1
|
Wen Y, Ullah H, Ma R, Farooqui NA, Li J, Alioui Y, Qiu J. Anemarrhena asphodeloides Bunge polysaccharides alleviate lipoteichoic acid-induced lung inflammation and modulate gut microbiota in mice. Heliyon 2024; 10:e39390. [PMID: 39469699 PMCID: PMC11513480 DOI: 10.1016/j.heliyon.2024.e39390] [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: 06/28/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
Pneumonia remains a prevalent infection primary ailment characterized by severe lung inflammation, leading to respiratory distress and significant mortality rates, particularly affecting young children in less developed regions. This study explores the therapeutic potential of low and high-molecular weight polysaccharides derived from Anemarrhena asphodeloides in a murine model of lipoteichoic acid (LTA)-induced pneumonia, which represents bacterial-induced lung inflammation. Administration of Anemarrhena asphodeloides polysaccharides effectively alleviated LTA-induced symptoms, including decreased lung and colon inflammation, and restored dysbiosis of gut microbiota. Polysaccharide treatment notably increased mucin-2 expression, reduced serum cytokine levels (IL-10, TNF-α), and increased tight junction protein production (ZO-1, Occludin, Claudin). Additionally, polysaccharides promoted a significant recovery in gut microbiota composition, indicating potential prebiotic effects. These findings highlight the therapeutic capability of Anemarrhena asphodeloides polysaccharides against LTA-induced pneumonia through gut microbiota modulation and restored intestinal homeostasis.
Collapse
Affiliation(s)
- Yuqi Wen
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Hidayat Ullah
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Renzhen Ma
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Nabeel Ahmad Farooqui
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Jiaxin Li
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Yamina Alioui
- Department of Biotechnology, College of Basic Medical Science, Dalian Medical University, Dalian 116044, China
| | - Juanjuan Qiu
- Central Lab, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Zhang Y, Wang Y, Xin E, Zhang Z, Ma D, Liu T, Gao F, Bian T, Sun Y, Wang M, Wang Z, Yan X, Li Y. Network pharmacology and experimental verification reveal the mechanism of Hedysari Radix and Curcumae Rhizoma with the optimal compatibility ratio against colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117555. [PMID: 38110130 DOI: 10.1016/j.jep.2023.117555] [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: 09/22/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The herb pair Astragali Radix (AR) and Curcumae Rhizoma (vinegar-processed, VPCR), derived from the traditional Chinese medicine (TCM) text 'Yixuezhongzhongcanxilu', have long been used to treat gastrointestinal diseases, notably colitis-associated colorectal cancer (CAC). Hedysari Radix (HR), belonging to the same Leguminosae family as AR but from a different genus, is traditionally used as a substitute for AR when paired with VPCR in the treatment of CAC. However, the optimal compatibility ratio for HR-VPCR against CAC and the underlying mechanisms remain unclear. AIM OF THE STUDY To investigate the optimal compatibility ratio and underlying mechanisms of HR-VPCR against CAC using a combination of comparative pharmacodynamics, network pharmacology, and experimental verification. MATERIALS AND METHODS The efficacy of different compatibility ratios of HR-VPCR against CAC was evaluated using various indicators, including the body weight, colon length, tumor count, survival rate, disease activity index (DAI) score, Haemotoxylin and Eosin (H&E) pathological sections, inflammation cytokines (IL-1β, IL-6, IL-10, TNF-α), tumor markers (K-Ras, p53), and intestinal permeability proteins (claudin-1, E-cadherin, mucin-2). Then, the optimal compatibility ratio of HR-VPCR against CAC was determined based on the fuzzy matter-element analysis by integrating the above indicators. After high-performance liquid chromatography (HPLC) analysis for the optimal compatibility ratio of HR-VPCR, potential active components of HR-VPCR were identified by TCMSP and the previous bibliographies. Swiss Targets and GeneCards were adopted to predict the targets of the active components and the targets of CAC, respectively. Then, the common targets of HR-VPCR against CAC were obtained by Venn analysis. PPI networks were constructed in STRING. GO and KEGG enrichments were visualized by the David database. Finally, the predicted pathway was experimentally validated via Western blot. RESULTS Various compatibility ratios of HR-VPCR demonstrated notable therapeutic effects to some extent, evidenced by improvements in body weight, colon length, tumor count, pathological symptoms (DAI score), colon and organ indexes, survival rate, and modulation of inflammation factors (IL-1β, IL-6, IL-10, TNF-α), as well as tumor markers (K-Ras, p53), and down-regulation of intestinal permeability proteins (claudin-1, E-cadherin, mucin-2) in CAC mice. Among these ratios, the ratio 4:1 represents the optimal compatibility ratio by the fuzzy matter-element analysis. Thirty active components of HR-VPCR were carefully selected, targeting 553 specific genes. Simultaneously, 2022 targets associated with CAC were identified. 88 common targets were identified after generating a Venn plot. Following PPI network analysis, 29 core targets were established, with AKT1 ranking highest among them. Further analysis via GO and KEGG enrichment identified the PI3K-AKT signaling pathway as a potential mechanism. Experimental validation confirmed that HR-VPCR intervention effectively reversed the activated PI3K-AKT signaling pathway. CONCLUSIONS The optimal compatibility ratio for the HR-VPCR herb pair in alleviating CAC is 4:1. HR-VPCR exerts its effects by alleviating intestinal inflammation, improving intestinal permeability, and regulating the PI3K-AKT signaling pathway.
Collapse
Affiliation(s)
- Yugui Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yanjun Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Erdan Xin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Zhuanhong Zhang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Dingcai Ma
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Ting Liu
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Feiyun Gao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Tiantian Bian
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yujing Sun
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Maomao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Zhe Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Xingke Yan
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| | - Yuefeng Li
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Key Laboratory of Standard and Quality of Chinese Medicine Research of Gansu, Engineering Research Center of Chinese Medicine Pharmaceutical Process of Gansu, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China; Scientific Research and Experimental Center, Gansu University of Chinese Medicine, Lanzhou, 730000, PR China.
| |
Collapse
|
3
|
Ding J, Zhang H, Dai T, Gao X, Yin Z, Wang Q, Long M, Tan S. TPGS-b-PBAE Copolymer-Based Polyplex Nanoparticles for Gene Delivery and Transfection In Vivo and In Vitro. Pharmaceutics 2024; 16:213. [PMID: 38399267 PMCID: PMC10891721 DOI: 10.3390/pharmaceutics16020213] [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: 11/09/2023] [Revised: 01/19/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Poly (β-amino ester) (PBAE) is an exceptional non-viral vector that is widely used in gene delivery, owing to its exceptional biocompatibility, easy synthesis, and cost-effectiveness. However, it carries a high surface positive charge that may cause cytotoxicity. Therefore, hydrophilic d-α-tocopherol polyethylene glycol succinate (TPGS) was copolymerised with PBAE to increase the biocompatibility and to decrease the potential cytotoxicity of the cationic polymer-DNA plasmid polyplex nanoparticles (NPs) formed through electrostatic forces between the polymer and DNA. TPGS-b-PBAE (TBP) copolymers with varying feeding molar ratios were synthesised to obtain products of different molecular weights. Their gene transfection efficiency was subsequently evaluated in HEK 293T cells using green fluorescent protein plasmid (GFP) as the model because free GFP is unable to easily pass through the cell membrane and then express as a protein. The particle size, ζ-potential, and morphology of the TBP2-GFP polyplex NPs were characterised, and plasmid incorporation was confirmed through gel retardation assays. The TBP2-GFP polyplex NPs effectively transfected multiple cells with low cytotoxicity, including HEK 293T, HeLa, Me180, SiHa, SCC-7 and C666-1 cells. We constructed a MUC2 (Mucin2)-targeting CRISPR/cas9 gene editing system in HEK 293T cells, with gene disruption supported by oligodeoxynucleotide (ODN) insertion in vitro. Additionally, we developed an LMP1 (latent membrane protein 1)-targeting CRISPR/cas9 gene editing system in LMP1-overexpressing SCC7 cells, which was designed to cleave fragments expressing the LMP1 protein (related to Epstein-Barr virus infection) and thus to inhibit the growth of the cells in vivo. As evidenced by in vitro and in vivo experiments, this system has great potential for gene therapy applications.
Collapse
Affiliation(s)
- Jiahui Ding
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.D.); (H.Z.)
| | - Handan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.D.); (H.Z.)
| | - Tianli Dai
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.D.); (H.Z.)
| | - Xueqin Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhongyuan Yin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China (Q.W.)
| | - Qiong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China (Q.W.)
| | - Mengqi Long
- Department of Otolaryngology, The Fifth Affiliated Hospital of Sun Yat-sen University, Meihua 52nd Road, Xiangzhou District, Zhuhai 510009, China
| | - Songwei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.D.); (H.Z.)
| |
Collapse
|
4
|
Wei J, Chen C, Feng J, Zhou S, Feng X, Yang Z, Lu H, Tao H, Li L, Xv H, Xuan J, Wang F. Muc2 mucin O-glycosylation interacts with enteropathogenic Escherichia coli to influence the development of ulcerative colitis based on the NF-kB signaling pathway. J Transl Med 2023; 21:793. [PMID: 37940996 PMCID: PMC10631195 DOI: 10.1186/s12967-023-04687-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine characterized by a compromised intestinal epithelial barrier. Mucin glycans are crucial in preserving barrier function during bacterial infections, although the underlying mechanisms remain largely unexplored. METHODS A cohort comprising 15 patients diagnosed with UC and 15 healthy individuals was recruited. Stool samples were collected to perform 16S rRNA gene sequencing, while biopsy samples were subjected to nanocapillary liquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) to assess O-glycosylation. Gene expression was evaluated through qPCR analysis and Western blotting. Furthermore, animal experiments were conducted to investigate the effects of Escherichia coli and/or O-glycan inhibitor benzyl-α-GalNAc on the development of colitis in mice. RESULTS Our findings revealed that the mucus barrier was disrupted during the early stages of UC, while the MUC2 protein content remained unaltered. Additionally, a noteworthy reduction in the O-glycosylation of MUC2 was observed, along with significant changes in the intestinal microbiota during the early stages of UC. These changes included a decrease in intestinal species richness and an increase in the abundance of Escherichia coli (E. coli). Moreover, subsequent to the administration of galactose or O-glycan inhibitor to intestinal epithelial cells, it was observed that the cell culture supernatant had the ability to modify the proliferation and adhesive capacity of E. coli. Furthermore, when pathogenic E. coli or commensal E. coli were cocultured with intestinal epithelium, both strains elicited activation of the NF-KB signaling pathway in epithelial cells and facilitated the expression of serine protease in comparison to the untreated control. Consistently, the inhibition of O-glycans has been observed to enhance the pathogenicity of E. coli in vivo. Furthermore, a correlation has been established between the level of O-glycans and the development of ulcerative colitis. Specifically, a reduction in the O-glycan content of MUC2 cells has been found to increase the virulence of E. coli, thereby compromising the integrity of the intestinal epithelial barrier. CONCLUSIONS Together, there exist complex interactions between the intestinal epithelium, O-glycans, and the intestinal microbiota, which may inform the development of novel therapeutic strategies for the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Juan Wei
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Chunyan Chen
- Department of Gastroenterology and Hepatology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jing Feng
- Department of Gastroenterology and Hepatology, The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuping Zhou
- Department of Gastroenterology and Hepatology, Huainan First People's Hospital and, First Affiliated Hospital of The Medical College of Anhui, University of Science and Technology, Huainan, 232000, Anhui, People's Republic of China
| | - Xiaoyue Feng
- Department of Gastroenterology and Hepatology, Jinling Clinical College of Nanjing Medical University, Nanjing, 210002, People's Republic of China
| | - Zhao Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Heng Lu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Hui Tao
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Liuying Li
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Huabing Xv
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China
| | - Ji Xuan
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China.
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 305 East Zhongshan Road, Nanjing, 210002, People's Republic of China.
| |
Collapse
|
5
|
Gu M, Yin W, Zhang J, Yin J, Tang X, Ling J, Tang Z, Yin W, Wang X, Ni Q, Zhu Y, Chen T. Role of gut microbiota and bacterial metabolites in mucins of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1119992. [PMID: 37265504 PMCID: PMC10229905 DOI: 10.3389/fcimb.2023.1119992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
Colorectal cancer (CRC) is a major health burden, accounting for approximately 10% of all new cancer cases worldwide. Accumulating evidence suggests that the crosstalk between the host mucins and gut microbiota is associated with the occurrence and development of CRC. Mucins secreted by goblet cells not only protect the intestinal epithelium from microorganisms and invading pathogens but also provide a habitat for commensal bacteria. Conversely, gut dysbiosis results in the dysfunction of mucins, allowing other commensals and their metabolites to pass through the intestinal epithelium, potentially triggering host responses and the subsequent progression of CRC. In this review, we summarize how gut microbiota and bacterial metabolites regulate the function and expression of mucin in CRC and novel treatment strategies for CRC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiangjun Wang
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Qing Ni
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Yunxiang Zhu
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| | - Tuo Chen
- *Correspondence: Xiangjun Wang, ; Qing Ni, ; Yunxiang Zhu, ; Tuo Chen,
| |
Collapse
|
6
|
Liu Y, Yu Z, Zhu L, Ma S, Luo Y, Liang H, Liu Q, Chen J, Guli S, Chen X. Orchestration of MUC2 - The key regulatory target of gut barrier and homeostasis: A review. Int J Biol Macromol 2023; 236:123862. [PMID: 36870625 DOI: 10.1016/j.ijbiomac.2023.123862] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut mucosa of human is covered by mucus, functioning as a crucial defense line for the intestine against external stimuli and pathogens. Mucin2 (MUC2) is a subtype of secretory mucins generated by goblet cells and is the major macromolecular component of mucus. Currently, there is an increasing interest on the investigations of MUC2, noting that its function is far beyond a maintainer of the mucus barrier. Moreover, numerous gut diseases are associated with dysregulated MUC2 production. Appropriate production level of MUC2 and mucus contributes to gut barrier function and homeostasis. The production of MUC2 is regulated by a series of physiological processes, which are orchestrated by various bioactive molecules, signaling pathways and gut microbiota, etc., forming a complex regulatory network. Incorporating the latest findings, this review provided a comprehensive summary of MUC2, including its structure, significance and secretory process. Furthermore, we also summarized the molecular mechanisms of the regulation of MUC2 production aiming to provide developmental directions for future researches on MUC2, which can act as a potential prognostic indicator and targeted therapeutic manipulation for diseases. Collectively, we elucidated the micro-level mechanisms underlying MUC2-related phenotypes, hoping to offer some constructive guidance for intestinal and overall health of mankind.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Lanping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Shuang Ma
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Yang Luo
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Huixi Liang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Qinlingfei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Sitan Guli
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China.
| |
Collapse
|
7
|
Li J, Wang L, Zhang X, Liu P, Deji Z, Xing Y, Zhou Y, Lin X, Huang Z. Per- and polyfluoroalkyl substances exposure and its influence on the intestinal barrier: An overview on the advances. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 852:158362. [PMID: 36055502 DOI: 10.1016/j.scitotenv.2022.158362] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/06/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Per- and polyfluoroalkyl substances (PFAS) are a class of artificially synthetic organic compounds that are hardly degraded in the natural environment. PFAS have been widely used for many decades, and the persistence and potential toxicity of PFAS are an emerging concern in the world. PFAS exposed via diet can be readily absorbed by the intestine and enter the circulatory system or accumulate directly at intestinal sites, which could interact with the intestine and cause the destruction of intestinal barrier. This review summarizes current relationships between PFAS exposure and intestinal barrier damage with a focus on more recent toxicological studies. Exposure to PFAS could cause inflammation in the gut, destruction of the gut epithelium and tight junction structure, reduction of the mucus layer, and induction of the toxicity of immune cells. PFAS accumulation could also induce microbial disorders and metabolic products changes. In addition, there are limited studies currently, and most available studies converge on the health risk of PFAS exposure for human intestinal disease. Therefore, more efforts are deserved to further understand potential associations between PFAS exposure and intestinal dysfunction and enable better assessment of exposomic toxicology and health risks for humans in the future.
Collapse
Affiliation(s)
- Jiaoyang Li
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Lei Wang
- School of Agriculture, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xin Zhang
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Peng Liu
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Zhuoma Deji
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Yudong Xing
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Yan Zhou
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Xia Lin
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China
| | - Zhenzhen Huang
- Department of Preventive Medicine, School of Public Health, Wuhan University, Wuhan 430071, PR China.
| |
Collapse
|
8
|
Zhang Y, Chen Y, Zhou J, Wang X, Ma L, Li J, Yang L, Yuan H, Pang D, Ouyang H. Porcine Epidemic Diarrhea Virus: An Updated Overview of Virus Epidemiology, Virulence Variation Patterns and Virus-Host Interactions. Viruses 2022; 14:2434. [PMID: 36366532 PMCID: PMC9695474 DOI: 10.3390/v14112434] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
The porcine epidemic diarrhea virus (PEDV) is a member of the coronavirus family, causing deadly watery diarrhea in newborn piglets. The global pandemic of PEDV, with significant morbidity and mortality, poses a huge threat to the swine industry. The currently developed vaccines and drugs are only effective against the classic GI strains that were prevalent before 2010, while there is no effective control against the GII variant strains that are currently a global pandemic. In this review, we summarize the latest progress in the biology of PEDV, including its transmission and origin, structure and function, evolution, and virus-host interaction, in an attempt to find the potential virulence factors influencing PEDV pathogenesis. We conclude with the mechanism by which PEDV components antagonize the immune responses of the virus, and the role of host factors in virus infection. Essentially, this review serves as a valuable reference for the development of attenuated virus vaccines and the potential of host factors as antiviral targets for the prevention and control of PEDV infection.
Collapse
Affiliation(s)
- Yuanzhu Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Yiwu Chen
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Jian Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Xi Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Lerong Ma
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Jianing Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Lin Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
| | - Hongming Yuan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
| | - Daxin Pang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
| | - Hongsheng Ouyang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Sciences, Jilin University, Changchun 130062, China
- Chongqing Research Institute, Jilin University, Chongqing 401120, China
- Chongqing Jitang Biotechnology Research Institute Co., Ltd., Chongqing 401120, China
| |
Collapse
|
9
|
Choi J, Marshall B, Ko H, Shi H, Singh AK, Thippareddi H, Holladay S, Gogal RM, Kim WK. Antimicrobial and immunomodulatory effects of tannic acid supplementation in broilers infected with Salmonella Typhimurium. Poult Sci 2022; 101:102111. [PMID: 36081234 PMCID: PMC9465346 DOI: 10.1016/j.psj.2022.102111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/03/2022] [Accepted: 07/27/2022] [Indexed: 12/23/2022] Open
Abstract
Infection by Salmonella Typhimurium, a food-borne pathogen, can reduce the poultry production efficiency. The objective of this study was to investigate the effects of tannic acid (TA) supplementation on growth performance, Salmonella colonization, gut barrier integrity, serum endotoxin levels, antioxidant capacity, gut health, and immune function in broilers infected with the Salmonella enterica serovar Typhimurium nalidixic acid resistant strain (STNR). A total of 546 one-day-old broilers were arbitrarily allocated into 6 treatments including 1) Sham-challenged control (SCC; birds fed a basal diet and administrated peptone water); 2) Challenged control (CC; birds fed a basal diet and inoculated with 108 STNR); 3) Tannic acid 0.25 (TA0.25; CC + 0.25 g/kg TA); 4) TA0.5 (CC + 0.5 g/kg TA); 5) TA1 (CC + 1 g/kg TA); and 6) TA2 (CC + 2 g/kg TA). On D 7, supplemental TA linearly reduced STNR colonization in the ceca (P < 0.01), and TA1 and TA2 group had significantly lower reduced STNR colonization in the ceca (P < 0.01). On D 7 to 21, average daily gain tended to be linearly increased by supplemental TA (P = 0.097). The serum endotoxin levels were quadratically decreased by supplemental TA on D 21 (P < 0.05). Supplemental TA quadratically increased ileal villus height (VH; P < 0.05), and the TA0.25 group had higher ileal VH compared to the CC group (P < 0.05). Supplemental TA linearly increased percentage of peripheral blood CD8+ T cells on D 18 (P < 0.01). The TA0.5 group had significantly lower lymphocyte numbers compared to the CC groups (P < 0.05). The abundance of monocytes linearly increased with TA supplementation (P < 0.01). Therefore, broilers fed TA had reduced STNR colonization, increased growth performance, decreased serum endotoxin levels, enhanced gut health in the broilers, and stimulated the immune system in broilers infected with STNR. Supplementation of TA (1-2 g/kg) enhanced growth performance and gut health via antimicrobial and immunostimulatory effects in broilers infected with STNR.
Collapse
Affiliation(s)
- Janghan Choi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Brett Marshall
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Hanseo Ko
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Hanyi Shi
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | - Amit Kumar Singh
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| | | | - Steven Holladay
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Robert M Gogal
- Department of Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
10
|
Ma M, Bai X, Wang Z, Dong Y, Chen Y, Cao J. Distribution of intraepithelial lymphocytes, mast cells, and goblet cells in the intestine of alpaca. Anat Histol Embryol 2022; 51:501-508. [PMID: 35656747 DOI: 10.1111/ahe.12823] [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: 04/27/2021] [Revised: 07/19/2021] [Accepted: 05/24/2022] [Indexed: 11/28/2022]
Abstract
Intestinal diseases in ruminants are frequent and susceptible to invasion by exogenous substances, and the intestinal mucosal barrier is the first line of defence of the body's immune defence. At present, the study on the structure of intestinal mucosal immune barrier in alpaca is incomplete. Therefore, the alpaca intestines were studied to show the distribution characteristics of intestinal mucosal barrier structure and cells associated with immune system using histology, histochemistry, and immunohistochemistry. The results showed that the intestinal tract of alpaca was composed of mucosa, submucosa, muscularis, and serosa. Intraepithelial lymphocytes were distributed in mucosal epithelium and glands of the large intestine. Mast cells were distributed in each segment of the intestine, mainly in the intestinal lamina propria, intestinal glands, and duodenal glands around, as well as in the muscularis, and the particles of cytoplasm were obvious. Acidic goblet cells were mainly distributed in the ileal mucosal epithelium and ileal intestinal glands, while sialomucins were mainly expressed in the colon. The cells associated with the immune system in the intestinal mucosa of alpaca play an important role in protecting against foreign microbial invasion and infection, and this result provides a theoretical basis for revealing the occurrence of gastrointestinal diseases in alpaca.
Collapse
Affiliation(s)
- Meng Ma
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xue Bai
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zixu Wang
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yulan Dong
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yaoxing Chen
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jing Cao
- Laboratory of Anatomy of Domestic Animal, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
11
|
Chen W, Lv X, Zhang W, Hu T, Cao X, Ren Z, Getachew T, Mwacharo JM, Haile A, Sun W. Insights Into Long Non-Coding RNA and mRNA Expression in the Jejunum of Lambs Challenged With Escherichia coli F17. Front Vet Sci 2022; 9:819917. [PMID: 35498757 PMCID: PMC9039264 DOI: 10.3389/fvets.2022.819917] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
It has long been recognized that enterotoxigenic Escherichia coli (ETEC) is the major pathogen responsible for vomiting and diarrhea. E. coli F17, a main subtype of ETEC, is characterized by high morbidity and mortality in young livestock. However, the transcriptomic basis underlying E. coli F17 infection has not been fully understood. In the present study, RNA sequencing was conducted to explore the expression profiles of mRNAs and long non-coding RNAs (lncRNAs) in the jejunum of lambs who were identified as resistant or sensitive to E. coli F17 that was obtained in a challenge experiment. A total of 772 differentially expressed (DE) mRNAs and 190 DE lncRNAs were detected between the E. coli F17—resistance and E. coli F17-sensitive lambs (i.e., TFF2, LOC105606142, OLFM4, LYPD8, REG4, APOA4, TCONS_00223467, and TCONS_00241897). Then, a two-step machine learning approach (RX) combination Random Forest and Extreme Gradient Boosting were performed, which identified 16 mRNAs and 17 lncRNAs as potential biomarkers, within which PPP2R3A and TCONS_00182693 were prioritized as key biomarkers involved in E. coli F17 infection. Furthermore, functional enrichment analysis showed that peroxisome proliferator-activated receptor (PPAR) pathway was significantly enriched in response to E. coli F17 infection. Our finding will help to improve the knowledge of the mechanisms underlying E. coli F17 infection and may provide novel targets for future treatment of E. coli F17 infection.
Collapse
Affiliation(s)
- Weihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiaoyang Lv
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Weibo Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tingyan Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiukai Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ziming Ren
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tesfaye Getachew
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa, Ethiopia
| | - Joram M. Mwacharo
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa, Ethiopia
| | - Aynalem Haile
- International Centre for Agricultural Research in the Dry Areas, Addis Ababa, Ethiopia
| | - Wei Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
- *Correspondence: Wei Sun
| |
Collapse
|
12
|
Haderer M, Neubert P, Rinner E, Scholtis A, Broncy L, Gschwendtner H, Kandulski A, Pavel V, Mehrl A, Brochhausen C, Schlosser S, Gülow K, Kunst C, Müller M. Novel pathomechanism for spontaneous bacterial peritonitis: disruption of cell junctions by cellular and bacterial proteases. Gut 2022; 71:580-592. [PMID: 33707230 PMCID: PMC8862089 DOI: 10.1136/gutjnl-2020-321663] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 02/17/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Spontaneous bacterial peritonitis (SBP) is a life-threatening complication of liver cirrhosis with a 1-year mortality of 66%. Bacterial translocation (BT) from the intestine to the mesenteric lymph nodes is crucial for the pathogenesis of SBP. DESIGN Since BT presupposes a leaky intestinal epithelium, the integrity of mucus and epithelial cell junctions (E-cadherin and occludin) was examined in colonic biopsies from patients with liver cirrhosis and controls. SBP-inducing Escherichia coli (E. coli) and Proteus mirabilis (P. mirabilis) were isolated from ascites of patients with liver cirrhosis and co-cultured with Caco-2 cells to characterise bacteria-to-cell effects. RESULTS SBP-derived E. coli and P. mirabilis led to a marked reduction of cell-to-cell junctions in a dose-dependent and time-dependent manner. This effect was enhanced by a direct interaction of live bacteria with epithelial cells. Degradation of occludin is mediated via increased ubiquitination by the proteasome. Remarkably, a novel bacterial protease activity is of pivotal importance for the cleavage of E-cadherin. CONCLUSION Patients with liver cirrhosis show a reduced thickness of colonic mucus, which allows bacteria-to-epithelial cell contact. Intestinal bacteria induce degradation of occludin by exploiting the proteasome of epithelial cells. We identified a novel bacterial protease activity of patient-derived SBP-inducing bacteria, which is responsible for the cleavage of E-cadherin structures. Inhibition of this protease activity leads to stabilisation of cell junctions. Thus, targeting these mechanisms by blocking the ubiquitin-proteasome system and/or the bacterial protease activity might interfere with BT and constitute a novel innovative therapeutic strategy to prevent SBP in patients with liver cirrhosis.
Collapse
Affiliation(s)
- Marika Haderer
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Philip Neubert
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Eva Rinner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Annika Scholtis
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Lucile Broncy
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Heidi Gschwendtner
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Arne Kandulski
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Vlad Pavel
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Alexander Mehrl
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Christoph Brochhausen
- Department of Pathology, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Sophie Schlosser
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Karsten Gülow
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Claudia Kunst
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Martina Müller
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious diseases, University Hospital Regensburg, Regensburg, Bavaria, Germany
| |
Collapse
|
13
|
Lin YL, Li Y. The Biological Synthesis and the Function of Mucin 2 in Pseudomyxoma Peritonei. Cancer Manag Res 2021; 13:7909-7917. [PMID: 34703312 PMCID: PMC8527350 DOI: 10.2147/cmar.s324982] [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: 06/17/2021] [Accepted: 08/11/2021] [Indexed: 11/25/2022] Open
Abstract
Excessive mucus secretion is the most prominent feature of pseudomyxoma peritonei (PMP), which often leads to significant increase in abdominal circumference, intractable abdominal pain, progressive intestinal obstruction, abdominal organ adhesions, and cachexia. Excessive mucus secretion is also the main cause of death. Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is the recommended treatment for PMP. However, recurrence is frequently observed even after CRS and HIPEC, presenting similar clinical manifestations. Mucin 2 (MUC2) is the main type of mucin in PMP and plays a key role in the progressive sclerosis of mucus. To comprehensively demonstrate the biosynthetic process and molecular features of MUC2 and to provide new directions for the development of PMP mucolytic strategies, this review systematically summarizes the molecular biology of MUC2, including MUC2 gene structure, transcription, translation, post-translational modification, tertiary structure, and factors regulating mucus viscoelasticity. The results show that MUC2 is a highly glycosylated protein, with glycan accounts for 80% to 90% of the dry weight. The assembly pattern of MUC2 is highly complicated, presenting a bead-like filament. Salt concentration, pH, mucin concentration and trefoil factor family may contribute to the increase in mucus viscoelasticity and sclerosis, which could be used to develop drugs to soften or even dissolve mucus in the future.
Collapse
Affiliation(s)
- Yu-Lin Lin
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University (Beijing Technical Training Base of Tumor Deep Hyperthermia and Whole-Body Hyperthermia), Department of Oncology, Capital Medical University, Beijing, 100038, People's Republic of China
| | - Yan Li
- Department of Peritoneal Cancer Surgery, Beijing Shijitan Hospital, Capital Medical University (Beijing Technical Training Base of Tumor Deep Hyperthermia and Whole-Body Hyperthermia), Department of Oncology, Capital Medical University, Beijing, 100038, People's Republic of China
| |
Collapse
|
14
|
Bredeck G, Kämpfer AAM, Sofranko A, Wahle T, Büttner V, Albrecht C, Schins RPF. Ingested Engineered Nanomaterials Affect the Expression of Mucin Genes-An In Vitro-In Vivo Comparison. NANOMATERIALS 2021; 11:nano11102621. [PMID: 34685068 PMCID: PMC8537393 DOI: 10.3390/nano11102621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022]
Abstract
The increasing use of engineered nanomaterials (ENM) in food has fueled the development of intestinal in vitro models for toxicity testing. However, ENM effects on intestinal mucus have barely been addressed, although its crucial role for intestinal health is evident. We investigated the effects of ENM on mucin expression and aimed to evaluate the suitability of four in vitro models of increasing complexity compared to a mouse model exposed through feed pellets. We assessed the gene expression of the mucins MUC1, MUC2, MUC5AC, MUC13 and MUC20 and the chemokine interleukin-8 in pre-confluent and confluent HT29-MTX-E12 cells, in stable and inflamed triple cultures of Caco-2, HT29-MTX-E12 and THP-1 cells, and in the ileum of mice following exposure to TiO2, Ag, CeO2 or SiO2. All ENM had shared and specific effects. CeO2 downregulated MUC1 in confluent E12 cells and in mice. Ag induced downregulation of Muc2 in mice. Overall, the in vivo data were consistent with the findings in the stable triple cultures and the confluent HT29-MTX-E12 cells but not in pre-confluent cells, indicating the higher relevance of advanced models for hazard assessment. The effects on MUC1 and MUC2 suggest that specific ENM may lead to an elevated susceptibility towards intestinal infections and inflammations.
Collapse
|
15
|
Yang S, Yu M. Role of Goblet Cells in Intestinal Barrier and Mucosal Immunity. J Inflamm Res 2021; 14:3171-3183. [PMID: 34285541 PMCID: PMC8286120 DOI: 10.2147/jir.s318327] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022] Open
Abstract
Goblet cells and the mucus they secrete serve as an important barrier, preventing pathogens from invading the mucosa to cause intestinal inflammation. The perspective regarding goblet cells and mucus has changed, with current evidence suggesting that they are not passive but play a positive role in maintaining intestinal tract immunity and mucosal homeostasis. Goblet cells could obtain luminal antigens, presenting them to the underlying antigen-presenting cells (APCs) that induces adaptive immune responses. Various immunomodulatory factors can promote the differentiation and maturation of goblet cells, and the secretion of mucin. The abnormal proliferation and differentiation of goblet cells, as well as the deficiency synthesis and secretion of mucins, result in intestinal mucosal barrier dysfunction. This review provides an extensive outline of the signaling pathways that regulate goblet cell proliferation and differentiation and control mucins synthesis and secretion to elucidate how altering these pathways affects goblet functionality. Furthermore, the interaction between mucins and goblet cells in intestinal mucosal immunology is described. Therefore, the contribution of goblet cells and mucus in promoting gut defense and homeostasis is illustrated, while clarifying the regulatory mechanisms involved may allow the development of new therapeutic strategies for intestinal disorders.
Collapse
Affiliation(s)
- Songwei Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, People's Republic of China
| |
Collapse
|
16
|
Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut 2020; 69:2232-2243. [PMID: 32917747 PMCID: PMC7677487 DOI: 10.1136/gutjnl-2020-322260] [Citation(s) in RCA: 770] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 02/07/2023]
Abstract
The gastrointestinal tract is often considered as a key organ involved in the digestion of food and providing nutrients to the body for proper maintenance. However, this system is composed of organs that are extremely complex. Among the different parts, the intestine is viewed as an incredible surface of contact with the environment and is colonised by hundreds of trillions of gut microbes. The role of the gut barrier has been studied for decades, but the exact mechanisms involved in the protection of the gut barrier are various and complementary. Among them, the integrity of the mucus barrier is one of the first lines of protection of the gastrointestinal tract. In the past, this 'slimy' partner was mostly considered a simple lubricant for facilitating the progression of the food bolus and the stools in the gut. Since then, different researchers have made important progress, and currently, the regulation of this mucus barrier is gaining increasing attention from the scientific community. Among the factors influencing the mucus barrier, the microbiome plays a major role in driving mucus changes. Additionally, our dietary habits (ie, high-fat diet, low-fibre/high-fibre diet, food additives, pre- probiotics) influence the mucus at different levels. Given that the mucus layer has been linked with the appearance of diseases, proper knowledge is highly warranted. Here, we debate different aspects of the mucus layer by focusing on its chemical composition, regulation of synthesis and degradation by the microbiota as well as some characteristics of the mucus layer in both physiological and pathological situations.
Collapse
Affiliation(s)
- Paola Paone
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
17
|
Du X, Yang Y, Xiao G, Yang M, Yuan L, Qin L, He R, Wang L, Wu M, Wu S, Feng J, Xiang Y, Qu X, Liu H, Qin X, Liu C. Respiratory syncytial virus infection-induced mucus secretion by down-regulation of miR-34b/c-5p expression in airway epithelial cells. J Cell Mol Med 2020; 24:12694-12705. [PMID: 32939938 PMCID: PMC7687004 DOI: 10.1111/jcmm.15845] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/15/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Severe RSV infection is the main cause of hospitalization to children under the age of five. The regulation of miRNAs on the severity of RSV infection is unclear. The aim of the study was to identify the critical differential expression miRNAs (DE miRNAs) that can regulate the pathological response in RSV‐infected airway epithelial cells. In this study, miRNA and mRNA chips of RSV‐infected airway epithelia from Gene Expression Omnibus (GEO) were screened and analysed, separately. DE miRNAs‐targeted genes were performed for further pathway and process enrichment analysis. DE miRNA‐targeted gene functional network was constructed on the basis of miRNA‐mRNA interaction. The screened critical miRNA was also investigated by bioinformatics analysis. Then, RSV‐infected human bronchial epithelial cells (HBECs) were constructed to verify the expression of the DE miRNAs. Finally, specific synthetic DE miRNAs mimics were used to confirm the effect of DE miRNAs on the RSV‐infected HBECs. 45 DE miRNAs were identified from GEO62306 dataset. Our results showed that hsa‐mir‐34b‐5p and hsa‐mir‐34c‐5p decreased significantly in HBECs after RSV infection. Consistent with the biometric analysis, hsa‐mir‐34b/c‐5p is involved in the regulation of mucin expression gene MUC5AC. In RSV‐infected HBECs, the inducement of MUC5AC production by decreased hsa‐mir‐34b/c‐5p was partly mediated through activation of c‐Jun. These findings provide new insights into the mechanism of mucus obstruction after RSV infection and represent valuable targets for RSV infection and airway obstruction treatment.
Collapse
Affiliation(s)
- Xizi Du
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Yu Yang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia.,School of Basic Medical Sciences & Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Lin Yuan
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Ruoxi He
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Mengping Wu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - ShuangYan Wu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Juntao Feng
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Changsha, China
| | - Yang Xiang
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Huijun Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
18
|
Han HA, Pang JKS, Soh BS. Mitigating off-target effects in CRISPR/Cas9-mediated in vivo gene editing. J Mol Med (Berl) 2020; 98:615-632. [PMID: 32198625 PMCID: PMC7220873 DOI: 10.1007/s00109-020-01893-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/28/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022]
Abstract
The rapid advancement of genome editing technologies has opened up new possibilities in the field of medicine. Nuclease-based techniques such as the CRISPR/Cas9 system are now used to target genetically linked disorders that were previously hard-to-treat. The CRISPR/Cas9 gene editing approach wields several advantages over its contemporary editing systems, notably in the ease of component design, implementation and the option of multiplex genome editing. While results from the early phase clinical trials have been encouraging, the small patient population recruited into these trials hinders a conclusive assessment on the safety aspects of the CRISPR/Cas9 therapy. Potential safety concerns include the lack of fidelity in the CRISPR/Cas9 system which may lead to unintended DNA modifications at non-targeted gene loci. This review focuses modifications to the CRISPR/Cas9 components that can mitigate off-target effects in in vitro and preclinical models and its translatability to gene therapy in patient populations.
Collapse
Affiliation(s)
- Hua Alexander Han
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Jeremy Kah Sheng Pang
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.
- Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543, Singapore.
- Key Laboratory for Major Obstetric Disease of Guangdong Province, The Third Affliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
19
|
Sanaksenaho G, Mutanen A, Godbole N, Kyrönlahti A, Koivusalo A, Lohi J, Pihlajoki M, Heikinheimo M, Pakarinen MP. Parenteral Nutrition-Dependent Children With Short-Bowel Syndrome Lack Duodenal-Adaptive Hyperplasia but Show Molecular Signs of Altered Mucosal Function. JPEN J Parenter Enteral Nutr 2020; 44:1291-1300. [PMID: 31985858 DOI: 10.1002/jpen.1763] [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: 09/01/2019] [Revised: 10/30/2019] [Accepted: 12/04/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Although adaptive mucosal growth of the remaining small intestine is an essential compensatory mechanism to bowel resection in experimental short-bowel syndrome (SBS), only scarce clinical data are available. We studied structural and molecular mechanisms of intestinal adaptation in children with SBS. METHODS Fourteen patients, who had been dependent on parenteral nutrition (PN) since neonatal period for a median (interquartile range)1.4 (0.7-6.5) years, were studied at the age of 1.5 (1.0-6.5) years. Median length of remaining small bowel was 33 (12-60) cm, and 6 patients had their ileocecal valve preserved. Six children without gastrointestinal disorders served as age-matched and gender-matched controls. All patients underwent duodenal biopsies. Mucosal microarchitecture, proliferation, apoptosis, inflammation, and epithelial-barrier function were addressed using histology, immunohistochemistry, and quantitative real-time polymerase chain reaction. RESULTS Villus height, crypt depth, enterocyte proliferation, and apoptosis were similar in patients and matched controls. Messenger RNA (mRNA) expression of numerous genes regulating gut epithelial-barrier function (TGFB2, CAV1, CLDN1, MUC2, and NLRC4) was significantly altered. Of various nutrient transporters studied, only expression of SLC2A1 encoding facilitative glucose transporter GLUT1 was increased among patients, whereas RNA expression of genes encoding sodium-dependent glucose, sterol, fatty-acid, and peptide transport remained unchanged. CONCLUSION Duodenal mucosal hyperplasia has a limited role in mediating physiological adaptation following intestinal resection among PN-dependent children with SBS. Further clinical studies addressing functional significance of the observed alterations in mucosal RNA expression are warranted.
Collapse
Affiliation(s)
- Galina Sanaksenaho
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Annika Mutanen
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Nimish Godbole
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Kyrönlahti
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Antti Koivusalo
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jouko Lohi
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Marjut Pihlajoki
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Markku Heikinheimo
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko P Pakarinen
- Section of Pediatric Surgery, Pediatric Liver and Gut Research Group, Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
20
|
Carmicheal J, Atri P, Sharma S, Kumar S, Chirravuri Venkata R, Kulkarni P, Salgia R, Ghersi D, Kaur S, Batra SK. Presence and structure-activity relationship of intrinsically disordered regions across mucins. FASEB J 2020; 34:1939-1957. [PMID: 31908009 DOI: 10.1096/fj.201901898rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/18/2019] [Accepted: 12/05/2019] [Indexed: 12/24/2022]
Abstract
Many members of the mucin family are evolutionarily conserved and are often aberrantly expressed and glycosylated in various benign and malignant pathologies leading to tumor invasion, metastasis, and immune evasion. The large size and extensive glycosylation present challenges to study the mucin structure using traditional methods, including crystallography. We offer the hypothesis that the functional versatility of mucins may be attributed to the presence of intrinsically disordered regions (IDRs) that provide dynamism and flexibility and that the IDRs offer potential therapeutic targets. Herein, we examined the links between the mucin structure and function based on IDRs, posttranslational modifications (PTMs), and potential impact on their interactome. Using sequence-based bioinformatics tools, we observed that mucins are predicted to be moderately (20%-40%) to highly (>40%) disordered and many conserved mucin domains could be disordered. Phosphorylation sites overlap with IDRs throughout the mucin sequences. Additionally, the majority of predicted O- and N- glycosylation sites in the tandem repeat regions occur within IDRs and these IDRs contain a large number of functional motifs, that is, molecular recognition features (MoRFs), which directly influence protein-protein interactions (PPIs). This investigation provides a novel perspective and offers an insight into the complexity and dynamic nature of mucins.
Collapse
Affiliation(s)
- Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sunandini Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, California
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska Omaha, Omaha, Nebraska
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
21
|
Gan GL, Liu J, Chen WJ, Ye QQ, Xu Y, Wu HT, Li W. The Diverse Roles of the Mucin Gene Cluster Located on Chromosome 11p15.5 in Colorectal Cancer. Front Cell Dev Biol 2020; 8:514. [PMID: 32695780 PMCID: PMC7338833 DOI: 10.3389/fcell.2020.00514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/28/2020] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC), the third most common malignant tumor in the world, shows multiple complex and pathologies based on the impaired structure and function of the intestinal mucosal barrier. Goblet cells secrete mucins, which are involved in the formation of the intestinal mucosal barrier and not only lubricate and protect the intestinal mucosa but also participate in the processes of cell adhesion, intercellular signal transduction, and immune regulation. It is accepted that the disordered expression and dysfunction of mucins are associated with the occurrence and development of CRC. This article focuses on the secretory mucins encoded by a gene cluster located on chromosome 11p15.5 and systematically reviews their composition, regulation, function, and role in CRC, to deepen the understanding of the pathogeneses of CRC and to provide a new basis and ideas for the treatment of CRC.
Collapse
Affiliation(s)
- Guo-Lian Gan
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jing Liu
- Changjiang Scholar’s Laboratory/Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou University Medical College, Shantou, China
- Department of Physiology/Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Wen-Jia Chen
- Changjiang Scholar’s Laboratory/Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou University Medical College, Shantou, China
- Department of Physiology/Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Qian-Qian Ye
- Changjiang Scholar’s Laboratory/Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou University Medical College, Shantou, China
- Department of Physiology/Cancer Research Center, Shantou University Medical College, Shantou, China
| | - Ya Xu
- Changjiang Scholar’s Laboratory/Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Shantou University Medical College, Shantou, China
| | - Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- *Correspondence: Hua-Tao Wu,
| | - Wei Li
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Wei Li,
| |
Collapse
|
22
|
Zhou N, Shen Y, Fan L, Sun Q, Huang C, Hao J, Lan J, Yan H. The Characteristics of Intestinal-Barrier Damage in Rats With IgA Nephropathy. Am J Med Sci 2019; 359:168-176. [PMID: 32089158 DOI: 10.1016/j.amjms.2019.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intestinal-barrier damage plays an important pathogenic role in immunoglobulin A nephropathy (IgAN). In this study, we explored the characteristics of the intestinal barrier in rats with IgAN. MATERIALS AND METHODS We randomly divided 17 Sprague Dawley (SD) male rats into a normal control group (NC; n = 9) and an IgAN model group (n = 8). Feces in the distal ileum were taken for intestinal-microbiota 16sDNA sequencing. We also took a segment of terminal ileum to analyze intestinal morphology and to detect mRNA and protein expression of the tight-junction proteins zonula occludens-1 (ZO-1) and occludin (OCLN), as well as of mucin 2 (MUC2). We then measured levels of serum diamine oxidase (DAO) and D-lactic acid (D-LA), the biomarkers of intestinal permeability. RESULTS Compared with the NC group, mRNA expression levels of ZO-1 (t = 4.216, P = 0.0007), OCLN (t = 2.413, P = 0.029) and MUC2 (t = 0.859, P < 0.0001) were significantly decreased in the IgAN model group. Protein expression of ZO-1 (t = 7.349, P < 0.0001) and OCLN (t = 6.367, P < 0.0001) was also decreased in the IgAN model group. Conversely, serum DAO (t = 3.758, P = 0.0024) and D-LA (t = 2.246, P = 0.0427) levels increased in this group. At the genus level, the relative abundance of Ruminococcus2 (P = 0.0086) was increased in the IgAN model group. CONCLUSIONS Decreased expression of ZO-1, OCLN and MUC2, plus intestinal-microbiota dysbiosis, are associated with intestinal-barrier damage in IgAN rats.
Collapse
Affiliation(s)
- Nan Zhou
- Department of Nephrology,; National Center for Children's Health (Beijing), Beijing, China; Key Laboratory of Chronic Kidney Disease and Blood Purification in Childhood (Beijing), Beijing, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Ying Shen
- Department of Nephrology,; National Center for Children's Health (Beijing), Beijing, China; Key Laboratory of Chronic Kidney Disease and Blood Purification in Childhood (Beijing), Beijing, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Lirong Fan
- Department of Traditional Chinese Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health (Beijing), Beijing, China
| | - Qiang Sun
- Department of Nephrology,; National Center for Children's Health (Beijing), Beijing, China; Key Laboratory of Chronic Kidney Disease and Blood Purification in Childhood (Beijing), Beijing, China; Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| | - Canxing Huang
- Department of Neonatology, Longyan People's Hospital, Longyan City, Fujian, China
| | - Jing Hao
- Department of Traditional Chinese Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health (Beijing), Beijing, China
| | - Jingchao Lan
- Department of Nephrology,; National Center for Children's Health (Beijing), Beijing, China
| | - Huimin Yan
- Department of Traditional Chinese Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health (Beijing), Beijing, China.
| |
Collapse
|
23
|
Baněčková M, Michal M, Laco J, Leivo I, Ptáková N, Horáková M, Michal M, Skálová A. Immunohistochemical and genetic analysis of respiratory epithelial adenomatoid hamartomas and seromucinous hamartomas: are they precursor lesions to sinonasal low-grade tubulopapillary adenocarcinomas? Hum Pathol 2019; 97:94-102. [PMID: 31698004 DOI: 10.1016/j.humpath.2019.09.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 01/05/2023]
Abstract
Respiratory epithelial adenomatoid hamartoma (REAH) and seromucinous hamartoma (SH) are rare tumor-like lesions of the nasal cavity, paranasal sinuses, and nasopharynx. The pathogenesis of REAH/SH is still unclear. Neoplastic proliferation, chronic mechanical irritation, inflammation, or possible embryological tissue misplacement are speculated as possible mechanisms of their development. Low-grade tubulopapillary adenocarcinoma (LGTA) is a rare variant of nonsalivary, nonintestinal type sinonasal adenocarcinoma. The aim of this study was to evaluate the immunohistochemical and genetic profiles of 10 cases of REAH/SH, with serous, mucinous, and respiratory components evaluated separately and to compare these findings with the features of 9 cases of LGTA. All cases of REAH/SH and LGTA were analyzed immunohistochemically with a cocktail of mucin antigens (MUC1, MUC2, MUC4, MUC5AC, MUC6) and with epithelial (CK7, CK20, CDX2, SATB2) and myoepithelial markers (S100 protein, p63, SOX10). The next-generation sequencing assay was performed using FusionPlex Solid Tumor Kit (ArcherDx) in 10 cases of REAH/SH, and the EGFR-ZNF267 gene fusion was detected in 1 of them. Two female REAH/SH cases were assessed for the presence of clonality. Using the human androgen receptor assay, 1 case was proved to be clonal. The serous component of REAH/SH was positive for CK7/MUC1 and SOX10 similarly to LGTA. Although REAH/SH and LGTA are histopathologically and clinically separate entities, the overlap in their morphological and immunohistochemical profiles suggests that REAH/SH might be a precursor lesion of LGTA.
Collapse
Affiliation(s)
- Martina Baněčková
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen 30605, Czech Republic; Bioptic Laboratory, ltd, Plzen 32600, Czech Republic.
| | - Michael Michal
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen 30605, Czech Republic; Bioptic Laboratory, ltd, Plzen 32600, Czech Republic; Biomedical Center, Charles University, Faculty of Medicine in Plzen, Plzen 32300, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine and University Hospital, Hradec Kralove 50005, Czech Republic
| | - Ilmo Leivo
- Institute of Biomedicine, Pathology, University of Turku, and Turku University Hospital, Turku 50521, Finland
| | | | - Markéta Horáková
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen 30605, Czech Republic; Bioptic Laboratory, ltd, Plzen 32600, Czech Republic
| | - Michal Michal
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen 30605, Czech Republic; Bioptic Laboratory, ltd, Plzen 32600, Czech Republic
| | - Alena Skálová
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Plzen 30605, Czech Republic; Bioptic Laboratory, ltd, Plzen 32600, Czech Republic
| |
Collapse
|
24
|
Intestinal Barrier Function in Gluten-Related Disorders. Nutrients 2019; 11:nu11102325. [PMID: 31581491 PMCID: PMC6835310 DOI: 10.3390/nu11102325] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022] Open
Abstract
Gluten-related disorders include distinct disease entities, namely celiac disease, wheat-associated allergy and non-celiac gluten/wheat sensitivity. Despite having in common the contact of the gastrointestinal mucosa with components of wheat and other cereals as a causative factor, these clinical entities have distinct pathophysiological pathways. In celiac disease, a T-cell mediate immune reaction triggered by gluten ingestion is central in the pathogenesis of the enteropathy, while wheat allergy develops as a rapid immunoglobulin E- or non-immunoglobulin E-mediated immune response. In non-celiac wheat sensitivity, classical adaptive immune responses are not involved. Instead, recent research has revealed that an innate immune response to a yet-to-be-defined antigen, as well as the gut microbiota, are pivotal in the development in this disorder. Although impairment of the epithelial barrier has been described in all three clinical conditions, its role as a potential pathogenetic co-factor, specifically in celiac disease and non-celiac wheat sensitivity, is still a matter of investigation. This article gives a short overview of the mucosal barrier of the small intestine, summarizes the aspects of barrier dysfunction observed in all three gluten-related disorders and reviews literature data in favor of a primary involvement of the epithelial barrier in the development of celiac disease and non-celiac wheat sensitivity.
Collapse
|