loos, France
| | - Juanita Zuluaga-Arango
- Transformations et Agroressources, ULR 7519, Institut Polytechnique UniLaSalle, Université d'Artois, 60000 Beauvais, France
| | - Hugo Mathieu
- Transformations et Agroressources, ULR 7519, Institut Polytechnique UniLaSalle, Université d'Artois, 60000 Beauvais, France
| | - Nicolas Barbezier
- Transformations et Agroressources, ULR 7519, Institut Polytechnique UniLaSalle, Université d'Artois, 60000 Beauvais, France
| | - Pauline M Anton
- Transformations et Agroressources, ULR 7519, Institut Polytechnique UniLaSalle, Université d'Artois, 60000 Beauvais, France
| |
Collapse
|
27
|
Shi F, Ma L, Chen Z, Huang Y, Lin L, Qin Z. Long-term disinfectant exposure on intestinal immunity and microbiome variation of grass carp. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 272:106942. [PMID: 38788458 DOI: 10.1016/j.aquatox.2024.106942] [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: 10/25/2023] [Revised: 04/19/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024]
Abstract
The gut microbiome is crucial in maintaining fish health homeostasis. Disinfectants can kill important pathogens and disinfect fish eggs, yet their effect on the immune pathways and intestinal microbiome in healthy fish remains unknown. In this study, we investigated the effects of two disinfectants on the transcriptome profiles, immunological response, and gut microbiota dynamics of grass carp over a four-week trial. In particular, aquatic water was disinfected with 80 μg/L glutaraldehyde or 50 μg/L povidone-iodine. We found that glutaraldehyde and povidone-iodine induced gut antioxidant system and depressed the function of grass carp digestive enzymes. The results of the 16S rDNA high-throughput sequencing identified a reduction in the diversity of grass carp gut microbiota following the disinfectant treatment. Moreover, transcriptome profiling revealed that disinfectant exposure altered the immune-related pathways of grass carp and inhibited the expression of inflammation and tight junction related genes. Finally, the histopathological observation and apoptosis detection results suggested that the long-term diet of disinfectant destroyed intestinal structural integrity and promoted apoptosis. In conclusion, long-term exposure to disinfectants was observed to reduce oxidation resistance, suppress the immune response, dysbiosis of the intestinal flora, and resulted in increasing the apoptosis in intestinal of grass carp.
Collapse
Affiliation(s)
- Fei Shi
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Lixin Ma
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Zhilong Chen
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Yao Huang
- The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Li Lin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China.
| | - Zhendong Qin
- Guangdong Provincial Water Environment and Aquatic Products Security Engineering Technology Research Center, Guangzhou Key Laboratory of Aquatic Animal Diseases and Waterfowl Breeding, College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China.
| |
Collapse
|
28
|
Diez-Martin E, Hernandez-Suarez L, Muñoz-Villafranca C, Martin-Souto L, Astigarraga E, Ramirez-Garcia A, Barreda-Gómez G. Inflammatory Bowel Disease: A Comprehensive Analysis of Molecular Bases, Predictive Biomarkers, Diagnostic Methods, and Therapeutic Options. Int J Mol Sci 2024; 25:7062. [PMID: 39000169 PMCID: PMC11241012 DOI: 10.3390/ijms25137062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
In inflammatory bowel diseases (IBDs), such as Crohn's disease (CD) and ulcerative colitis (UC), the immune system relentlessly attacks intestinal cells, causing recurrent tissue damage over the lifetime of patients. The etiology of IBD is complex and multifactorial, involving environmental, microbiota, genetic, and immunological factors that alter the molecular basis of the organism. Among these, the microbiota and immune cells play pivotal roles; the microbiota generates antigens recognized by immune cells and antibodies, while autoantibodies target and attack the intestinal membrane, exacerbating inflammation and tissue damage. Given the altered molecular framework, the analysis of multiple molecular biomarkers in patients proves exceedingly valuable for diagnosing and prognosing IBD, including markers like C reactive protein and fecal calprotectin. Upon detection and classification of patients, specific treatments are administered, ranging from conventional drugs to new biological therapies, such as antibodies to neutralize inflammatory molecules like tumor necrosis factor (TNF) and integrin. This review delves into the molecular basis and targets, biomarkers, treatment options, monitoring techniques, and, ultimately, current challenges in IBD management.
Collapse
Affiliation(s)
- Eguzkiñe Diez-Martin
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Leidi Hernandez-Suarez
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Carmen Muñoz-Villafranca
- Department of Gastroenterology, University Hospital of Basurto, Avda Montevideo 18, 48013 Bilbao, Spain
| | - Leire Martin-Souto
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Egoitz Astigarraga
- Research and Development Department, IMG Pharma Biotech S.L., 48170 Zamudio, Spain
| | - Andoni Ramirez-Garcia
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | | |
Collapse
|
29
|
Yao Z, Bai R, Liu W, Liu Y, Zhou W, Xu Z, Sheng J. Activation of angiogenin expression in macrophages by lipopolysaccharide via the TLR4/NF-κB pathway in colitis. Acta Biochim Biophys Sin (Shanghai) 2024; 56:857-865. [PMID: 38567413 PMCID: PMC11214953 DOI: 10.3724/abbs.2024013] [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: 11/29/2023] [Accepted: 01/10/2024] [Indexed: 04/04/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a debilitating condition that can lead to life-threatening complications. Macrophages are crucial in IBD management because they secrete various cytokines and regulate tissue repair. Macrophage-derived angiogenin (ANG) has been shown to be essential for limiting colonic inflammation, but its upstream regulatory pathway and role in macrophages remain unclear. Here we show that ANG expression is up-regulated in macrophages during colitis treatment or upon lipopolysaccharides (LPS) treatment. Mechanistically, LPS activates Toll-like receptor 4 (TLR4) to initiate NF-κB translocation from the cytoplasm to the nucleus, where it binds to the ANG promoter and enhances its transcriptional activity, leading to increased ANG expression. Interestingly, our data also reveal that the deletion of ANG in macrophages has no adverse effect on key macrophage functions, such as phagocytosis, chemotaxis, and cell survival. Our findings establish a "LPS-TLR4-NF-κB-ANG" regulatory axis in inflammatory disorders and confirm that ANG controls inflammation in a paracrine manner, highlighting the importance of ANG as a key mediator in the complex network of inflammatory processes.
Collapse
Affiliation(s)
- Zhengrong Yao
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Rongpan Bai
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Wei Liu
- Department of General SurgerySir Run Run Shaw Hospital.Zhejiang University School of MedicineHangzhou310016China
| | - Yaxing Liu
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| | - Wei Zhou
- Department of General SurgerySir Run Run Shaw Hospital.Zhejiang University School of MedicineHangzhou310016China
| | - Zhengping Xu
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Cancer CenterZhejiang UniversityHangzhou310012China
- Zhejiang Provincial Key Laboratory of BioelectromagneticsHangzhou310058China
| | - Jinghao Sheng
- Institute of Environmental Medicine and Department of General SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Cancer CenterZhejiang UniversityHangzhou310012China
- Zhejiang Provincial Key Laboratory of BioelectromagneticsHangzhou310058China
| |
Collapse
|
30
|
Che S, Yan Z, Feng Y, Zhao H. Unveiling the intratumoral microbiota within cancer landscapes. iScience 2024; 27:109893. [PMID: 38799560 PMCID: PMC11126819 DOI: 10.1016/j.isci.2024.109893] [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] [Indexed: 05/29/2024] Open
Abstract
Recent advances in cancer research have unveiled a significant yet previously underappreciated aspect of oncology: the presence and role of intratumoral microbiota. These microbial residents, encompassing bacteria, fungi, and viruses within tumor tissues, have been found to exert considerable influence on tumor development, progression, and the efficacy of therapeutic interventions. This review aims to synthesize these groundbreaking discoveries, providing an integrated overview of the identification, characterization, and functional roles of intratumoral microbiota in cancer biology. We focus on elucidating the complex interactions between these microorganisms and the tumor microenvironment, highlighting their potential as novel biomarkers and therapeutic targets. The purpose of this review is to offer a comprehensive understanding of the microbial dimension in cancer, paving the way for innovative approaches in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Shusheng Che
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266005, Shandong, China
| | - Zhiyong Yan
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266005, Shandong, China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266005, Shandong, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266005, Shandong, China
| |
Collapse
|
31
|
Chen T, Aly RSS, Shen Y, Tang S, Zhao Y, Zhao J, Chen X. The silent threat: Nanopolystyrene and chrysene pollutants disrupt the intestinal mucosal barrier, new insights from juvenile Siniperca chuatsi. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 926:172001. [PMID: 38552987 DOI: 10.1016/j.scitotenv.2024.172001] [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: 11/20/2023] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024]
Abstract
The intestinal mucosal barrier-comprising microbial, mechanical, chemical, and immunological barriers-is critical to protection against pathogens and maintenance of host health; however, it remains unclear whether it is affected by environmental contaminants. Therefore, the present study assessed whether exposure to ambient concentrations of nanopolystyrene (NP) and chrysene (CHR)-two ubiquitous environmental pollutants in the aquatic environment-affect the intestinal mucosal barrier in juvenile Siniperca chuatsi. After exposure for 21 days, S. chuatsi exhibited intestinal oxidative stress and imbalance of intestinal microbial homeostasis. NP and/or CHR exposure also disrupted the intestinal mechanical barrier, as evidenced by the altered intestinal epithelial cell morphology, disrupted structure of intercellular tight junctions, and decreased expression of tight junction proteins. Damage to the intestinal chemical barrier manifested as thinning of the mucus layer owing to the loss and damage of goblet cells. Furthermore, the intestinal immunological barrier was impaired as indicated by the loss of intestinal intraepithelial lymphocytes and increase in pro-inflammatory cytokines, chemokines, and immunoglobulins. These findings collectively suggest that the intestinal mucosal barrier was damaged. This study is, to the best of our knowledge, the first to report that exposure to NP and/or CHR at environmentally relevant concentrations disrupts the intestinal mucosal barrier in organisms and highlight the significance of nanoplastic/CHR pollution for intestinal health.
Collapse
Affiliation(s)
- Tiantian Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Rahma Sakina Said Aly
- Centre for Research on Environmental Ecology and Fish Nutrition of the Ministry of Agriculture, Shanghai Ocean University, Shanghai 201306, China
| | - Yawei Shen
- College of Fisheries, Henan Normal University, Xinxiang 453007, Henan, China
| | - Shoujie Tang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Yan Zhao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Jinliang Zhao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
| | - Xiaowu Chen
- Centre for Research on Environmental Ecology and Fish Nutrition of the Ministry of Agriculture, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
32
|
Wang J, He M, Yang M, Ai X. Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci 2024; 345:122612. [PMID: 38588949 DOI: 10.1016/j.lfs.2024.122612] [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: 02/04/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Gut microbiota is a complex microbial community with the ability of maintaining intestinal health. Intestinal homeostasis largely depends on the mucosal immune system to defense external pathogens and promote tissue repair. In recent years, growing evidence revealed the importance of gut microbiota in shaping intestinal mucosal immunity. Therefore, according to the existing findings, this review first provided an overview of intestinal mucosal immune system before summarizing the regulatory roles of gut microbiota in intestinal innate and adaptive immunity. Specifically, this review delved into the gut microbial interactions with the cells such as intestinal epithelial cells (IECs), macrophages, dendritic cells (DCs), neutrophils, and innate lymphoid cells (ILCs) in innate immunity, and T and B lymphocytes in adaptive immunity. Furthermore, this review discussed the main effects of gut microbiota dysbiosis in intestinal diseases and offered future research prospects. The review highlighted the key regulatory roles of gut microbiota in intestinal mucosal immunity via various host-microbe interactions, providing valuable references for the development of microbial therapy in intestinal diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Mei He
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
33
|
Ambrogi M, Vezina CM. Roles of airway and intestinal epithelia in responding to pathogens and maintaining tissue homeostasis. Front Cell Infect Microbiol 2024; 14:1346087. [PMID: 38736751 PMCID: PMC11082347 DOI: 10.3389/fcimb.2024.1346087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/10/2024] [Indexed: 05/14/2024] Open
Abstract
Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.
Collapse
Affiliation(s)
| | - Chad M. Vezina
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
34
|
Shin Y, Kim J, Song Y, Kim S, Kong H. Efficacy of Laurus nobilis L. for Tight Junction Protein Imbalance in Leaky Gut Syndrome. Nutrients 2024; 16:1250. [PMID: 38732497 PMCID: PMC11085348 DOI: 10.3390/nu16091250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Laurus nobilis L. (LNL) belongs to the evergreen Lauraceae family. It is native to the Mediterranean and widely distributed in the southern United States, Europe, and the Middle East. LNL is rich in active ingredients of the sesquiterpene lactone series and has been reported to have antioxidant, anti-inflammatory, and anticancer effects. And parthenolide, known as a sesquiterpene lactone-based compound, inhibits the activation of lipopolysaccharide-binding protein (LBP), which is a major trigger for leaky gut syndrome. However, the effectiveness of LNL in improving the state of increased intestinal permeability has not yet been reported. Therefore, we demonstrated the efficacy of LNL, which is known to be rich in parthenolide, in improving intestinal permeability induced by IL-13. We investigated the improvement in permeability and analyzed major tight junction proteins (TJs), permeability-related mechanisms, weight and disease activity indices, and corresponding cytokine mechanisms. LNL maintained TJs homeostasis and clinical improvement by reducing increased claudin-2 through the inhibition of IL-13/STAT6 activation in TJ-damaged conditions. These results are expected to be effective in preventing leaky gut syndrome through the TJ balance and to further improve intestinal-related diseases, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Yelim Shin
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea; (Y.S.); (J.K.); (Y.S.); (S.K.)
| | - Jiyeon Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea; (Y.S.); (J.K.); (Y.S.); (S.K.)
- KOSA BIO Inc., 272, Namyangju-si 12106, Republic of Korea
| | - Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea; (Y.S.); (J.K.); (Y.S.); (S.K.)
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
| | - Sangbum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea; (Y.S.); (J.K.); (Y.S.); (S.K.)
| | - Hyunseok Kong
- PADAM Natural Material Research Institute, Sahmyook University, Seoul 01795, Republic of Korea
- College of Animal Resources Science, Seoul 01795, Republic of Korea
| |
Collapse
|
35
|
Ashkenazi-Preiser H, Reuven O, Uzan-Yulzari A, Komisarov S, Cirkin R, Turjeman S, Even C, Twaik N, Ben-Meir K, Mikula I, Cohen-Daniel L, Meirow Y, Pikarsky E, Louzoun Y, Koren O, Baniyash M. The Cross-talk Between Intestinal Microbiota and MDSCs Fuels Colitis-associated Cancer Development. CANCER RESEARCH COMMUNICATIONS 2024; 4:1063-1081. [PMID: 38506672 PMCID: PMC11017962 DOI: 10.1158/2767-9764.crc-23-0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/24/2023] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
Intestinal chronic inflammation is associated with microbial dysbiosis and accumulation of various immune cells including myeloid-derived suppressor cells (MDSC), which profoundly impact the immune microenvironment, perturb homeostasis and increase the risk to develop colitis-associated colorectal cancer (CAC). However, the specific MDSCs-dysbiotic microbiota interactions and their collective impact on CAC development remain poorly understood. In this study, using a murine model of CAC, we demonstrate that CAC-bearing mice exhibit significantly elevated levels of highly immunosuppressive MDSCs, accompanied by microbiota alterations. Both MDSCs and bacteria that infiltrate the colon tissue and developing tumors can be found in close proximity, suggesting intricate MDSC-microbiota cross-talk within the tumor microenvironment. To investigate this phenomenon, we employed antibiotic treatment to disrupt MDSC-microbiota interactions. This intervention yielded a remarkable reduction in intestinal inflammation, decreased MDSC levels, and alleviated immunosuppression, all of which were associated with a significant reduction in tumor burden. Furthermore, we underscore the causative role of dysbiotic microbiota in the predisposition toward tumor development, highlighting their potential as biomarkers for predicting tumor load. We shed light on the intimate MDSCs-microbiota cross-talk, revealing how bacteria enhance MDSC suppressive features and activities, inhibit their differentiation into mature beneficial myeloid cells, and redirect some toward M2 macrophage phenotype. Collectively, this study uncovers the role of MDSC-bacteria cross-talk in impairing immune responses and promoting tumor growth, providing new insights into potential therapeutic strategies for CAC. SIGNIFICANCE MDSCs-dysbiotic bacteria interactions in the intestine play a crucial role in intensifying immunosuppression within the CAC microenvironment, ultimately facilitating tumor growth, highlighting potential therapeutic targets for improving the treatment outcomes of CAC.
Collapse
Affiliation(s)
- Hadas Ashkenazi-Preiser
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Or Reuven
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | | | - Sharon Komisarov
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Roy Cirkin
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Carmel Even
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Nira Twaik
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Kerem Ben-Meir
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Ivan Mikula
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Leonor Cohen-Daniel
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Yaron Meirow
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Eli Pikarsky
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| | - Yoram Louzoun
- Department of mathematics, Bar-Ilan University, Ramat Gan, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Michal Baniyash
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, POB 12272, Jerusalem 91120, Israel
| |
Collapse
|
36
|
Cicchinelli S, Gemma S, Pignataro G, Piccioni A, Ojetti V, Gasbarrini A, Franceschi F, Candelli M. Intestinal Fibrogenesis in Inflammatory Bowel Diseases: Exploring the Potential Role of Gut Microbiota Metabolites as Modulators. Pharmaceuticals (Basel) 2024; 17:490. [PMID: 38675450 PMCID: PMC11053610 DOI: 10.3390/ph17040490] [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: 03/04/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Fibrosis, sustained by the transformation of intestinal epithelial cells into fibroblasts (epithelial-to-mesenchymal transition, EMT), has been extensively studied in recent decades, with the molecular basis well-documented in various diseases, including inflammatory bowel diseases (IBDs). However, the factors influencing these pathways remain unclear. In recent years, the role of the gut microbiota in health and disease has garnered significant attention. Evidence suggests that an imbalanced or dysregulated microbiota, along with environmental and genetic factors, may contribute to the development of IBDs. Notably, microbes produce various metabolites that interact with host receptors and associated signaling pathways, influencing physiological and pathological changes. This review aims to present recent evidence highlighting the emerging role of the most studied metabolites as potential modulators of molecular pathways implicated in intestinal fibrosis and EMT in IBDs. These studies provide a deeper understanding of intestinal inflammation and fibrosis, elucidating the molecular basis of the microbiota role in IBDs, paving the way for future treatments.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
37
|
Kaur H, Kaur G, Ali SA. Postbiotics Implication in the Microbiota-Host Intestinal Epithelial Cells Mutualism. Probiotics Antimicrob Proteins 2024; 16:443-458. [PMID: 36933160 DOI: 10.1007/s12602-023-10062-w] [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: 03/08/2023] [Indexed: 03/19/2023]
Abstract
To sustain host health and provide the microbial community with a nutrient-rich environment, the host and gut microbiota must interact with one another. These interactions between commensal bacterial and intestinal epithelial cells (IECs) serve as the first line of defense against gut microbiota in preserving intestinal homeostasis. In this microenvironment, the post-biotics and similar molecules such as p40 exert several beneficial effects through regulation of IECs. Importantly, post-biotics were discovered to be transactivators of the EGF receptor (EGFR) in IECs, inducing protective cellular responses and alleviating colitis. The transient exposure to post-biotics such as p40 during the neonatal period reprograms IECs by upregulation of a methyltransferase, Setd1β, leading to a sustained increase in TGF- β release for the expansion of regulatory T cells (Tregs) in the intestinal lamina propria and durable protection against colitis in adulthood. This crosstalk between the IECs and post-biotic secreted factors was not reviewed previously. Therefore, this review describes the role of probiotic-derived factors in the sustainability of intestinal health and improving gut homeostasis via certain signaling pathways. In the era of precision medicine and targeted therapies, more basic, preclinical, and clinical evidence is needed to clarify the efficacy of probiotics released as functional factors in maintaining intestinal health and preventing and treating disease.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, Karnal, 132001, India
| | - Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
- Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal, 132001, India.
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, 69120, Germany.
| |
Collapse
|
38
|
Didriksen BJ, Eshleman EM, Alenghat T. Epithelial regulation of microbiota-immune cell dynamics. Mucosal Immunol 2024; 17:303-313. [PMID: 38428738 PMCID: PMC11412483 DOI: 10.1016/j.mucimm.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
The mammalian gastrointestinal tract hosts a diverse community of trillions of microorganisms, collectively termed the microbiota, which play a fundamental role in regulating tissue physiology and immunity. Recent studies have sought to dissect the cellular and molecular mechanisms mediating communication between the microbiota and host immune system. Epithelial cells line the intestine and form an initial barrier separating the microbiota from underlying immune cells, and disruption of epithelial function has been associated with various conditions ranging from infection to inflammatory bowel diseases and cancer. From several studies, it is now clear that epithelial cells integrate signals from commensal microbes. Importantly, these non-hematopoietic cells also direct regulatory mechanisms that instruct the recruitment and function of microbiota-sensitive immune cells. In this review, we discuss the central role that has emerged for epithelial cells in orchestrating intestinal immunity and highlight epithelial pathways through which the microbiota can calibrate tissue-intrinsic immune responses.
Collapse
Affiliation(s)
- Bailey J Didriksen
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Emily M Eshleman
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| | - Theresa Alenghat
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.
| |
Collapse
|
39
|
Xi Y, Yu M, Li X, Zeng X, Li J. The coming future: The role of the oral-microbiota-brain axis in aroma release and perception. Compr Rev Food Sci Food Saf 2024; 23:e13303. [PMID: 38343293 DOI: 10.1111/1541-4337.13303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
The field of aroma release and perception during the oral process has been well studied. However, the traditional approaches have not fully explored the integration of oral biology, microbiology, and neurology to further understand aroma release and perception mechanisms. Herein, to address the existing challenges in this field, we introduce the oral-microbiota-brain axis (OMBA), an innovative framework that encapsulates the interactive relationships among saliva and the oral mucosa, the oral microbiota, and the brain in aroma release and perception. This review introduces the OMBA and highlights its role as a key interface facilitating the sensory experience of aroma. Based on a comprehensive literature survey, the specific roles of the oral mucosa, oral microbiota, saliva, and brain in the OMBA are discussed. This integrated approach reveals the importance of each component and the interconnected relationships within this axis in the overall process of aroma release and perception. Saliva and the oral mucosa play fundamental roles in aroma release and perception; the oral microbiota regulates aroma release and impacts olfactory perception; and the brain's intricate neural circuitry is central to the decoding and interpretation of aroma signals. The components of this axis are interdependent, and imbalances can disrupt aroma perception. The OMBA framework not only enhances our comprehension of aroma release and perception but also paves the way for innovative applications that could heighten sensory experiences.
Collapse
Affiliation(s)
- Yu Xi
- Laboratory of Green and Low-carbon Processing Technology for Plant-based Food of China National Light Industry Council, and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Meihong Yu
- Laboratory of Green and Low-carbon Processing Technology for Plant-based Food of China National Light Industry Council, and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Xuejie Li
- Laboratory of Green and Low-carbon Processing Technology for Plant-based Food of China National Light Industry Council, and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Xiangquan Zeng
- Laboratory of Green and Low-carbon Processing Technology for Plant-based Food of China National Light Industry Council, and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| | - Jian Li
- Laboratory of Green and Low-carbon Processing Technology for Plant-based Food of China National Light Industry Council, and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
40
|
Gu Q, Xia H, Song YQ, Duan J, Chen Y, Zhang Y, Chen HP, Zhang L. SLC6A14 promotes ulcerative colitis progression by facilitating NLRP3 inflammasome-mediated pyroptosis. World J Gastroenterol 2024; 30:252-267. [PMID: 38314135 PMCID: PMC10835529 DOI: 10.3748/wjg.v30.i3.252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/08/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory condition with frequent relapse and recurrence. Evidence suggests the involvement of SLC6A14 in UC pathogenesis, but the central regulator remains unknown. AIM To explore the role of SLC6A14 in UC-associated pyroptosis. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR), immunoblotting, and immunohistochemical were used to assess SLC6A14 in human UC tissues. Lipopolysaccharide (LPS) was used to induce inflammation in FHC and NCM460 cells and model enteritis, and SLC6A14 levels were assessed. Pyroptosis markers were quantified using enzyme-linked immunosorbent assay, Western blotting, and qRT-PCR, and EdU incubation, CCK-8 assays and flow cytometry were used to examine proliferation and apoptosis. Mouse models of UC were used for verification. RESULTS SLC6A14 was increased and correlated with NLRP3 in UC tissues. LPS-induced FHC and NCM460 cells showed increased SLC6A14 levels. Reducing SLC6A14 increased cell proliferation and suppressed apoptosis. Reducing SLC6A14 decreased pyroptosis-associated proteins (ASC, IL-1β, IL-18, NLRP3). NLRP3 overexpression counteracted the effects of sh-SLC6A14 on LPS-induced FHC and NCM460 cell pyroptosis. SLC6A14 improved the mucosa in mice with dextran sulfate sodium-induced colitis. CONCLUSION SLC6A14 promotes UC pyroptosis by regulating NLRP3, suggesting the therapeutic potential of modulating the SLC6A14/NLRP3 axis.
Collapse
Affiliation(s)
- Qing Gu
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
| | - Huan Xia
- Geriatrics Research Institute, Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| | - Yue-Qiong Song
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
| | - Jun Duan
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
| | - Yun Chen
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
| | - You Zhang
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
| | - He-Ping Chen
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
- Geriatrics Research Institute, Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| | - Li Zhang
- Department of Elderly Digestive, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu 610072, Sichuan Province, China
- Geriatrics Research Institute, Sichuan Provincial People’s Hospital, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
41
|
Zhang J, Zhao Q, Hu Z. Clinical predictive value of the initial neutrophils to lymphocytes and platelets ratio for prognosis of sepsis patients in the intensive care unit: a retrospective study. Front Med (Lausanne) 2024; 11:1351492. [PMID: 38318247 PMCID: PMC10840849 DOI: 10.3389/fmed.2024.1351492] [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: 12/07/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
Background Neutrophils to lymphocytes and platelets (N/LP) ratio has been confirmed as an indirect marker of inflammation. In this study, we aimed to further evaluate the prognostic significance of the N/LP ratio in sepsis patients admitted to the ICU. Methods Sepsis patients from the Affiliated Hospital of Jiangsu University were retrospectively enrolled from January 2015 and July 2023. The primary outcomes were 30/60 days mortality. The secondary outcomes included the incidence of AKI, vasoactive drug, CRRT, invasive ventilation, length of ICU stay, length of hospital stay and ICU mortality. Results A total of 1,066 sepsis patients were enrolled with a median age of 75.0 (66.0, 85.0) years, and 62.5% of them being male. The 30 days and 60 days mortality rates were found to be 28.7 and 34.0%, respectively, while the incidence of AKI was 45.2%. Based on their N/LP ratios, we classified the sepsis patients into three groups: low, middle, and high, consisting of 266, 534, and 266 patients, respectively. According the Cox proportional hazard model, the middle and high N/LP groups were associated with a 1.990/3.106-fold increase in 30 days mortality risk and a 2.066/3.046-fold increase in 60 days mortality risk compared with the low N/LP group. Besides, multivariate logistic regression model suggested that the risk of AKI occurrence increased 2.460 fold in the high group compared to the low group. However, through subgroup analyses, we observed substantial variations in the association between N/LP ratios and 30/60 days mortality rates as well as the incidence of AKI among different populations. Notably, the N/LP ratio measured at ICU admission exhibited a higher AUC for predicting 30/60 days mortality (0.684/0.687). Additionally, we observed a good predictive power for the occurrence of AKI (AUC: 0.645) using the N/LP ratio measured at sepsis prognosis. Regarding the other secondary outcomes, the N/LP ratio was associated with disease severity in sepsis patients, including the need for vasoactive drugs, length of ICU stay, and ICU mortality. Conclusion The N/LP ratio at ICU admission was found to have a significant independent association with 30/60 days mortality and the incidence of AKI in sepsis patients.
Collapse
Affiliation(s)
| | | | - Zhenkui Hu
- Department of Critical Care Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, China
| |
Collapse
|
42
|
Kim HB, Go EJ, Baek JS. Effect of hot-melt extruded Morus alba leaves on intestinal microflora and epithelial cells. Heliyon 2024; 10:e23954. [PMID: 38332870 PMCID: PMC10851307 DOI: 10.1016/j.heliyon.2023.e23954] [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: 09/22/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024] Open
Abstract
Although rutin and isoquercitrin have many effects, they are insoluble substances, making it difficult to obtain pure substances. This study was to investigate whether Morus alba leaves containing rutin and isoquercitrin could improve intestinal health by making a sustained-release formulation through a hot-melt extrusion (HME) process with improved stability and solubility and determine whether it could upregulate the balance of intestinal microorganisms and intestinal epithelial cells. A sustained-release formulation was prepared by the HME process using Morus alba leaves and a hydrophilic polymer matrix. Antibacterial activities of pathogenic microorganisms (Escherichia coli, Streptococcus aureus, Enterococcus faecalis) and proliferative effect of probiotics (Lactobacillus rhamnosus, Pediococcus pentosaceus) were tested against intestinal microorganisms. Regarding intestinal epithelial cells, a co-culture model of Caco-2 cells and RAW 264.7 cells was used. It was confirmed that the extrudate exhibited high antibacterial activities against pathogenic microorganisms and affected the proliferation of probiotics. Furthermore, after inducing inflammation through LPS, it recovered transepithelial electrical resistance-increased levels of tight junction proteins and decreased expression levels of pro-inflammatory cytokines. HME of Morus alba leaves containing rutin and isoquercitrin can upregulate intestinal microbial balance and intestinal epithelial cells.
Collapse
Affiliation(s)
- Hyun Bok Kim
- National Institute of Agricultural Sciences, RDA, Wanju 55365, South Korea
| | - Eun Ji Go
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, South Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, South Korea
- Department of Bio-Functional Materials, Kangwon National University, Samcheok 25949. South Korea
- BeNatureBioLab, Chuncheon 24206, South Korea
| |
Collapse
|
43
|
Riehl L, Fürst J, Kress M, Rykalo N. The importance of the gut microbiome and its signals for a healthy nervous system and the multifaceted mechanisms of neuropsychiatric disorders. Front Neurosci 2024; 17:1302957. [PMID: 38249593 PMCID: PMC10797776 DOI: 10.3389/fnins.2023.1302957] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/13/2023] [Indexed: 01/23/2024] Open
Abstract
Increasing evidence links the gut microbiome and the nervous system in health and disease. This narrative review discusses current views on the interaction between the gut microbiota, the intestinal epithelium, and the brain, and provides an overview of the communication routes and signals of the bidirectional interactions between gut microbiota and the brain, including circulatory, immunological, neuroanatomical, and neuroendocrine pathways. Similarities and differences in healthy gut microbiota in humans and mice exist that are relevant for the translational gap between non-human model systems and patients. There is an increasing spectrum of metabolites and neurotransmitters that are released and/or modulated by the gut microbiota in both homeostatic and pathological conditions. Dysbiotic disruptions occur as consequences of critical illnesses such as cancer, cardiovascular and chronic kidney disease but also neurological, mental, and pain disorders, as well as ischemic and traumatic brain injury. Changes in the gut microbiota (dysbiosis) and a concomitant imbalance in the release of mediators may be cause or consequence of diseases of the central nervous system and are increasingly emerging as critical links to the disruption of healthy physiological function, alterations in nutrition intake, exposure to hypoxic conditions and others, observed in brain disorders. Despite the generally accepted importance of the gut microbiome, the bidirectional communication routes between brain and gut are not fully understood. Elucidating these routes and signaling pathways in more detail offers novel mechanistic insight into the pathophysiology and multifaceted aspects of brain disorders.
Collapse
Affiliation(s)
| | | | | | - Nadiia Rykalo
- Institute of Physiology, Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
44
|
Wang Y, Chen C, Yan W, Fu Y. Epigenetic modification of m 6A methylation: Regulatory factors, functions and mechanism in inflammatory bowel disease. Int J Biochem Cell Biol 2024; 166:106502. [PMID: 38030117 DOI: 10.1016/j.biocel.2023.106502] [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: 11/19/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
Although the exact cause of inflammatory bowel disease (IBD) is still unknown, there is a lot of evidence to support the notion that it results from a combination of environmental factors, immune system issues, gut microbial changes, and genetic susceptibility. In recent years, the role of epigenetics in the pathogenesis of IBD has drawn increasing attention. The regulation of IBD-related immunity, the preservation of the intestinal epithelial barrier, and autophagy are all significantly influenced by epigenetic factors. The most extensive epigenetic methylation modification of mammalian mRNA among them is N6-methyladenosine (m6A). It summarizes the general structure and function of the m6A regulating factors, as well as their complex effects on IBD by regulating the intestinal mucous barrier, intestine mucosal immunity, epidermal cell death, and intestinal microorganisms.This paper provides key insights for the future identification of potential new targets for the diagnosis and treatment of IBD.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyue Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
45
|
Martins-Gomes C, Nunes FM, Silva AM. Natural Products as Dietary Agents for the Prevention and Mitigation of Oxidative Damage and Inflammation in the Intestinal Barrier. Antioxidants (Basel) 2024; 13:65. [PMID: 38247489 PMCID: PMC10812469 DOI: 10.3390/antiox13010065] [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: 12/07/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Food intake is a basic need to sustain life, but foodborne pathogens and food-related xenobiotics are also the main health concerns regarding intestinal barrier homeostasis. With a predominant role in the well-being of the entire human body, intestinal barrier homeostasis is strictly regulated by epithelial and immune cells. These cells are also the main intervenients in oxidative stress and inflammation-related diseases in the intestinal tract, triggered, for example, by genetic/epigenetic factors, food additives, pesticides, drugs, pathogens, and their metabolites. Nevertheless, the human diet can also be seen as a solution for the problem, mainly via the inclusion of functional foods or nutraceuticals that may act as antioxidant/anti-inflammatory agents to prevent and mitigate acute and chronic oxidative damage and inflammation. A literature analysis of recent advances in this topic highlights the significant role of Nrf2 (nuclear factor erythroid 2-related factor 2) and NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells) pathways in these biological processes, with many natural products and phytochemicals targeting endogenous antioxidant systems and cytokine production and balance. In this review, we summarized and discussed studies using in vitro and in vivo models of the intestinal tract used to reproduce oxidative damage and inflammatory events, as well as the role of natural products as modulators of Nrf2 and NK-kB pathways.
Collapse
Affiliation(s)
- Carlos Martins-Gomes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Laboratory, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Laboratory, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
| | - Fernando M. Nunes
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Laboratory, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal;
- Department of Chemistry, School of Life Sciences and Environment, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Amélia M. Silva
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Laboratory, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-food Production (Inov4gro), University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801 Vila Real, Portugal
- Department of Biology and Environment, School of Life Sciences and Environment, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| |
Collapse
|
46
|
Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases. Eur J Immunol 2024; 54:e2249982. [PMID: 37804068 DOI: 10.1002/eji.202249982] [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: 05/17/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Epithelial/immune interactions are characterized by the different properties of the various epithelial tissues, the mediators involved, and the varying immune cells that initiate, sustain, or abrogate allergic diseases on the surface. The intestinal mucosa, respiratory mucosa, and regular skin feature structural differences according to their primary function and surroundings. In the context of these specialized functions, the active role of the epithelium in shaping immune responses is increasingly recognizable. Crosstalk between epithelial and immune cells plays an important role in maintaining homeostatic conditions. While cells of the myeloid cell lineage, mainly macrophages, are the dominating immune cell population in the skin and the respiratory tract, lymphocytes comprise most intraepithelial immune cells in the intestine under healthy conditions. Common to all surface epithelia is the fact that innate immune cells represent the first line of immunosurveillance that either directly defeats invading pathogens or initiates and coordinates more effective successive immune responses involving adaptive immune cells and effector cells. Pharmacological approaches for the treatment of allergic and chronic inflammatory diseases involving epithelial barriers target immunological mediators downstream of the epithelium (such as IL-4, IL-5, IL-13, and IgE). The next generation of therapeutics involves upstream events of the inflammatory cascade, such as epithelial-derived alarmins and related mediators.
Collapse
Affiliation(s)
- Melanie Albrecht
- Molecular Allergology, Vice President´s Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University of Marburg, Marburg, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
47
|
Gu Q, Yan J, Lou Y, Zhang Z, Li Y, Zhu Z, Liu M, Wu D, Liang Y, Pu J, Zhao X, Xiao H, Li P. Bacteriocins: Curial guardians of gastrointestinal tract. Compr Rev Food Sci Food Saf 2024; 23:e13292. [PMID: 38284593 DOI: 10.1111/1541-4337.13292] [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: 08/30/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 01/30/2024]
Abstract
The human gastrointestinal (GI) tract microbiome secretes various metabolites that play pivotal roles in maintaining host physiological balance and influencing disease progression. Among these metabolites, bacteriocins-small, heat-stable peptides synthesized by ribosomes-are notably prevalent in the GI region. Their multifaceted benefits have garnered significant interest in the scientific community. This review comprehensively explores the methods for mining bacteriocins (traditional separation and purification, bioinformatics, and artificial intelligence), their effects on the stomach and intestines, and their complex bioactive mechanisms. These mechanisms include flora regulation, biological barrier restoration, and intervention in epithelial cell pathways. By detailing each well-documented bacteriocin, we reveal the diverse ways in which bacteriocins interact with the GI environment. Moreover, the future research direction is prospected. By further studying the function and interaction of intestinal bacteriocins, we can discover new pharmacological targets and develop drugs targeting intestinal bacteriocins to regulate and improve human health. It provides innovative ideas and infinite possibilities for further exploration, development, and utilization of bacteriocins. The inevitable fact is that the continuously exploration of bacteriocins is sure to bring the promising future for demic GI health understanding and interference strategy.
Collapse
Affiliation(s)
- Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Yan
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yeqing Lou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zihao Zhang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yonglu Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Zichun Zhu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Manman Liu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Danli Wu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ying Liang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jiaqian Pu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodan Zhao
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
48
|
Abstract
The remarkable diversity of lymphocytes, essential components of the immune system, serves as an ingenious mechanism for maximizing the efficient utilization of limited host defense resources. While cell adhesion molecules, notably in gut-tropic T cells, play a central role in this mechanism, the counterbalancing molecular details have remained elusive. Conversely, we've uncovered the molecular pathways enabling extracellular vesicles secreted by lymphocytes to reach the gut's mucosal tissues, facilitating immunological regulation. This discovery sheds light on immune fine-tuning, offering insights into immune regulation mechanisms.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
49
|
Kim S, Naziripour A, Prabhala P, Horváth V, Junaid A, Breault DT, Goyal G, Ingber DE. Direct therapeutic effect of sulfadoxine-pyrimethamine on nutritional deficiency-induced enteric dysfunction in a human Intestine Chip. EBioMedicine 2024; 99:104921. [PMID: 38101300 PMCID: PMC10733102 DOI: 10.1016/j.ebiom.2023.104921] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Sulfadoxine-pyrimethamine (SP) antimalarial therapy has been suggested to potentially increase the birth weight of infants in pregnant women in sub-Saharan Africa, independently of malarial infection. Here, we utilized female intestinal organoid-derived cells cultured within microfluidic Organ Chips to investigate whether SP could directly impact intestinal function and thereby improve the absorption of essential fats and nutrients crucial for fetal growth. METHODS Using a human organ-on-a-chip model, we replicated the adult female intestine with patient organoid-derived duodenal epithelial cells interfaced with human intestinal endothelial cells. Nutrient-deficient (ND) medium was perfused to simulate malnutrition, resulting in the appearance of enteric dysfunction indicators such as villus blunting, reduced mucus production, impaired nutrient absorption, and increased inflammatory cytokine secretion. SP was administered to these chips in the presence or absence of human peripheral blood mononuclear cells (PBMCs). FINDINGS Our findings revealed that SP treatment effectively reversed multiple intestinal absorptive abnormalities observed in malnourished female Intestine Chips, as validated by transcriptomic and proteomic analyses. SP also reduced the production of inflammatory cytokines and suppressed the recruitment of PBMCs in ND chips. INTERPRETATION Our results indicate that SP could potentially increase birth weights by preventing enteric dysfunction and suppressing intestinal inflammation. This underscores the potential of SP as a targeted intervention to improve maternal absorption, subsequently contributing to healthier fetal growth. While SP treatment shows promise in addressing malabsorption issues that can influence infant birth weight, we did not model pregnancy in our chips, and thus its usefulness for treatment of malnourished pregnant women requires further investigation through clinical trials. FUNDING The Bill and Melinda Gates Foundation, and the Wyss Institute for Biologically Inspired Engineering at Harvard University, and the HDDC Organoid Core of the P30 DK034854.
Collapse
Affiliation(s)
- Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Pranav Prabhala
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Viktor Horváth
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Abidemi Junaid
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Vascular Biology Program, Boston Children's Hospital and Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA 02139, USA.
| |
Collapse
|
50
|
Qazi AS, Rahman UU, Ahmad B, Safdar W, Ahmad S, Mumtaz S. Diet, Gut Microbes, and Cancer. Cancer Treat Res 2024; 191:163-190. [PMID: 39133408 DOI: 10.1007/978-3-031-55622-7_7] [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: 08/13/2024]
Abstract
Gut microbes are important and may play important role in spreading cancers specially the gastrointestinal malignancies preferably colorectal cancers. Gut microbes and diet can influence the tissues in gastrointestinal tract increasing the risk of cancer spread. Insufficient nutrient intake and imbalance diet can disturb the microbiome of gastrointestinal tract causing metabolism of xenobiotics which is beneficial as well as detrimental. Dietary imbalance may also weaken the immune system which is another reason for spreading and development of cancers. The triage of gut microbiome, host immune system, and dietary patterns may help the initiation of mechanism of carcinogenesis. In addition to its role in carcinogenesis and tumor development, there is still growing evidence as to how intestinal microflora influences the efficacy and toxicity of chemotherapy and immunotherapy by the gut microbiome. It can therefore be used as a biomarker to predict treatment response or poor response and can also be modified to improve cancer treatment.
Collapse
Affiliation(s)
- Asma Saleem Qazi
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan.
| | - Ubaid Ur Rahman
- Department of Microbiology, Quaid e Azam University, Islamabad, Pakistan
| | - Bilal Ahmad
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Waseem Safdar
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Saeed Ahmad
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| | - Sara Mumtaz
- Department of Biological Sciences, National University of Medical Sciences, Islamabad, Pakistan
| |
Collapse
|