1
|
Zhou HM, Yang XY, Yue SJ, Wang WX, Zhang Q, Xu DQ, Li JJ, Tang YP. The identification of metabolites from gut microbiota in coronary heart disease via network pharmacology. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:145-155. [PMID: 38412071 DOI: 10.1080/21691401.2024.2319827] [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: 08/30/2023] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Although the gut microbial metabolites exhibit potential effects on coronary heart disease (CHD), the underlying mechanism remains unclear. In this study, the active gut microbial metabolites acting on CHD and their potential mechanisms of action were explored through a network pharmacological approach. We collected a total of 208 metabolites from the gutMgene database and 726 overlapping targets from the similarity ensemble approach (SEA) and SwissTargetPrediction (STP) database, and ultimately identified 610 targets relevant to CHD. In conjunction with the gutMGene database, we identified 12 key targets. The targets of exogenous substances were removed, and 10 core targets involved in CHD were eventually retained. The microbiota-metabolites-targets-signalling pathways network analysis revealed that C-type lectin receptor signalling pathway, Lachnospiraceae, Escherichia, mitogen-activated protein kinase 1, prostaglandin-endoperoxidase synthase 2, phenylacetylglutamine and alcoholic acid are notable components of CHD and play important roles in the development of CHD. The results of molecular docking experiments demonstrated that AKT1-glycocholic acid and PTGS2-phenylacetylglutamine complexes may act on C-type lectin receptor signalling pathways. In this study, the key substances and potential mechanisms of gut microbial metabolites were analysed via network pharmacological methods, and a scientific basis and comprehensive idea were provided for the effects of gut microbial metabolites on CHD.
Collapse
Affiliation(s)
- Hao-Ming Zhou
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Xin-Yu Yang
- Department of Pharmacy, Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Wen-Xiao Wang
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Jia-Jia Li
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, China
| |
Collapse
|
2
|
Yang L, Hu M, Shao J. Integration of Gut Mycobiota and Oxidative Stress to Decipher the Roles of C-Type Lectin Receptors in Inflammatory Bowel Diseases. Immunol Invest 2024; 53:1177-1204. [PMID: 39115960 DOI: 10.1080/08820139.2024.2388164] [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: 08/10/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) and Crohn's disease (CD) are two subtypes of inflammatory bowel disease (IBD) with rapidly increased incidence worldwide. Although multiple factors contribute to the occurrence and progression of IBD, the role of intestinal fungal species (gut mycobiota) in regulating the severity of these conditions has been increasingly recognized. C-type lectin receptors (CLRs) on hematopoietic cells, including Dectin-1, Dectin-2, Dectin-3, Mincle and DC-SIGN, are a group of pattern recognition receptors (PRRs) that primarily recognize fungi and mediate defense responses, such as oxidative stress. Recent studies have demonstrated the indispensable role of CLRs in protecting the colon from intestinal inflammation and mucosal damage. METHODS AND RESULTS This review provides a comprehensive overview of the role of CLRs in the pathogenesis of IBD. Given the significant impact of mycobiota and oxidative stress in IBD, this review also discusses recent advancements in understanding how these factors exacerbate or ameliorate IBD. Furthermore, the latest developments in CLR-guided IBD therapy are examined to highlight the modulation of CLRs in fungal recognition and oxidative burst during the IBD process. CONCLUSION This review emphasizes the importance of CLRs in IBD, offering new perspectives on the etiology and therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Liu Yang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui 230012, P. R. China
| | - Min Hu
- Department of pathology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, P. R. China
| | - Jing Shao
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui 230012, P. R. China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, P. R. China
| |
Collapse
|
3
|
Yang H, Wang Z, Li L, Wang X, Wei X, Gou S, Ding Z, Cai Z, Ling Q, Hoffmann PR, He J, Liu F, Huang Z. Mannose coated selenium nanoparticles normalize intestinal homeostasis in mice and mitigate colitis by inhibiting NF-κB activation and enhancing glutathione peroxidase expression. J Nanobiotechnology 2024; 22:613. [PMID: 39385176 PMCID: PMC11465824 DOI: 10.1186/s12951-024-02861-2] [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: 03/27/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
Impaired intestinal homeostasis is a major pathological feature of inflammatory bowel diseases (IBD). Mannose and selenium (Se) both demonstrate potential anti-inflammatory and anti-oxidative properties. However, most lectin receptors bind free monosaccharide ligands with relatively low affinity and most Se species induce side effects beyond a very narrow range of dosage. This has contributed to a poorly explored therapies for IBD that combine mannose and Se to target intestinal epithelial cells (IECs) for normalization gut homeostasis. Herein, a facile and safe strategy for ulcerative colitis (UC) treatment was developed using optimized, mannose-functionalized Se nanoparticles (M-SeNPs) encapsulated within a colon-targeted hydrogel delivery system containing alginate (SA) and chitosan (CS). This biocompatible nanosystem was efficiently taken up by IECs and led to increased expression of Se-dependent glutathione peroxidases (GPXs), thereby modulating IECs' immune response. Using a mouse model of DSS-induced colitis, (CS/SA)-embedding M-SeNPs (C/S-MSe) were found to mitigate oxidative stress and inflammation through the inhibition of the NF-kB pathway in the colon. This stabilized mucosal homeostasis of IECs and ameliorated colitis-related symptoms, thereby providing a potential new approach for treatment of IBD.
Collapse
Affiliation(s)
- Hui Yang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhiyao Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Lixin Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xing Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xian Wei
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Shan Gou
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zimo Ding
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhihui Cai
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qinjie Ling
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, U.S.A
| | - Jingjun He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Fei Liu
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Zhi Huang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Wu X, Yang X, Dai Y, Zhao Z, Zhu J, Guo H, Yang R. Single-cell sequencing to multi-omics: technologies and applications. Biomark Res 2024; 12:110. [PMID: 39334490 PMCID: PMC11438019 DOI: 10.1186/s40364-024-00643-4] [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: 04/29/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Cells, as the fundamental units of life, contain multidimensional spatiotemporal information. Single-cell RNA sequencing (scRNA-seq) is revolutionizing biomedical science by analyzing cellular state and intercellular heterogeneity. Undoubtedly, single-cell transcriptomics has emerged as one of the most vibrant research fields today. With the optimization and innovation of single-cell sequencing technologies, the intricate multidimensional details concealed within cells are gradually unveiled. The combination of scRNA-seq and other multi-omics is at the forefront of the single-cell field. This involves simultaneously measuring various omics data within individual cells, expanding our understanding across a broader spectrum of dimensions. Single-cell multi-omics precisely captures the multidimensional aspects of single-cell transcriptomes, immune repertoire, spatial information, temporal information, epitopes, and other omics in diverse spatiotemporal contexts. In addition to depicting the cell atlas of normal or diseased tissues, it also provides a cornerstone for studying cell differentiation and development patterns, disease heterogeneity, drug resistance mechanisms, and treatment strategies. Herein, we review traditional single-cell sequencing technologies and outline the latest advancements in single-cell multi-omics. We summarize the current status and challenges of applying single-cell multi-omics technologies to biological research and clinical applications. Finally, we discuss the limitations and challenges of single-cell multi-omics and potential strategies to address them.
Collapse
Affiliation(s)
- Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xin Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yunhan Dai
- Medical School, Nanjing University, Nanjing, China
| | - Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Junmeng Zhu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
5
|
Nemli DD, Jiang X, Jakob RP, Gloder LM, Schwardt O, Rabbani S, Maier T, Ernst B, Cramer J. Thermodynamics-Guided Design Reveals a Cooperative Hydrogen Bond in DC-SIGN-targeted Glycomimetics. J Med Chem 2024; 67:13813-13828. [PMID: 38771131 DOI: 10.1021/acs.jmedchem.4c00623] [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: 05/22/2024]
Abstract
Due to the shallow and hydrophilic binding sites of carbohydrate-binding proteins, the design of glycomimetics is often complicated by high desolvation costs as well as competition with solvent. Therefore, a careful optimization of interaction vectors and ligand properties is required in the design and optimization of glycomimetics. Here, we employ thermodynamics-guided design to optimize mannose-based glycomimetics targeting the human C-type lectin receptor dendritic cell-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN), a pathogenic host factor in viral infections. By exploring ligand rigidification and hydrogen bond engineering, a monovalent glycomimetic with an unprecedented affinity for DC-SIGN in the low μM range was discovered. A matched molecular pair analysis based on microcalorimetric data revealed a stereospecific hydrogen bond interaction with Glu358/Ser360 as the origin of this cooperative and enthalpically dominated interaction. This detailed insight into the binding mechanism paves the way for an improvement of monovalent glycomimetics targeting DC-SIGN.
Collapse
Affiliation(s)
- Dilara D Nemli
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Xiaohua Jiang
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Roman P Jakob
- Department Biozentrum, Structural Area Focal Biology, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| | - Laura Muñoz Gloder
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Oliver Schwardt
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Said Rabbani
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Timm Maier
- Department Biozentrum, Structural Area Focal Biology, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| | - Beat Ernst
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| |
Collapse
|
6
|
Yao J, Chen Y, Zhang L, Cheng Y, Chen Z, Zhang Y, Zheng X, Lv Y, Wang S, Li Z, Zhao J. pH-responsive CuS/DSF/EL/PVP nanoplatform alleviates inflammatory bowel disease in mice via regulating gut immunity and microbiota. Acta Biomater 2024; 178:265-286. [PMID: 38417643 DOI: 10.1016/j.actbio.2024.02.034] [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: 11/02/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
The clinical treatment of inflammatory bowel disease (IBD) is challenging. We developed copper sulfate (CuS)/disulfiram (DSF)/methacrylic acid-ethyl acrylate copolymer (EL)/polyvinylpyrrolidone (PVP) nanoplatform (CuS/DSF/EL/PVP) and evaluated its efficiency for treating IBD. After oral administration, the pH-sensitive EL protected the CuS/DSF/EL/PVP against degradation by acidic gastric juices. Once the colon was reached, EL was dissolved, releasing DSF and Cu2+. Further, the main in vivo metabolite of DSF can bind to Cu2+ and form copper (II) N, N-diethyldithiocarbamate (CuET), which significantly alleviated acute colitis in mice. Notably, CuS/DSF/EL/PVP outperformed CuS/EL/PVP and DSF/EL/PVP nanoplatforms in reducing colonic pathology and improving the secretion of inflammation-related cytokines (such as IL-4 and IL-10) in the colonic mucosa. RNA-seq analysis revealed that the nanoplatform reduced colonic inflammation and promoted intestinal mucosal repair by upregulating C-type lectin receptor (CLR)-related genes and signaling pathways. Furthermore, CuS/DSF/EL/PVP showed potential for improving colitis Th1/Th17 cells through innate immunity stimulation, down-regulation of inflammatory cytokines, and upregulation of anti-inflammatory cytokines. Additionally, the intervention with CuS/DSF/EL/PVP led to increased intestinal flora diversity, decreased Escherichia-Shigella abundance, and elevated levels of short-chain fatty acid (SCFA)-producing bacteria Prevotella, Lactobacillus, and Bifidobacterium, indicating their potential to modulate the dysregulated intestinal flora and suppress inflammation. STATEMENT OF SIGNIFICANCE: Our study introduces the CuS/DSF/EL/PVP nanoplatform as a therapeutic strategy for treating inflammatory bowel disease (IBD). This approach demonstrates significant efficacy in targeting the colon and alleviating acute colitis in mice. It uniquely modulates gut immunity and microbiota, exhibiting a notable impact on inflammation-related cytokines and promoting intestinal mucosal repair. The nanoplatform's ability to regulate gut flora diversity, combined with its cost-effective and scalable production, positions it as a potentially transformative treatment for IBD, offering new avenues for personalized medical interventions.
Collapse
Affiliation(s)
- Jinpeng Yao
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yu Chen
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China
| | - Liang Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yuancun Cheng
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Zheng Chen
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Yanhui Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Xiaoyi Zheng
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yanwei Lv
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Shige Wang
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China.
| | - Zhaoshen Li
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| | - Jiulong Zhao
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| |
Collapse
|
7
|
Wang Z, Li X, Tian L, Sha D, Sun Q, Wang J. Application of Bioinformatics in Predicting the Efficacy of Digestive Tumour Immunotherapy Target TIM-3 and its Inhibitors. J Cancer 2024; 15:1954-1965. [PMID: 38434966 PMCID: PMC10905402 DOI: 10.7150/jca.92446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 03/05/2024] Open
Abstract
Background: Our main objective is to apply bioinformatics in predicting the efficacy of digestive tumour immunotherapy target TIM-3 and its inhibitors. Methods: Our study used the gene expression omnibus (GEO) database to identify datasets associated with digestive tumours and the action of TIM-3. The GSE427729 dataset based on the GPL10192 platform. The dataset consisted of six samples of total RNA derived from TIM-3 control and knockdown RAW 264.7 cells. We used GEO2R tool to identify DEGs before performing Gene Ontology and identifying the kyoto encyclopedia of genes and genomes (KEGG) pathways. Lastly, we determined the PPI networks to identify hub genes. Results: Our study identified 57 differentially expressed genes based on an adjusted p-value of less than 0.05 and a log2 fold change of 2.0. There were 26 down-regulated genes with 31 up-regulated genes while 22, 404 genes were non-significant. The DEGs were enriched in biological pathways such as activating leukocytes, cells, and development of the immune system. Additionally, we identified four significant KEGG pathways that were implicated in digestive tumour immunotherapy and TIM-3; pathways of pancreatic cancer, NF-Kappa B signalling pathway, Toll-like receptor signalling pathway and C-type lectin receptor signalling pathway. The PPI networks identified 10 hub genes that were implicated in digestive tumour immunotherapy target TIM-3 (Myd88, Traf6, Irf7, Cdk4, Ccnd2, Mapkap1, Prr5, Mpp3, Serpinb6b and Pvrl3). Conclusion: Targeting these biological pathways, KEGG pathways, molecular functions and cellular processes can lead to novel therapeutic treatment and management in digestive tumours based on TIM-3 immunotherapy.
Collapse
Affiliation(s)
- Zexin Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| | - Xibin Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| | - Litao Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| | - Dan Sha
- Department of Minimally Invasive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| | - Qinhui Sun
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| | - Jinshen Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021
| |
Collapse
|
8
|
Zaher K, Basingab F. Interaction between Gut Microbiota and Dendritic Cells in Colorectal Cancer. Biomedicines 2023; 11:3196. [PMID: 38137417 PMCID: PMC10741039 DOI: 10.3390/biomedicines11123196] [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: 10/07/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
Colorectal cancer (CRC) is a malignancy that manifests in serial stages and has been observed to have an escalating incidence in modern societies, causing a significant global health problem. The development of CRC is influenced by various exogenous factors, including lifestyle, diet, nutrition, environment, and microbiota, that can affect host cells, including immune cells. Various immune dysfunctions have been recognized in patients with CRC at different stages of this disease. The signature of microbiota in the development of CRC-inflammation related to obesity, diet, and reactive host cells, such as dendritic cells (DCs)-has been highlighted by many studies. This study focuses on DCs, the primary cellular mediators linking innate and adaptive immune responses against cancer. In addition, this review focuses on the role of microbiota in dysbiosis and how it affects DCs and, in turn, the immune response and progression of CRC by stimulating different sets of T cells. Additionally, DCs' role in protecting this delicate balance is examined. This is to determine how gene yields of commensal microbiota may be critical in restoring this balance when disrupted. The stages of the disease and major checkpoints are discussed, as well as the role of the C-type lectin receptor of immature DCs pattern recognition receptor in CRC. Finally, based on a thorough examination of worldwide clinical studies and recent advancements in cancer immunotherapy, it is recommended that innovative approaches that integrate DC vaccination strategies with checkpoint inhibitors be considered. This approach holds great promise for improving CRC management.
Collapse
Affiliation(s)
- Kawther Zaher
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 21859, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21859, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21585, Saudi Arabia
| |
Collapse
|
9
|
Sun M, Ju J, Xu H, Wang Y. Intestinal fungi and antifungal secretory immunoglobulin A in Crohn's disease. Front Immunol 2023; 14:1177504. [PMID: 37359518 PMCID: PMC10285161 DOI: 10.3389/fimmu.2023.1177504] [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: 03/01/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
The human gastrointestinal tract harbors trillions of commensal microorganisms. Emerging evidence points to a possible link between intestinal fungal dysbiosis and antifungal mucosal immunity in inflammatory bowel disease, especially in Crohn's disease (CD). As a protective factor for the gut mucosa, secretory immunoglobulin A (SIgA) prevents bacteria from invading the intestinal epithelium and maintains a healthy microbiota community. In recent years, the roles of antifungal SIgA antibodies in mucosal immunity, including the regulation of intestinal immunity binding to hyphae-associated virulence factors, are becoming increasingly recognized. Here we review the current knowledge on intestinal fungal dysbiosis and antifungal mucosal immunity in healthy individuals and in patients with CD, discuss the factors governing antifungal SIgA responses in the intestinal mucosa in the latter group, and highlight potential antifungal vaccines targeting SIgA to prevent CD.
Collapse
|
10
|
Ashkar F, Wu J. Effects of Food Factors and Processing on Protein Digestibility and Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37267055 DOI: 10.1021/acs.jafc.3c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Protein is an essential macronutrient. The nutritional needs of dietary proteins are met by digestion and absorption in the small intestine. Indigestible proteins are further metabolized in the gut and produce metabolites via protein fermentation. Thus, protein indigestibility exerts a wide range of effects on gut microbiota composition and function. This review aims to discuss protein digestibility, the effects of food factors, such as protein sources, intake level, and amino acid composition, and making meat analogues. Besides, it provides an inventory of antinutritional factors and processing techniques that influence protein digestibility and, consequently, the diversity and composition of intestinal microbiota. Future studies are warranted to understand the implication of plant-based analogues on protein digestibility and gut microbiota and to elucidate the mechanisms concerning protein digestibility to host gut microbiota using various omics techniques.
Collapse
Affiliation(s)
- Fatemeh Ashkar
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Jianping Wu
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
11
|
Madel MB, Halper J, Ibáñez L, Claire L, Rouleau M, Boutin A, Mahler A, Pontier-Bres R, Ciucci T, Topi M, Hue C, Amiaud J, Iborra S, Sancho D, Heymann D, Garchon HJ, Czerucka D, Apparailly F, Duroux-Richard I, Wakkach A, Blin-Wakkach C. Specific targeting of inflammatory osteoclastogenesis by the probiotic yeast S. boulardii CNCM I-745 reduces bone loss in osteoporosis. eLife 2023; 12:e82037. [PMID: 36848406 PMCID: PMC9977286 DOI: 10.7554/elife.82037] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/12/2023] [Indexed: 03/01/2023] Open
Abstract
Bone destruction is a hallmark of chronic inflammation, and bone-resorbing osteoclasts arising under such a condition differ from steady-state ones. However, osteoclast diversity remains poorly explored. Here, we combined transcriptomic profiling, differentiation assays and in vivo analysis in mouse to decipher specific traits for inflammatory and steady-state osteoclasts. We identified and validated the pattern-recognition receptors (PRR) Tlr2, Dectin-1, and Mincle, all involved in yeast recognition as major regulators of inflammatory osteoclasts. We showed that administration of the yeast probiotic Saccharomyces boulardii CNCM I-745 (Sb) in vivo reduced bone loss in ovariectomized but not sham mice by reducing inflammatory osteoclastogenesis. This beneficial impact of Sb is mediated by the regulation of the inflammatory environment required for the generation of inflammatory osteoclasts. We also showed that Sb derivatives as well as agonists of Tlr2, Dectin-1, and Mincle specifically inhibited directly the differentiation of inflammatory but not steady-state osteoclasts in vitro. These findings demonstrate a preferential use of the PRR-associated costimulatory differentiation pathway by inflammatory osteoclasts, thus enabling their specific inhibition, which opens new therapeutic perspectives for inflammatory bone loss.
Collapse
Affiliation(s)
- Maria-Bernadette Madel
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Julia Halper
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Lidia Ibáñez
- Department of Pharmacy, Cardenal Herrera-CEU UniversityValenciaSpain
| | | | - Matthieu Rouleau
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Antoine Boutin
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Adrien Mahler
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Rodolphe Pontier-Bres
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
- Centre Scientifiquede MonacoMonaco
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Majlinda Topi
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Christophe Hue
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammationMontigny-Le-BretonneuxFrance
| | | | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT. School of Medicine, Universidad Complutense de MadridMadridSpain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Dominique Heymann
- Université de Nantes, Institut de Cancérologie de l’OuestSaint HerblainFrance
| | - Henri-Jean Garchon
- Université Paris-Saclay, UVSQ, INSERM, Infection et inflammationMontigny-Le-BretonneuxFrance
- Genetics Division, Ambroise Paré Hospital, AP-HPBoulogne-BillancourtFrance
| | - Dorota Czerucka
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
- Centre Scientifiquede MonacoMonaco
| | | | | | - Abdelilah Wakkach
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| | - Claudine Blin-Wakkach
- Université Côte d’Azur, CNRS, LP2MNiceFrance
- LIA ROPSE, Laboratoire International Associé Université Côte d’Azur - Centre Scientifique de MonacoNice and MonacoFrance
| |
Collapse
|
12
|
Zhang Y, Wang Y, Li X, Nie D, Liu C, Gan Y. Ligand-modified nanocarriers for oral drug delivery: Challenges, rational design, and applications. J Control Release 2022; 352:813-832. [PMID: 36368493 DOI: 10.1016/j.jconrel.2022.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/15/2022]
Abstract
Ligand-modified nanocarriers (LMNCs) specific to their targets have attracted increasing interest for enhanced oral drug delivery in recent decades. Although the design of LMNCs for enhanced endocytosis and improved exposure of the loaded drugs through the oral route has received abundant attention, it remains unclear how the design influences their transcellular process, especially the key factors affecting their functions. This review discusses the extracellular and cellular barriers to orally administered LMNCs in the gastrointestinal (GI) tract and new discoveries regarding the GI protein corona and the sequential transport barriers that impede the preplanned movements of LMNCs after oral administration. Furthermore, innovative progress in considering key factors (including target selection, ligand properties, and other important factors) in the rational design of LMNCs for oral drug delivery is presented. In particular, some factors that endow LMNCs with efficient transcytosis rather than only endocytosis are highlighted. Finally, the prospects of orally administered LMNCs in disease therapy for the enhanced oral/local bioavailability of active pharmaceutical ingredients, as well as emerging delivery routes, such as lymphatic drug delivery and systemic location-specific drug release based on oral transcellular LMNCs, are discussed.
Collapse
Affiliation(s)
- Yaqi Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Nie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chang Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yong Gan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China.
| |
Collapse
|
13
|
Cheng YC, Kim SW. Use of Microorganisms as Nutritional and Functional Feedstuffs for Nursery Pigs and Broilers. Animals (Basel) 2022; 12:3141. [PMID: 36428369 PMCID: PMC9686830 DOI: 10.3390/ani12223141] [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: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
The objectives of this review paper are to introduce the structures and composition of various microorganisms, to show some applications of single cells as alternative protein supplements or energy feeds in swine and poultry diets, and to discuss the functional effects of microorganisms as feed additives on the growth performance and intestinal health of nursery pigs and broilers. Microorganisms, including bacteria, yeasts, and microalgae, have been commonly supplemented in animal diets because they are cost-effective, stable, and have quantitative production that provides nutritional and functional benefits to pigs and broilers. Microorganisms could be alternative antibiotics to enhance intestinal health due to bioactive components from cell wall components, which interact with receptors on epithelial and immune cells. In addition, bioactive components could be digested by intestinal microbiota to produce short-chain fatty acids and enhance energy utilization. Otherwise, microorganisms such as single-cell protein (SCP) and single-cell oils (SCOs) are sustainable and economic choices to replace conventional protein supplements and energy feeds. Supplementing microorganisms as feedstuffs and feed additives improved the average daily gain by 1.83%, the daily feed intake by 0.24%, and the feed efficiency by 1.46% in pigs and broilers. Based on the properties of each microorganism, traditional protein supplements, energy feeds, and functional feed additives could be replaced by microorganisms, which have shown benefits to animal's growth and health. Therefore, specific microorganisms could be promising alternatives as nutritional and functional feedstuffs in animal diets.
Collapse
Affiliation(s)
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
14
|
Cheng S, Liu Y, Jing Y, Jiang B, Wang D, Chu X, Jia L, Xin S. Identification of key monocytes/macrophages related gene set of the early-stage abdominal aortic aneurysm by integrated bioinformatics analysis and experimental validation. Front Cardiovasc Med 2022; 9:950961. [PMID: 36186997 PMCID: PMC9515382 DOI: 10.3389/fcvm.2022.950961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Objective Abdominal aortic aneurysm (AAA) is a lethal peripheral vascular disease. Inflammatory immune cell infiltration is a central part of the pathogenesis of AAA. It’s critical to investigate the molecular mechanisms underlying immune infiltration in early-stage AAA and look for a viable AAA marker. Methods In this study, we download several mRNA expression datasets and scRNA-seq datasets of the early-stage AAA models from the NCBI-GEO database. mMCP-counter and CIBERSORT were used to assess immune infiltration in early-stage experimental AAA. The scRNA-seq datasets were then utilized to analyze AAA-related gene modules of monocytes/macrophages infiltrated into the early-stage AAA by Weighted Correlation Network analysis (WGCNA). After that, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis for the module genes was performed by ClusterProfiler. The STRING database was used to create the protein-protein interaction (PPI) network. The Differentially Expressed Genes (DEGs) of the monocytes/macrophages were explored by Limma-Voom and the key gene set were identified. Then We further examined the expression of key genes in the human AAA dataset and built a logistic diagnostic model for distinguishing AAA patients and healthy people. Finally, real-time quantitative polymerase chain reaction (RT-qPCR) and Enzyme Linked Immunosorbent Assay (ELISA) were performed to validate the gene expression and serum protein level between the AAA and healthy donor samples in our cohort. Results Monocytes/macrophages were identified as the major immune cells infiltrating the early-stage experimental AAA. After pseudocell construction of monocytes/macrophages from scRNA-seq datasets and WGCNA analysis, four gene modules from two datasets were identified positively related to AAA, mainly enriched in Myeloid Leukocyte Migration, Collagen-Containing Extracellular matrix, and PI3K-Akt signaling pathway by functional enrichment analysis. Thbs1, Clec4e, and Il1b were identified as key genes among the hub genes in the modules, and the high expression of Clec4e, Il1b, and Thbs1 was confirmed in the other datasets. Then, in human AAA transcriptome datasets, the high expression of CLEC4E, IL1B was confirmed and a logistic regression model based on the two gene expressions was built, with an AUC of 0.9 in the train set and 0.79 in the validated set. Additionally, in our cohort, we confirmed the increased serum protein levels of IL-1β and CLEC4E in AAA patients as well as the increased expression of these two genes in AAA aorta samples. Conclusion This study identified monocytes/macrophages as the main immune cells infiltrated into the early-stage AAA and constructed a logistic regression model based on monocytes/macrophages related gene set. This study could aid in the early diagnostic of AAA.
Collapse
Affiliation(s)
- Shuai Cheng
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Yuanlin Liu
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Yuchen Jing
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Bo Jiang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Ding Wang
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Xiangyu Chu
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Longyuan Jia
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
| | - Shijie Xin
- Department of Vascular and Thyroid Surgery, The First Hospital, China Medical University, Shenyang, China
- Key Laboratory of Pathogenesis, Prevention, and Therapeutics of Aortic Aneurysm in Liaoning Province, Shenyang, China
- Regenerative Medicine Research Center of China Medical University, Shenyang, China
- *Correspondence: Shijie Xin,
| |
Collapse
|
15
|
Xie X, Wu Z, Wu Y, Liu J, Chen X, Shi X, Wei C, Li J, Lv J, Li Q, Tang L, He S, Zhan T, Tang Z. Cysteine protease of Clonorchis sinensis alleviates DSS-induced colitis in mice. PLoS Negl Trop Dis 2022; 16:e0010774. [PMID: 36084127 PMCID: PMC9491586 DOI: 10.1371/journal.pntd.0010774] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/21/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Currently, inflammatory bowel disease (IBD) has become a global chronic idiopathic disease with ever-rising morbidity and prevalence. Accumulating evidence supports the IBD-hygiene hypothesis that helminths and their derivatives have potential therapeutic value for IBD. Clonorchis sinensis (C. sinensis) mainly elicit Th2/Treg-dominated immune responses to maintain long-term parasitism in the host. This study aimed to evaluate the therapeutic effects of cysteine protease (CsCP) and adult crude antigen (CsCA) of C. sinensis, and C. sinensis (Cs) infection on DSS-induced colitis mice.
Methods
BALB/c mice were given 5% DSS daily for 7 days to induce colitis. During this period, mice were treated with rCsCP, CsCA or dexamethasone (DXM) every day, or Cs infection which was established in advance. Changes in body weight, disease activity index (DAI), colon lengths, macroscopic scores, histopathological findings, myeloperoxidase (MPO) activity levels, regulatory T cell (Treg) subset levels, colon gene expression levels, serum cytokine levels, and biochemical indexes were measured.
Results
Compared with Cs infection, rCsCP and CsCA alleviated the disease activity of acute colitis more significant without causing abnormal blood biochemical indexes. In comparison, rCsCP was superior to CsCA in attenuating colonic pathological symptoms, enhancing the proportion of Treg cells in spleens and mesenteric lymph nodes, and improving the secretion of inflammatory-related cytokines (e.g., IL-2, IL-4, IL-10 and IL-13) in serum. Combined with RNA-seq data, it was revealed that CsCA might up-regulate the genes related to C-type lectin receptor and intestinal mucosal repair related signal pathways (e.g., Cd209d, F13a1 and Cckbr) to reduce colon inflammation and benefit intestinal mucosal repair. Dissimilarly, rCsCP ameliorated colitis mainly through stimulating innate immunity, such as Toll like receptor (TLR) signaling pathway, down-regulating the expression of inflammatory cytokines (e.g., IL-12b, IL-23r and IL-7), thereby restraining the differentiation of Th1/Th17 cells.
Conclusions
Both rCsCP and CsCA showed good therapeutic effects on the treatment of acute colitis, but rCsCP is a better choice. rCsCP is a safe, effective, readily available and promising therapeutic agent against IBD mainly by activating innate immunity and regulating the IL-12/IL-23r axis.
Collapse
Affiliation(s)
- Xiaoying Xie
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Zhanshuai Wu
- Department of Immunology, Guangxi University of Chinese Medicine, Nanning, China
- GuangXi Medical Transformational Key Laboratory of Combine Traditional Chinese and Western Medicine and High Incidence of Infectious Diseases, Nanning, China
| | - Yuhong Wu
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
- Department of Cell Biology and Genetics, School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Jing Liu
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Xinyuan Chen
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Xiaoqian Shi
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Caiheng Wei
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Jiasheng Li
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
| | - Jiahui Lv
- School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
- Department of Parasitology, Guangxi Medical University, Nanning, China
| | - Qing Li
- Department of Cell Biology and Genetics, School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
| | - Lili Tang
- Department of Parasitology, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
| | - Shanshan He
- Department of Parasitology, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
| | - Tingzheng Zhan
- Department of Parasitology, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- * E-mail: (TZ); (ZT)
| | - Zeli Tang
- Department of Cell Biology and Genetics, School of Pre-clinical Medicine, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- * E-mail: (TZ); (ZT)
| |
Collapse
|
16
|
Oh KK, Choi YR, Gupta H, Ganesan R, Sharma SP, Won SM, Jeong JJ, Lee SB, Cha MG, Kwon GH, Kim DJ, Suk KT. Identification of Gut Microbiome Metabolites via Network Pharmacology Analysis in Treating Alcoholic Liver Disease. Curr Issues Mol Biol 2022; 44:3253-3266. [PMID: 35877448 PMCID: PMC9316215 DOI: 10.3390/cimb44070224] [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: 06/17/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/03/2022] Open
Abstract
Alcoholic liver disease (ALD) is linked to a broad spectrum of diseases, including diabetes, hypertension, atherosclerosis, and even liver carcinoma. The ALD spectrum includes alcoholic fatty liver disease (AFLD), alcoholic hepatitis, and cirrhosis. Most recently, some reports demonstrated that the pathogenesis of ALD is strongly associated with metabolites of human microbiota. AFLD was the onset of disease among ALDs, the initial cause of which is alcohol consumption. Thus, we analyzed the significant metabolites of microbiota against AFLD via the network pharmacology concept. The metabolites from microbiota were retrieved by the gutMGene database; sequentially, AFLD targets were identified by public databases (DisGeNET, OMIM). The final targets were utilized for protein–protein interaction (PPI) networks and signaling pathway analyses. Then, we performed a molecular docking test (MDT) to verify the affinity between metabolite(s) and target(s) utilizing the Autodock 1.5.6 tool. From a holistic viewpoint, we integrated the relationships of microbiota-signaling pathways-targets-metabolites (MSTM) using the R Package. We identified the uppermost six key targets (TLR4, RELA, IL6, PPARG, COX-2, and CYP1A2) against AFLD. The PPI network analysis revealed that TLR4, RELA, IL6, PPARG, and COX-2 had equivalent degrees of value (4); however, CYP1A2 had no associations with the other targets. The bubble chart showed that the PI3K-Akt signaling pathway in nine signaling pathways might be the most significant mechanism with antagonistic functions in the treatment of AFLD. The MDT confirmed that Icaritin is a promising agent to bind stably to RELA (known as NF-Κb). In parallel, Bacterium MRG-PMF-1, the PI3K-Akt signaling pathway, RELA, and Icaritin were the most significant components against AFLD in MSTM networks. In conclusion, we showed that the Icaritin–RELA complex on the PI3K-Akt signaling pathway by bacterial MRG-PMF-1 might have promising therapeutic effects against AFLD, providing crucial evidence for further research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Ki-Tae Suk
- Correspondence: ; Tel.: +82-10-5365-5700; Fax: +82-033-248-3481
| |
Collapse
|
17
|
Li M, Zhang R, Li J, Li J. The Role of C-Type Lectin Receptor Signaling in the Intestinal Microbiota-Inflammation-Cancer Axis. Front Immunol 2022; 13:894445. [PMID: 35619716 PMCID: PMC9127077 DOI: 10.3389/fimmu.2022.894445] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022] Open
Abstract
As a subset of pattern recognition receptors (PRRs), C-type lectin-like receptors (CLRs) are mainly expressed by myeloid cells as both transmembrane and soluble forms. CLRs recognize not only pathogen associated molecular patterns (PAMPs), but also damage-associated molecular patterns (DAMPs) to promote innate immune responses and affect adaptive immune responses. Upon engagement by PAMPs or DAMPs, CLR signaling initiates various biological activities in vivo, such as cytokine secretion and immune cell recruitment. Recently, several CLRs have been implicated as contributory to the pathogenesis of intestinal inflammation, which represents a prominent risk factor for colorectal cancer (CRC). CLRs function as an interface among microbiota, intestinal epithelial barrier and immune system, so we firstly discussed the relationship between dysbiosis caused by microbiota alteration and inflammatory bowel disease (IBD), then focused on the role of CLRs signaling in pathogenesis of IBD (including Mincle, Dectin-3, Dectin-1, DCIR, DC-SIGN, LOX-1 and their downstream CARD9). Given that CLRs mediate intricate inflammatory signals and inflammation plays a significant role in tumorigenesis, we finally highlight the specific effects of CLRs on CRC, especially colitis-associated cancer (CAC), hoping to open new horizons on pathogenesis and therapeutics of IBD and CAC.
Collapse
Affiliation(s)
- Muhan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Runfeng Zhang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ji Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Gut Microbiota Translational Medicine Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23073464. [PMID: 35408838 PMCID: PMC8998182 DOI: 10.3390/ijms23073464] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Microbe-host communication is essential to maintain vital functions of a healthy host, and its disruption has been associated with several diseases, including Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease (IBD). Although individual members of the intestinal microbiota have been associated with experimental IBD, identifying microorganisms that affect disease susceptibility and phenotypes in humans remains a considerable challenge. Currently, the lack of a definition between what is healthy and what is a dysbiotic gut microbiome limits research. Nevertheless, although clear proof-of-concept of causality is still lacking, there is an increasingly evident need to understand the microbial basis of IBD at the microbial strain, genomic, epigenomic, and functional levels and in specific clinical contexts. Recent information on the role of diet and novel environmental risk factors affecting the gut microbiome has direct implications for the immune response that impacts the development of IBD. The complexity of IBD pathogenesis, involving multiple distinct elements, suggests the need for an integrative approach, likely utilizing computational modeling of molecular datasets to identify more specific therapeutic targets.
Collapse
|
19
|
Liberti A, Natarajan O, Atkinson CGF, Dishaw LJ. Secreted immunoglobulin domain effector molecules of invertebrates and management of gut microbial ecology. Immunogenetics 2022; 74:99-109. [PMID: 34988622 DOI: 10.1007/s00251-021-01237-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/18/2021] [Indexed: 02/01/2023]
Abstract
The origins of a "pass-through" gut in early bilaterians facilitated the exploration of new habitats, motivated the innovation of feeding styles and behaviors, and helped drive the evolution of more complex organisms. The gastrointestinal tract has evolved to consist of a series of interwoven exchanges between nutrients, host immunity, and an often microbe-rich environmental interface. Not surprisingly, animals have expanded their immune repertoires to include soluble effectors that can be secreted into luminal spaces, e.g., in the gut, facilitating interactions with microbes in ways that influence their settlement dynamics, virulence, and their interaction with other microbes. The immunoglobulin (Ig) domain, which is also found in some non-immune molecules, is recognized as one of the most versatile recognition domains lying at the interface of innate and adaptive immunity; among vertebrates, secreted Igs are known to play crucial roles in the management of gut microbial communities. In this mini-review, we will focus on secreted immune effectors possessing Ig-like domains in invertebrates, such as the fibrinogen-related effector proteins first described in the gastropod Biomphalaria glabrata, the Down syndrome cellular adhesion molecule first described in the arthropod, Drosophila melanogaster, and the variable region-containing chitin-binding proteins of the protochordates. We will highlight our current understanding of their function and their potential role, if not yet recognized, in the establishment and maintenance of host-microbial interfaces and argue that these Igs are likely also essential to microbiome management.
Collapse
Affiliation(s)
- Assunta Liberti
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Ojas Natarajan
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Division of Molecular Genetics, Children's Research Institute, St. Petersburg, FL, USA
| | - Celine Grace F Atkinson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA.,Division of Molecular Genetics, Children's Research Institute, St. Petersburg, FL, USA
| | - Larry J Dishaw
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL, USA. .,Division of Molecular Genetics, Children's Research Institute, St. Petersburg, FL, USA.
| |
Collapse
|
20
|
Immune cell-specific smoking-related expression characteristics are revealed by re-analysis of transcriptomes from the CEDAR cohort. Cent Eur J Immunol 2022; 47:246-259. [PMID: 36817262 PMCID: PMC9896985 DOI: 10.5114/ceji.2022.120618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction Smoking is known to affect whole-blood expression and methylation profiles. Although whole-genome methylation studies indicated that effects observed in blood may be driven by changes within leukocyte subtypes, these phenomena have not been explored using expression profiling. Material and methods This study reanalyzed data from the Correlated Expression and Disease Association Research (CEDAR) patient cohort recruited by Momozawa et al. (E-MTAB-6667). Data from gene expression profiling of immunomagnetically sorted CD4+, CD8+, CD14+, CD15+, and CD19+ cells were processed. Differential expression analyses were conducted in each immune cell type, followed by gene ontology analysis and supplementary investigations. Results Ninety-four differentially expressed genes were found (CD8+ n = 58, CD14+ n = 20, CD4+ n = 14, CD19+ n = 2). Two key smoking-related genes were overexpressed in specific cell types: LRRN3 (CD4+, CD8+) and MMP25 (CD8+, CD14+). In CD4+ cells smoking was associated with reduced expression of the NK cell receptor KLRB1, suggesting CD4+ subpopulation shifts and differences in interferon signaling (reduced IRF1 and IL18RAP in smokers). Key results and their integration with an immune protein-protein interaction network revealed that smoking influences integrins in CD8+ cells (ITGB7, ITGAL, ITGAM, ITGB2). C-type lectin CLEC4A was reduced in CD8+ cells and CLEC10A was increased in CD14+ cells from smokers; moreover, CLEC5A (CD8+), CLEC7A (CD8+) and CLEC9A (CD19+) were related to smoking in supplementary analyses. CD14+ cells from smokers exhibited overexpression of LDLR and the formyl peptide receptor FPR3. Conclusions Smoking specifically alters vital immune regulation genes in lymphocyte subtypes, especially CD4+, CD8+ and CD14+ cells.
Collapse
|
21
|
Zhou R, He D, Xie J, Zhou Q, Zeng H, Li H, Huang L. The Synergistic Effects of Polysaccharides and Ginsenosides From American Ginseng ( Panax quinquefolius L.) Ameliorating Cyclophosphamide-Induced Intestinal Immune Disorders and Gut Barrier Dysfunctions Based on Microbiome-Metabolomics Analysis. Front Immunol 2021; 12:665901. [PMID: 33968068 PMCID: PMC8100215 DOI: 10.3389/fimmu.2021.665901] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/29/2021] [Indexed: 12/15/2022] Open
Abstract
Cyclophosphamide (CTX), used in cancer chemotherapy, a high dose of which would cause immunosuppressive effect and intestinal mucosa damage. American ginseng (Panax quinquefolius L.) has a long history of functional food use for immunological disorder, colitis, cancer, and so on. This study aimed to illustrate the underlying mechanism of American ginseng’s immunomodulatory effect in CTX-induced mice. In this study, all groups of American ginseng (American ginseng polysaccharide [AGP], American ginseng ginsenoside [AGG], co-treated with American ginseng polysaccharide and ginsenoside [AGP_AGG]) have relieve the immune disorder by reversing the lymphocyte subsets ratio in spleen and peripheral blood, as well as stimulating CD4+T cells and IgA-secreting cells in small intestine. These three treatment groups, especially AGP_AGG co-treated group recovered the intestine morphology that up-regulated villus height (VH)/crypt depth (CD) ratio, areas of mucins expression, quantity of goblet cells, and expression of tight junction proteins (ZO-1, occludin). Importantly, the microbiome-metabolomics analysis was applied in this study to illustrate the possible immuno-modulating mechanism. The synergistic effect of polysaccharides and ginsenosides (AGP_AGG group) restored the gut microbiota composition and increased various beneficial mucosa-associated bacterial taxa Clostridiales, Bifidobacterium, and Lachnospiraceae, while decreased harmful bacteria Escherichia-Shigella and Peptococcaceae. Also, AGP_AGG group altered various fecal metabolites such as uric acid, xanthurenic acid, acylcarnitine, 9,10-DHOME, 13-HDoHE, LysoPE15:0, LysoPC 16:0, LysoPI 18:0, and so on, that associated with immunometabolism or protective effect of gut barrier. These results suggest AG, particularly co-treated of polysaccharide and ginsenoside may be used as immunostimulants targeting microbiome-metabolomics axis to prevent CTX-induced side effects in cancer patients.
Collapse
Affiliation(s)
- Rongrong Zhou
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China.,National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dan He
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Xie
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qingyijun Zhou
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hongliang Zeng
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hongmei Li
- Insitute of Chinese Materia, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Palombo V, Alharthi A, Batistel F, Parys C, Guyader J, Trevisi E, D'Andrea M, Loor JJ. Unique adaptations in neonatal hepatic transcriptome, nutrient signaling, and one-carbon metabolism in response to feeding ethyl cellulose rumen-protected methionine during late-gestation in Holstein cows. BMC Genomics 2021; 22:280. [PMID: 33865335 PMCID: PMC8053294 DOI: 10.1186/s12864-021-07538-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/11/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Methionine (Met) supply during late-pregnancy enhances fetal development in utero and leads to greater rates of growth during the neonatal period. Due to its central role in coordinating nutrient and one-carbon metabolism along with immune responses of the newborn, the liver could be a key target of the programming effects induced by dietary methyl donors such as Met. To address this hypothesis, liver biopsies from 4-day old calves (n = 6/group) born to Holstein cows fed a control or the control plus ethyl-cellulose rumen-protected Met for the last 28 days prepartum were used for DNA methylation, transcriptome, metabolome, proteome, and one-carbon metabolism enzyme activities. RESULTS Although greater withers and hip height at birth in Met calves indicated better development in utero, there were no differences in plasma systemic physiological indicators. RNA-seq along with bioinformatics and transcription factor regulator analyses revealed broad alterations in 'Glucose metabolism', 'Lipid metabolism, 'Glutathione', and 'Immune System' metabolism due to enhanced maternal Met supply. Greater insulin sensitivity assessed via proteomics, and efficiency of transsulfuration pathway activity suggested beneficial effects on nutrient metabolism and metabolic-related stress. Maternal Met supply contributed to greater phosphatidylcholine synthesis in calf liver, with a role in very low density lipoprotein secretion as a mechanism to balance metabolic fates of fatty acids arising from the diet or adipose-depot lipolysis. Despite a lack of effect on hepatic amino acid (AA) transport, a reduction in metabolism of essential AA within the liver indicated an AA 'sparing effect' induced by maternal Met. CONCLUSIONS Despite greater global DNA methylation, maternal Met supply resulted in distinct alterations of hepatic transcriptome, proteome, and metabolome profiles after birth. Data underscored an effect on maintenance of calf hepatic Met homeostasis, glutathione, phosphatidylcholine and taurine synthesis along with greater efficiency of nutrient metabolism and immune responses. Transcription regulators such as FOXO1, PPARG, E2F1, and CREB1 appeared central in the coordination of effects induced by maternal Met. Overall, maternal Met supply induced better immunometabolic status of the newborn liver, conferring the calf a physiologic advantage during a period of metabolic stress and suboptimal immunocompetence.
Collapse
Affiliation(s)
- Valentino Palombo
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, via De Sanctis snc, 86100, Campobasso, Italy
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA
| | - Abdulrahman Alharthi
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Fernanda Batistel
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Claudia Parys
- Evonik Operations GmbH, Hanau-Wolfgang, 63457, Essen, Germany
| | - Jessie Guyader
- Evonik Operations GmbH, Hanau-Wolfgang, 63457, Essen, Germany
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, 29122, Piacenza, Italy
| | - Mariasilvia D'Andrea
- Dipartimento Agricoltura, Ambiente e Alimenti, Università degli Studi del Molise, via De Sanctis snc, 86100, Campobasso, Italy
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL, 61801, USA.
| |
Collapse
|
23
|
The Immunomodulatory Properties of β-2,6 Fructans: A Comprehensive Review. Nutrients 2021; 13:nu13041309. [PMID: 33921025 PMCID: PMC8071392 DOI: 10.3390/nu13041309] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/07/2023] Open
Abstract
Polysaccharides such as β-2,1-linked fructans including inulin or fructose oligosaccharides are well-known prebiotics with recognised immunomodulatory properties. In recent years, other fructan types covering β-2,6-linked fructans, particularly microbial levans, have gained increasing interest in the field. β-2,6-linked fructans of different degrees of polymerisation can be synthesised by plants or microbes including those that reside in the gastrointestinal tract. Accumulating evidence suggests a role for these β-2,6 fructans in modulating immune function. Here, we provide an overview of the sources and structures of β-2,6 fructans from plants and microbes and describe their ability to modulate immune function in vitro and in vivo along with the suggested mechanisms underpinning their immunomodulatory properties. Further, we discuss the limitations and perspectives pertinent to current studies and the potential applications of β-2,6 fructans including in gut health.
Collapse
|
24
|
Cheng L, Liu WL, Tsou YT, Li JC, Chien CH, Su MP, Liu KL, Huang YL, Wu SC, Tsai JJ, Hsieh SL, Chen CH. Transgenic Expression of Human C-Type Lectin Protein CLEC18A Reduces Dengue Virus Type 2 Infectivity in Aedes aegypti. Front Immunol 2021; 12:640367. [PMID: 33767710 PMCID: PMC7985527 DOI: 10.3389/fimmu.2021.640367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/17/2021] [Indexed: 01/15/2023] Open
Abstract
The C-type lectins, one family of lectins featuring carbohydrate binding domains which participate in a variety of bioprocesses in both humans and mosquitoes, including immune response, are known to target DENV. A human C-type lectin protein CLEC18A in particular shows extensive glycan binding abilities and correlates with type-I interferon expression, making CLEC18A a potential player in innate immune responses to DENV infection; this potential may provide additional regulatory point in improving mosquito immunity. Here, we established for the first time a transgenic Aedes aegypti line that expresses human CLEC18A. This expression enhanced the Toll immune pathway responses to DENV infection. Furthermore, viral genome and virus titers were reduced by 70% in the midgut of transgenic mosquitoes. We found significant changes in the composition of the midgut microbiome in CLEC18A expressing mosquitoes, which may result from the Toll pathway enhancement and contribute to DENV inhibition. Transgenic mosquito lines offer a compelling option for studying DENV pathogenesis, and our analyses indicate that modifying the mosquito immune system via expression of a human immune gene can significantly reduce DENV infection.
Collapse
Affiliation(s)
- Lie Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Liang Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Yun-Ting Tsou
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Hao Chien
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Matthew P Su
- Department of Biological Science, Nagoya University, Nagoya, Japan
| | - Kun-Lin Liu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Ya-Lang Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shih-Cheng Wu
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Jih-Jin Tsai
- Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan.,National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
25
|
Prado Acosta M, Goyette-Desjardins G, Scheffel J, Dudeck A, Ruland J, Lepenies B. S-Layer From Lactobacillus brevis Modulates Antigen-Presenting Cell Functions via the Mincle-Syk-Card9 Axis. Front Immunol 2021; 12:602067. [PMID: 33732234 PMCID: PMC7957004 DOI: 10.3389/fimmu.2021.602067] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
C-type lectin receptors (CLRs) are pattern recognition receptors that are crucial in the innate immune response. The gastrointestinal tract contributes significantly to the maintenance of immune homeostasis; it is the shelter for billions of microorganisms including many genera of Lactobacillus sp. Previously, it was shown that host-CLR interactions with gut microbiota play a crucial role in this context. The Macrophage-inducible C-type lectin (Mincle) is a Syk-coupled CLR that contributes to sensing of mucosa-associated commensals. In this study, we identified Mincle as a receptor for the Surface (S)-layer of the probiotic bacteria Lactobacillus brevis modulating GM-CSF bone marrow-derived cells (BMDCs) functions. We found that the S-layer/Mincle interaction led to a balanced cytokine response in BMDCs by triggering the release of both pro- and anti-inflammatory cytokines. In contrast, BMDCs derived from Mincle−/−, CARD9−/− or conditional Syk−/− mice failed to maintain this balance, thus leading to an increased production of the pro-inflammatory cytokines TNF and IL-6, whereas the levels of the anti-inflammatory cytokines IL-10 and TGF-β were markedly decreased. Importantly, this was accompanied by an altered CD4+ T cell priming capacity of Mincle−/− BMDCs resulting in an increased CD4+ T cell IFN-γ production upon stimulation with L. brevis S-layer. Our results contribute to the understanding of how commensal bacteria regulate antigen-presenting cell (APC) functions and highlight the importance of the Mincle/Syk/Card9 axis in APCs as a key factor in host-microbiota interactions.
Collapse
Affiliation(s)
- Mariano Prado Acosta
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| | - Guillaume Goyette-Desjardins
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| | - Jörg Scheffel
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke Universität Magdeburg, Magdeburg, Germany
| | - Jürgen Ruland
- School of Medicine, Institute of Clinical Chemistry and Pathobiochemistry, Technical University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Bernd Lepenies
- Research Center for Emerging Infections and Zoonoses, Institute for Immunology, University of Veterinary Medicine, Hannover, Germany
| |
Collapse
|
26
|
Baranyai Z, Soria‐Carrera H, Alleva M, Millán‐Placer AC, Lucía A, Martín‐Rapún R, Aínsa JA, la Fuente JM. Nanotechnology‐Based Targeted Drug Delivery: An Emerging Tool to Overcome Tuberculosis. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000113] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Zsuzsa Baranyai
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Héctor Soria‐Carrera
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - Maria Alleva
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
| | - Ana C. Millán‐Placer
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
| | - Ainhoa Lucía
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Rafael Martín‐Rapún
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Departamento de Química Orgánica Facultad de Ciencias Universidad de Zaragoza Zaragoza 50009 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| | - José A. Aínsa
- Departamento de Microbiología, Facultad de Medicina Universidad de Zaragoza C/ Domingo Miral s/n Zaragoza 50009 Spain
- Instituto de Investigación Sanitaria Aragón (IIS‐Aragón) Zaragoza 50009 Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI) Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Instituto de Salud Carlos III Madrid 28029 Spain
| | - Jesús M. la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA) CSIC–Universidad de Zaragoza C/ Mariano Esquillor s/n Zaragoza 50018 Spain
- Biomateriales y Nanomedicina (CIBER‐BBN), Instituto de Salud Carlos III CIBER de Bioingeniería Madrid 28029 Spain
| |
Collapse
|
27
|
Wang J, Tian J, He YH, Yang ZW, Wang L, Lai YX, Xu P. Role of CARD9 in inflammatory signal pathway of peritoneal macrophages in severe acute pancreatitis. J Cell Mol Med 2020; 24:9774-9785. [PMID: 32790017 PMCID: PMC7520331 DOI: 10.1111/jcmm.15559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/24/2020] [Accepted: 06/14/2020] [Indexed: 12/14/2022] Open
Abstract
Previous studies revealed that caspase recruitment domain protein 9 (CARD9) was involved in severe acute pancreatitis (SAP) inflammation and that interfering with its expression in vivo could inhibit inflammation. However, the specific mechanism is unknown. This study aimed to discover the related signal pathways of CARD9 in macrophages. SiRNA interference technology was used in vivo and in vitro to detect CARD9‐related signal pathways in peritoneal macrophages. Furthermore, Toll‐like receptor 4 (TLR4) and membrane‐associated C‐type lectin‐1 (Dectin‐1) pathways in macrophages were activated specially to looking for the upstream signal path of CARD9. Results showed up‐regulation of CARD9 expression in peritoneal macrophages of SAP rats (P < .05). CARD9 siRNA alleviated inflammatory cytokines, and inhibited the phosphorylation of NF‐κB and p38MAPK in peritoneal macrophages in vivo or in vitro. Meanwhile, CARD9 siRNA reduced the concentration of CARD9 and Bcl10 in peritoneal macrophages, and TLR4 and Dectin‐1 took part in CARD9 signal pathways in macrophages. In conclusion, there is an inflammation signal pathway comprised of TLR4/Dectin‐1‐CARD9‐NF‐κB/p38MAPK activated in macrophages in SAP. Blockade of CARD9 expression in macrophages can effectively alleviate SAP inflammation.
Collapse
Affiliation(s)
- Jing Wang
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China.,Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jun Tian
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
| | - Yang-Huan He
- Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhi-Wen Yang
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China.,Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Lin Wang
- Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yue-Xing Lai
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China.,Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ping Xu
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
| |
Collapse
|
28
|
Lindenwald DL, Lepenies B. C-Type Lectins in Veterinary Species: Recent Advancements and Applications. Int J Mol Sci 2020; 21:ijms21145122. [PMID: 32698416 PMCID: PMC7403975 DOI: 10.3390/ijms21145122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 02/06/2023] Open
Abstract
C-type lectins (CTLs), a superfamily of glycan-binding receptors, play a pivotal role in the host defense against pathogens and the maintenance of immune homeostasis of higher animals and humans. CTLs in innate immunity serve as pattern recognition receptors and often bind to glycan structures in damage- and pathogen-associated molecular patterns. While CTLs are found throughout the whole animal kingdom, their ligand specificities and downstream signaling have mainly been studied in humans and in model organisms such as mice. In this review, recent advancements in CTL research in veterinary species as well as potential applications of CTL targeting in veterinary medicine are outlined.
Collapse
|
29
|
Mnich ME, van Dalen R, van Sorge NM. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front Cell Infect Microbiol 2020; 10:309. [PMID: 32733813 PMCID: PMC7358460 DOI: 10.3389/fcimb.2020.00309] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antigen-presenting cells (APCs) are present throughout the human body—in tissues, at barrier sites and in the circulation. They are critical for processing external signals to instruct both local and systemic responses toward immune tolerance or immune defense. APCs express an extensive repertoire of pattern-recognition receptors (PRRs) to detect and transduce these signals. C-type lectin receptors (CLRs) comprise a subfamily of PRRs dedicated to sensing glycans, including those expressed by commensal and pathogenic bacteria. This review summarizes recent findings on the recognition of and responses to bacteria by membrane-expressed CLRs on different APC subsets, which are discussed according to the primary site of infection. Many CLR-bacterial interactions promote bacterial clearance, whereas other interactions are exploited by bacteria to enhance their pathogenic potential. The discrimination between protective and virulence-enhancing interactions is essential to understand which interactions to target with new prophylactic or treatment strategies. CLRs are also densely concentrated at APC dendrites that sample the environment across intact barrier sites. This suggests an–as yet–underappreciated role for CLR-mediated recognition of microbiota-produced glycans in maintaining tolerance at barrier sites. In addition to providing a concise overview of identified CLR-bacteria interactions, we discuss the main challenges and potential solutions for the identification of new CLR-bacterial interactions, including those with commensal bacteria, and for in-depth structure-function studies on CLR-bacterial glycan interactions. Finally, we highlight the necessity for more relevant tissue-specific in vitro, in vivo and ex vivo models to develop therapeutic applications in this area.
Collapse
Affiliation(s)
- Malgorzata E Mnich
- Medical Microbiology, UMC Utrecht, Utrecht University, Utrecht, Netherlands.,GSK, Siena, Italy
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
30
|
Ortiz-Perez A, Donnelly B, Temple H, Tiao G, Bansal R, Mohanty SK. Innate Immunity and Pathogenesis of Biliary Atresia. Front Immunol 2020; 11:329. [PMID: 32161597 PMCID: PMC7052372 DOI: 10.3389/fimmu.2020.00329] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Biliary atresia (BA) is a devastating fibro-inflammatory disease characterized by the obstruction of extrahepatic and intrahepatic bile ducts in infants that can have fatal consequences, when not treated in a timely manner. It is the most common indication of pediatric liver transplantation worldwide and the development of new therapies, to alleviate the need of surgical intervention, has been hindered due to its complexity and lack of understanding of the disease pathogenesis. For that reason, significant efforts have been made toward the development of experimental models and strategies to understand the etiology and disease mechanisms and to identify novel therapeutic targets. The only characterized model of BA, using a Rhesus Rotavirus Type A infection of newborn BALB/c mice, has enabled the identification of key cellular and molecular targets involved in epithelial injury and duct obstruction. However, the establishment of an unleashed chronic inflammation followed by a progressive pathological wound healing process remains poorly understood. Like T cells, macrophages can adopt different functional programs [pro-inflammatory (M1) and resolutive (M2) macrophages] and influence the surrounding cytokine environment and the cell response to injury. In this review, we provide an overview of the immunopathogenesis of BA, discuss the implication of innate immunity in the disease pathogenesis and highlight their suitability as therapeutic targets.
Collapse
Affiliation(s)
- Ana Ortiz-Perez
- Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, Netherlands
| | - Bryan Donnelly
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Haley Temple
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Greg Tiao
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ruchi Bansal
- Department of Biomaterials Science and Technology, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, Netherlands
| | - Sujit Kumar Mohanty
- Department of Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
31
|
Choi HH, Lee MH. C-type lectin receptors as potential targets for the treatment of gastrointestinal diseases related to fungal infection. Gastroenterol Rep (Oxf) 2019; 7:376-377. [PMID: 31687160 PMCID: PMC6821261 DOI: 10.1093/gastro/goz048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 08/27/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Hyun Ho Choi
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Mong-Hong Lee
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|