1
|
Long L, Li M, Wang M, Liang B, Huang M, Yuan X, Wu X, Guo X, Li S, Liu Z, Liu W, Chen W, Wang W, Lyu Q, Li C. Activation of mannose receptor C type 1 in macrophages improves renal fibrosis through mediating fibronectin endocytosis. Life Sci 2025; 371:123593. [PMID: 40164332 DOI: 10.1016/j.lfs.2025.123593] [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: 01/21/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
AIMS Excess extracellular matrix (ECM) deposition is the characteristic of renal fibrosis, owing to the imbalance between synthesis and degradation. Fibronectin could regulate the deposition of other ECM, thus plays a crucial role in the progression of renal fibrosis. Mannose receptor C type 1 (MRC1), largely expressed on macrophages, owns an extracellular fibronectin type II domain that binds to and internalizes collagen and thus involves in fibrosis modulation. The purpose of the present study was to investigate whether MRC1 participates in the internalization of fibronectin and whether alginate oligosaccharides (AOSC), a degradation product of alginate, has beneficial effects in the resolution of renal fibrosis via MRC1. MATERIALS AND METHODS Renal fibrosis models were constructed by unilateral ureteral obstruction (UUO) and unilateral ischemia-reperfusion injury (UIRI) in MRC1-WT and MRC1-KO mice. RAW264.7 cells were treated with TGF-β1 to induce pro-fibrotic responses. Expression of fibrotic markers and fibronectin endocytosis were examined. KEY FINDINGS MRC1 gene knockout aggravated renal fibrosis in UUO and UIRI models. Inhibition of MRC1 exacerbated TGF-β1-induced pro-fibrotic responses in RAW264.7 cells. MRC1 regulated integrin β1-mediated fibronectin endocytosis through Arp2/3-Kindlin-2 signaling pathway. AOSC improved renal fibrosis by increasing MRC1 expression and endocytosis of fibronectin. SIGNIFICANCE Our findings highlight the importance of MRC1 and fibronectin endocytosis in the development of renal fibrosis, suggesting that activation of MRC1 by AOSC is probably a therapeutic option to delay the progress of kidney fibrosis.
Collapse
Affiliation(s)
- Luosha Long
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathology and Pathophysiology, Pu Ai Medical School, Shaoyang University, Shaoyang, China
| | - Meng Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Minghui Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baien Liang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Meiying Huang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xi Yuan
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinyan Wu
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiangdong Guo
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Suchun Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, China
| | - Zhende Liu
- Haitang (Jiangsu) Biotechnology Co, Ltd., Nantong, Jiangsu, China
| | - Weizhi Liu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Nephrology, National Health Commission and Guangdong Province, Guangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qianqian Lyu
- Fang Zongxi Center, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| | - Chunling Li
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Wei ZX, Jiang SH, Qi XY, Cheng YM, Liu Q, Hou XY, He J. scRNA-seq of the intestine reveals the key role of mast cells in early gut dysfunction associated with acute pancreatitis. World J Gastroenterol 2025; 31:103094. [PMID: 40182603 PMCID: PMC11962851 DOI: 10.3748/wjg.v31.i12.103094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Intestinal barrier dysfunction is a prevalent and varied manifestation of acute pancreatitis (AP). Molecular mechanisms underlying the early intestinal barrier in AP remain poorly understood. AIM To explore the biological processes and mechanisms of intestinal injury associated with AP, and to find potential targets for early prevention or treatment of intestinal barrier injury. METHODS This study utilized single-cell RNA sequencing of the small intestine, alongside in vitro and in vivo experiments, to examine intestinal barrier function homeostasis during the early stages of AP and explore involved biological processes and potential mechanisms. RESULTS Seventeen major cell types and 33232 cells were identified across all samples, including normal, AP1 (4x caerulein injections, animals sacrificed 2 h after the last injection), and AP2 (8x caerulein injections, animals sacrificed 4 h after the last injection). An average of 980 genes per cell was found in the normal intestine, compared to 927 in the AP1 intestine and 1382 in the AP2 intestine. B cells, dendritic cells, mast cells (MCs), and monocytes in AP1 and AP2 showed reduced numbers compared to the normal intestine. Enterocytes, brush cells, enteroendocrine cells, and goblet cells maintained numbers similar to the normal intestine, while cytotoxic T cells and natural killer (NK) cells increased. Enterocytes in early AP exhibited elevated programmed cell death and intestinal barrier dysfunction but retained absorption capabilities. Cytotoxic T cells and NK cells showed enhanced pathogen-fighting abilities. Activated MCs, secreted chemokine (C-C motif) ligand 5 (CCL5), promoted neutrophil and macrophage infiltration and contributed to barrier dysfunction. CONCLUSION These findings enrich our understanding of biological processes and mechanisms in AP-associated intestinal injury, suggesting that CCL5 from MCs is a potential target for addressing dysfunction.
Collapse
Affiliation(s)
- Zu-Xing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shi-He Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Yan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yi-Miao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Qiong Liu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xu-Yang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
3
|
Dong L, Dong F, Guo P, Li T, Fang Y, Dong Y, Xu X, Cai T, Liang S, Song X, Li L, Sun W, Zheng Y. Gut microbiota as a new target for hyperuricemia: A perspective from natural plant products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156402. [PMID: 39874797 DOI: 10.1016/j.phymed.2025.156402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/29/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Hyperuricemia, a prevalent chronic metabolic disorder caused by purine metabolism disturbances, is characterized by elevated serum uric acid (UA) levels. Prolonged hyperuricemia can cause severe complications such as gout or kidney damage. However, the toxic side effects of and adverse reactions to UA-lowering drugs are becoming increasingly prominent. Therefore, new targets and drugs for hyperuricemia are needed. PURPOSE This review aims to summarize recent research progress on the prevention and treatment mechanisms for gut microbiota-hyperuricemia from the perspective of plant-derived natural products. METHODS Data from PubMed, Web of Science, ScienceDirect, and the CNKI databases spanning from January 2020 to December 2024 were reviewed. The aim of this study is to categorize and summarize the relevant mechanisms through which natural products improve hyperuricemia via the gut microbiota. The retrieved data followed PRISMA criteria (Preferred Reporting Items for Systematic reviews and Meta-Analyses). RESULTS Regulating gut microbiota as a treatment for hyperuricemia. Targeting the gut microbiota could reduce host UA levels by promoting purine degradation, reducing UA production, and increasing UA excretion. Moreover, the gut microbiota also exerts anti-inflammatory and antioxidant effects that alleviate complications such as renal damage caused by hyperuricemia. Due to their diverse sources, multicomponent synergy, multitarget effects, and minimal side effects, plant-derived natural products have been extensively utilized in the management of hyperuricemia. Especially, utilizing natural products from plants to regulate the gut microbiota has become a new strategy for reducing UA levels. CONCLUSION This review comprehensively summarizes recent advances in understanding the preventive and therapeutic mechanisms of plant-derived natural products in ameliorating hyperuricemia and its comorbidities through gut microbiota modulation. This review contributes a novel perspective for the development of safer and more efficacious UA-lowering products.
Collapse
Affiliation(s)
- Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Fengying Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Pingping Guo
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China; Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100007, China
| | - Yini Fang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China; Basic Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yang Dong
- Monitoring and Statistical Research Center, National Administration of Traditional Chinese Medicine, Beijing, 100021, China
| | - Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, China.
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, China.
| |
Collapse
|
4
|
Du Y, Fan Y, Li X, Chen F. Novel anti-inflammatory properties of mannose oligosaccharides in the treatment of inflammatory bowel disease via LGALS3 modulation. NPJ Biofilms Microbiomes 2025; 11:26. [PMID: 39920168 PMCID: PMC11806110 DOI: 10.1038/s41522-025-00648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025] Open
Abstract
This study investigates the role of Gum Arabic Mannose Oligosaccharides (GA-MOS) in modulating gut microbiota and alleviating symptoms of Inflammatory Bowel Disease (IBD). Employing both in vitro and in vivo models, we explored how GA-MOS influences microbial communities, particularly focusing on their capacity to enhance health-associated bacteria and reduce pathogenic species within the gut environment. Our findings reveal that GA-MOS treatment significantly altered the gut microbiota composition, increasing the abundance of anti-inflammatory bacteria while decreasing pro-inflammatory species, thus contributing to a reduction in gut inflammation and an improvement in intestinal barrier function. Detailed molecular analyses further demonstrated that these changes in microbiota were associated with modifications in the host's immune response, particularly through the suppression of key inflammatory pathways and cytokines involved in IBD progression. These results underscore the potential of dietary polysaccharides like GA-MOS as therapeutic agents in managing dysbiosis and inflammatory conditions in the gut, offering a promising approach for enhancing microbial health and overall disease management in IBD. This study provides novel insights into the bioactive properties of MOS and their interactions with gut microbiota, suggesting broader implications for their use in microbiome-centered therapies.
Collapse
Affiliation(s)
- Yaqi Du
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, P.R. China
| | - Xin Li
- Department of Gastrointestinal Surgery, The First Hospital, China Medical University, Shenyang, P.R. China.
| | - Fenqin Chen
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, P.R. China.
| |
Collapse
|
5
|
Zhao H, Zhang Z, Liu H, Ma M, Sun P, Zhao Y, Liu X. Multi-omics perspective: mechanisms of gastrointestinal injury repair. BURNS & TRAUMA 2025; 13:tkae057. [PMID: 39845194 PMCID: PMC11752642 DOI: 10.1093/burnst/tkae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 01/24/2025]
Abstract
In this review, we examine the significance of multi-omics technologies in understanding the plethora of intricate processes that activate gastrointestinal (GI) injury repair. Multi-omics, which includes genomics, transcriptomics, proteomics, and metabolomics, allows intricate mapping of cellular responses and molecular pathways involved in GI repair. We highlight the potential of multi-omics to discover previously unknown therapeutic targets or elucidate the molecular basis of the pathogenesis of GI. Furthermore, we explore the possibilities of integrating omics data to improve prediction models, and summarize the state-of-the-art technological developments and persisting obstacles that hinder the translation of multi-omics into clinical practice. Finally, innovative multi-omics approaches that can improve patient outcomes and advance therapeutic strategies in GI medicine are discussed.
Collapse
Affiliation(s)
- Haibin Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Zhigang Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Hongyu Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Mingxiu Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Peng Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Yang Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| |
Collapse
|
6
|
Zhou Y, Wei Z, Gao Y, Zhang H, Schroyen M. The role of alginate oligosaccharide on boar semen quality: A research review. Int J Biol Macromol 2024; 277:134492. [PMID: 39106929 DOI: 10.1016/j.ijbiomac.2024.134492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/24/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
Alginate is the general term of a polysaccharide which is widely used in the area of pharmaceutics and the food industry and is known for its unique biological activities. However, due to the low water solubility and large viscosity of alginate, its development and utilization in the agricultural field are limited. Alginate oligosaccharide (AOS) is a degradable product derived from alginate and has attracted much attention in recent years because of its specific characteristics such as a low molecular weight, high water solubility, and non-toxicity. Boar semen quality, which is affected by various factors, is an important indicator for measuring reproductive performance of boars. With the development of artificial insemination technology, high quality semen has been more and more important. Therefore, increasing semen quality is an important means to improve the reproductive performance in swine industry. In this research review, we used the PubMed database and Google Scholar and web of science to search for relevant literature on the topic of AOS in relation to boar semen quality. Key words used were alginate oligosaccharide, boars, semen quality, microbiota and metabolites. The purpose of this review article was to describe the current knowledge on the relationship between AOS and boar semen quality, and provide an overview of solutions for the decline in the boar semen quality in specific conditions. Based on the existing literature, it is evident that AOS can be used as a new type of food additive. This review paper provides a theoretical basis for the production of high-quality boar sperm and, suggests that, in the future, AOS can even aid in treating human infertility.
Collapse
Affiliation(s)
- Yexun Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Zeou Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; School of Agriculture and Food Science, University College Dublin, Belfeld, Dublin 4, Ireland
| | - Yang Gao
- College of Life Science, Baicheng Normal University, Baicheng, Jilin 137000, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
7
|
Wang J, Zhang Z, Dai T, Zhang Z, Zhang Q, Yao J, Wang L, He N, Li S. The therapeutic effect and possible mechanisms of alginate oligosaccharide on metabolic syndrome by regulating gut microbiota. Food Funct 2024; 15:9632-9661. [PMID: 39239698 DOI: 10.1039/d4fo02802c] [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: 09/07/2024]
Abstract
Metabolic syndrome (MetS) is a disease condition incorporating the abnormal accumulation of various metabolic components, including overweight or abdominal obesity, insulin resistance and abnormal glucose tolerance, hypertension, atherosclerosis, or dyslipidemia. It has been proved that the gut microbiota and microbial-derived products play an important role in regulating lipid metabolism and thus the onset and development of MetS. Previous studies have demonstrated that oligosaccharides with prebiotic effects, such as chitosan oligosaccharides, can regulate the structure of the microbial community and its derived products to control weight and reduce MetS associated with obesity. Alginate oligosaccharides (AOS), natural products extracted from degraded alginate salts with high solubility and extensive biological activity, have also been found to modulate gut microbiota. This review aims to summarize experimental evidence on the positive effects of AOS on different types of MetS while providing insights into mechanisms through which AOS regulates gut microbiota for preventing and treating MetS.
Collapse
Affiliation(s)
- Jingyi Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
| | - Zixuan Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Ziheng Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Qingfeng Zhang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Jingtong Yao
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Lijing Wang
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, 266000, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao, University, Qingdao 266071, China.
| |
Collapse
|
8
|
Teng Y, Li J, Guo J, Yan C, Wang A, Xia X. Alginate oligosaccharide improves 5-fluorouracil-induced intestinal mucositis by enhancing intestinal barrier and modulating intestinal levels of butyrate and isovalerate. Int J Biol Macromol 2024; 276:133699. [PMID: 38972652 DOI: 10.1016/j.ijbiomac.2024.133699] [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: 01/16/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Chemotherapy-induced mucositis (CIM) is the typical side effect of chemotherapy. This study investigates the potential of alginate oligosaccharide (AOS) in ameliorating CIM induced by 5-fluorouracil (5-FU) in a murine model and its underlying mechanisms. AOS effectively mitigated body weight loss and histopathological damage, modulated inflammatory cytokines and attenuated the oxidative stress. AOS restored intestinal barrier integrity through enhancing expression of tight junction proteins via MLCK signaling pathway. AOS alleviated intestinal mucosal damage by inhibiting TLR4/MyD88/NF-κB signaling pathway, downregulating the pro-apoptotic protein Bax and upregulating the anti-apoptotic protein Bcl-2. Moreover, AOS significantly enriched intestinal Akkermansiaceae and increased the production of short-chain fatty acids (SCFAs), most notably butyrate and isovalerate. Pre-treatment with butyrate and isovalerate also alleviated 5-FU-induced CIM. In conclusion, AOS effectively mitigated CIM through strenghthening intestinal barrier, attenuating inflammation, and modulating gut microbiota and intestianl levels of butyrate and isovalerate. These finding indicate that AOS could be potentially utilized as a supplemental strategy for prevention or mitigation of CIM.
Collapse
Affiliation(s)
- Yue Teng
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiahui Li
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jian Guo
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Ailing Wang
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
9
|
Liu M, Deng X, Zhao Y, Everaert N, Zhang H, Xia B, Schroyen M. Alginate Oligosaccharides Enhance Antioxidant Status and Intestinal Health by Modulating the Gut Microbiota in Weaned Piglets. Int J Mol Sci 2024; 25:8029. [PMID: 39125598 PMCID: PMC11311613 DOI: 10.3390/ijms25158029] [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: 06/16/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Alginate oligosaccharides (AOSs), which are an attractive feed additive for animal production, exhibit pleiotropic bioactivities. In the present study, we investigated graded doses of AOS-mediated alterations in the physiological responses of piglets by determining the intestinal architecture, barrier function, and microbiota. A total of 144 weaned piglets were allocated into four dietary treatments in a completely random design, which included a control diet (CON) and three treated diets formulated with 250 mg/kg (AOS250), 500 mg/kg (AOS500), and 1000 mg/kg AOS (AOS1000), respectively. The trial was carried out for 28 days. Our results showed that AOS treatment reinforced the intestinal barrier function by increasing the ileal villus height, density, and fold, as well as the expression of tight junction proteins, especially at the dose of 500 mg/kg AOS. Meanwhile, supplementations with AOSs showed positive effects on enhancing antioxidant capacity and alleviating intestinal inflammation by elevating the levels of antioxidant enzymes and inhibiting excessive inflammatory cytokines. The DESeq2 analysis showed that AOS supplementation inhibited the growth of harmful bacteria Helicobacter and Escherichia_Shigella and enhanced the relative abundance of Faecalibacterium and Veillonella. Collectively, these findings suggested that AOSs have beneficial effects on growth performance, antioxidant capacity, and gut health in piglets.
Collapse
Affiliation(s)
- Ming Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (M.L.)
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés 2, 5030 Gembloux, Belgium
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China (H.Z.)
| | - Xiong Deng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (M.L.)
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China (H.Z.)
| | - Nadia Everaert
- Nutrition and Animal Microbiota Ecosystems Laboratory, Department of Biosystems, KU Leuven, 3001 Leuven, Belgium
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China (H.Z.)
| | - Bing Xia
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China; (M.L.)
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés 2, 5030 Gembloux, Belgium
| |
Collapse
|
10
|
Ma XQ, Wang B, Wei W, Tan FC, Su H, Zhang JZ, Zhao CY, Zheng HJ, Feng YQ, Shen W, Yang JB, Li FL. Alginate oligosaccharide assimilation by gut microorganisms and the potential role in gut inflammation alleviation. Appl Environ Microbiol 2024; 90:e0004624. [PMID: 38563787 PMCID: PMC11107165 DOI: 10.1128/aem.00046-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
Dietary fiber metabolism by gut microorganisms plays important roles in host physiology and health. Alginate, the major dietary fiber of daily diet seaweeds, is drawing more attention because of multiple biological activities. To advance the understanding of alginate assimilation mechanism in the gut, we show the presence of unsaturated alginate oligosaccharides (uAOS)-specific alginate utilization loci (AUL) in human gut microbiome. As a representative example, a working model of the AUL from the gut microorganism Bacteroides clarus was reconstructed from biochemistry and transcriptome data. The fermentation of resulting monosaccharides through Entner-Doudoroff pathway tunes the metabolism of short-chain fatty acids and amino acids. Furthermore, we show that uAOS feeding protects the mice against dextran sulfate sodium-induced acute colitis probably by remodeling gut microbiota and metabolome. IMPORTANCE Alginate has been included in traditional Chinese medicine and daily diet for centuries. Recently discovered biological activities suggested that alginate-derived alginate oligosaccharides (AOS) might be an active ingredient in traditional Chinese medicine, but how these AOS are metabolized in the gut and how it affects health need more information. The study on the working mechanism of alginate utilization loci (AUL) by the gut microorganism uncovers the role of unsaturated alginate oligosaccharides (uAOS) assimilation in tuning short-chain fatty acids and amino acids metabolism and demonstrates that uAOS metabolism by gut microorganisms results in a variation of cell metabolites, which potentially contributes to the physiology and health of gut.
Collapse
Affiliation(s)
- Xiao-Qing Ma
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Bing Wang
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Wei Wei
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Fang-Cheng Tan
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Hang Su
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Jun-Zhe Zhang
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Chen-Yang Zhao
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Hua-Jun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Yan-Qin Feng
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Jin-Bo Yang
- Innovation Center of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Fu-Li Li
- Qingdao C1 Refinery Engineering Research Center, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| |
Collapse
|
11
|
Bao L, Fu L, Su Y, Chen Z, Peng Z, Sun L, Gonzalez FJ, Wu C, Zhang H, Shi B, Shi YB. Amino acid transporter SLC7A5 regulates cell proliferation and secretary cell differentiation and distribution in the mouse intestine. Int J Biol Sci 2024; 20:2187-2201. [PMID: 38617535 PMCID: PMC11008275 DOI: 10.7150/ijbs.94297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/16/2024] [Indexed: 04/16/2024] Open
Abstract
The intestine is critical for not only processing nutrients but also protecting the organism from the environment. These functions are mainly carried out by the epithelium, which is constantly being self-renewed. Many genes and pathways can influence intestinal epithelial cell proliferation. Among them is mTORC1, whose activation increases cell proliferation. Here, we report the first intestinal epithelial cell (IEC)-specific knockout (ΔIEC) of an amino acid transporter capable of activating mTORC1. We show that the transporter, SLC7A5, is highly expressed in mouse intestinal crypt and Slc7a5ΔIEC reduces mTORC1 signaling. Surprisingly, adult Slc7a5ΔIEC intestinal crypts have increased cell proliferation but reduced mature Paneth cells. Goblet cells, the other major secretory cell type in the small intestine, are increased in the crypts but reduced in the villi. Analyses with scRNA-seq and electron microscopy have revealed dedifferentiation of Paneth cells in Slc7a5ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. Thus, SLC7A5 likely regulates secretory cell differentiation to affect stem cell niche and indirectly regulate cell proliferation.
Collapse
Affiliation(s)
- Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Liezhen Fu
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| | - Yijun Su
- Laboratory of High Resolution Optical Imaging and Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
- Janelia Research Campus, Howard Hughes Medical Institute (HHMI), Ashburn, VA, USA
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhaoyi Peng
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongen Zhang
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University School of Medicine. No.277, Yanta West Road, Xi'an, Shaanxi, 710061, P.R. China
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD, USA
| |
Collapse
|
12
|
Krishna Perumal P, Huang CY, Chen CW, Anisha GS, Singhania RR, Dong CD, Patel AK. Advances in oligosaccharides production from brown seaweeds: extraction, characterization, antimetabolic syndrome, and other potential applications. Bioengineered 2023; 14:2252659. [PMID: 37726874 PMCID: PMC10512857 DOI: 10.1080/21655979.2023.2252659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 09/21/2023] Open
Abstract
Brown seaweeds are a promising source of bioactive substances, particularly oligosaccharides. This group has recently gained considerable attention due to its diverse cell wall composition, structure, and wide-spectrum bioactivities. This review article provides a comprehensive update on advances in oligosaccharides (OSs) production from brown seaweeds and their potential health applications. It focuses on advances in feedstock pretreatment, extraction, characterization, and purification prior to OS use for potential health applications. Brown seaweed oligosaccharides (BSOSs) are extracted using various methods. Among these, enzymatic hydrolysis is the most preferred, with high specificity, mild reaction conditions, and low energy consumption. However, the enzyme selection and hydrolysis conditions need to be optimized for desirable yield and oligosaccharides composition. Characterization of oligosaccharides is essential to determine their structure and properties related to bioactivities and to predict their most suitable application. This is well covered in this review. Analytical techniques such as high-performance liquid chromatography (HPLC), gas chromatography (GC), and nuclear magnetic resonance (NMR) spectroscopy are commonly applied to analyze oligosaccharides. BSOSs exhibit a range of biological properties, mainly antimicrobial, anti-inflammatory, and prebiotic properties among others. Importantly, BSOSs have been linked to possible health advantages, including metabolic syndrome management. Metabolic syndrome is a cluster of conditions, such as obesity, hypertension, and dyslipidemia, which increase the risk of cardiovascular disease and type 2 diabetes. Furthermore, oligosaccharides have potential applications in the food and pharmaceutical industries. Future research should focus on improving industrial-scale oligosaccharide extraction and purification, as well as researching their potential utility in the treatment of various health disorders.[Figure: see text].
Collapse
Affiliation(s)
- Pitchurajan Krishna Perumal
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Chun-Yung Huang
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Chiu-Wen Chen
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Sustainable Environment Research Center, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Department of Marine Environmental Engineering, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
| | - Grace Sathyanesan Anisha
- Post-Graduate and Research Department of Zoology, Government College for Women, Thiruvananthapuram, India
| | - Reeta Rani Singhania
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| | - Cheng-Di Dong
- Sustainable Environment Research Center, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Department of Marine Environmental Engineering, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| | - Anil Kumar Patel
- Institute of Aquatic Science and Technology, College of Hydrosphere, National Kaohsiung University of Science and Technology, Kaohsiung City, Taiwan
- Centre for Energy and Environmental Sustainability, Lucknow, Uttar Pradesh, India
| |
Collapse
|
13
|
Mavrogeni ME, Asadpoor M, Judernatz JH, van Ark I, Wösten MMSM, Strijbis K, Pieters RJ, Folkerts G, Braber S. Protective Effects of Alginate and Chitosan Oligosaccharides against Clostridioides difficile Bacteria and Toxin. Toxins (Basel) 2023; 15:586. [PMID: 37888617 PMCID: PMC10610568 DOI: 10.3390/toxins15100586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023] Open
Abstract
Clostridioides difficile infection is expected to become the most common healthcare-associated infection worldwide. C. difficile-induced pathogenicity is significantly attributed to its enterotoxin, TcdA, which primarily targets Rho-GTPases involved in regulating cytoskeletal and tight junction (TJ) dynamics, thus leading to cytoskeleton breakdown and ultimately increased intestinal permeability. This study investigated whether two non-digestible oligosaccharides (NDOs), alginate (AOS) and chitosan (COS) oligosaccharides, possess antipathogenic and barrier-protective properties against C. difficile bacteria and TcdA toxin, respectively. Both NDOs significantly reduced C. difficile growth, while cell cytotoxicity assays demonstrated that neither COS nor AOS significantly attenuated the TcdA-induced cell death 24 h post-exposure. The challenge of Caco-2 monolayers with increasing TcdA concentrations increased paracellular permeability, as measured by TEER and LY flux assays. In this experimental setup, COS completely abolished, and AOS mitigated, the deleterious effects of TcdA on the monolayer's integrity. These events were not accompanied by alterations in ZO-1 and occludin protein levels; however, immunofluorescence microscopy revealed that both AOS and COS prevented the TcdA-induced occludin mislocalization. Finally, both NDOs accelerated TJ reassembly upon a calcium-switch assay. Overall, this study established the antipathogenic and barrier-protective capacity of AOS and COS against C. difficile and its toxin, TcdA, while revealing their ability to promote TJ reassembly in Caco-2 cells.
Collapse
Affiliation(s)
- Maria Eleni Mavrogeni
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Jo H Judernatz
- Structural Biochemistry Group, Bijvoet Centre for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Ingrid van Ark
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Marc M S M Wösten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Karin Strijbis
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - Roland J Pieters
- Division of Medicinal Chemistry and Chemical Biology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
14
|
Li L, Jiang J, Yao Z, Zhu B. Recent advances in the production, properties and applications of alginate oligosaccharides - a mini review. World J Microbiol Biotechnol 2023; 39:207. [PMID: 37221433 DOI: 10.1007/s11274-023-03658-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/20/2023] [Indexed: 05/25/2023]
Abstract
Alginate oligosaccharides (AOS) made from the degradation of alginate, to some extent, makes up for the poor solubility and bioavailability of alginate as a macromolecular substance and possess several beneficial biological activities that are absent in alginate. These properties include prebiotic, glycolipid regulatory, immunomodulatory, antimicrobial, antioxidant, anti-tumor, promoting plant growth and other activities. Consequently, AOS has significant potential for use in the agricultural, biomedical, and food industries, and has been the focus of research in the field of marine biological resources. This review comprehensively covers methods (physical, chemical, and enzymatic methods) for the production of AOS from alginate. More importantly, this paper reviews recent advances in the biological activity and potentially industrial and therapeutic applications of AOS, providing a reference for future research and applications of AOS.
Collapse
Affiliation(s)
- Li Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China
| | - Jinju Jiang
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao, 266400, China
| | - Zhong Yao
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China
| | - Benwei Zhu
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, 211816, China.
| |
Collapse
|
15
|
Zhou J, Qaing S, Yang B, Wang Y, Wang J, Yang T, Zhang Y, Chen Y, Li S. Cold plasma treatment with alginate oligosaccharide improves the digestive stability and bioavailability of nutrient-delivered particles: An in vitro INFOGEST gastrointestinal study. Int J Biol Macromol 2023; 232:123309. [PMID: 36652987 DOI: 10.1016/j.ijbiomac.2023.123309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
To improve the stability and bioavailability of the delivered hydrophobic nutrients, the zein-based delivery system was modified by alginate oligosaccharide (AOS), cold plasma (CP) treatments, and synergistically. The digestive behavior of each was investigated in an INFOGEST static in vitro digestion model. The results showed that AOS and CP treatments and their synergistic effects improved the dispersion and stability of the delivery system, leading to a more concentrated particle size distribution and higher particle surface charge. Both CP treatments and AOS increased the release rate of Curcumin (Cur) at small intestine (11.8 % to 20.5 % and 11.8 % to 24.64 %, respectively), and the synergistic effect was higher (11.8 % to 43.84 %). The wall material modified showed a higher encapsulation efficiency of Cur (52.83 % to 85.17 %). Cur release rate measurements showed that the wall material modified could have a positive effect on the slow release of Cur. SDS-page electrophoresis revealed that the slow release was due to the enhanced resistance of wall material to digestive fluids. Thus, treatment with AOS and CP treatments, and the synergism are suitable for modifying zein-based delivery systems for the encapsulation, stabilization, and slow release of hydrophobic nutrients during digestion in the field of functional foods.
Collapse
Affiliation(s)
- Junjun Zhou
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Siqi Qaing
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Bowen Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuhe Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jiake Wang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tongliang Yang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yifu Zhang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Ye Chen
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shuhong Li
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Quality and Health, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China.
| |
Collapse
|
16
|
Lu S, Na K, Wei J, Tao T, Zhang L, Fang Y, Li X, Guo X. Alginate oligosaccharide structures differentially affect DSS-induced colitis in mice by modulating gut microbiota. Carbohydr Polym 2023; 312:120806. [PMID: 37059538 DOI: 10.1016/j.carbpol.2023.120806] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/25/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Alginate oligosaccharides (AOS) are divided by their monomer sequences into three types: oligomannuronate (MAOS), oligoguluronate (GAOS), and heterogeneous AOS (HAOS). However, how these AOS structures differentially regulate health and modulate gut microbiota is unclear. We explored the structure-function relationship of AOS both in an in vivo colitis model and an in vitro enterotoxigenic Escherichia coli (ETEC)-challenged cell model. We found that MAOS administration significantly alleviated the symptom of experimental colitis and improved the gut barrier function in vivo and in vivo. Nevertheless, HAOS and GAOS were less effective than MAOS. The abundance and diversity of gut microbiota are obviously increased by MAOS intervention, but not by HAOS or GAOS. Importantly, microbiota from MAOS-dosed mice through FMT decreased the disease index level, alleviated histopathological changes, and improved gut barrier function in the colitis model. Super FMT donors induced by MAOS but not by HAOS or GAOS, seemed to exert potential in colitis bacteriotherapy. These findings may aid in establishing precise pharmaceutical applications based on the targeted production of AOS.
Collapse
|
17
|
Gut-Spleen Axis: Microbiota via Vascular and Immune Pathways Improve Busulfan-Induced Spleen Disruption. mSphere 2023; 8:e0058122. [PMID: 36511706 PMCID: PMC9942571 DOI: 10.1128/msphere.00581-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fecal microbiota transplantation (FMT) is an effective means of modulating gut microbiota for the treatment of many diseases, including Clostridioides difficile infections. The gut-spleen axis has been established, and this is involved in the development and function of the spleen. However, it is not understood whether gut microbiota can be used to improve spleen function, especially in spleens disrupted by a disease or an anti-cancer treatment. In the current investigation, we established that alginate oligosaccharide (AOS)-improved gut microbiota (A10-FMT) can rescue anticancer drug busulfan-disrupted spleen vasculature and spleen function. A10-FMT improved the gene and/or protein expression of genes involved in vasculature development, increased the cell proliferation rate, enhanced the endothelial progenitor cell capability, and elevated the expression of the cell junction molecules to increase the vascularization of the spleen. This investigation found for the first time that the reestablishment of spleen vascularization restored spleen function by improving spleen immune cells and iron metabolism. These findings may be used as a strategy to minimize the side effects of anti-cancer drugs or to improve spleen vasculature-related diseases. IMPORTANCE Alginate oligosaccharide (AOS)-improved gut microbiota (A10-FMT) can rescue busulfan disrupted spleen vasculature. A10-FMT improved the cell proliferation rate, endothelial progenitor cell capability, and cell junction molecules to increase vasculature formation in the spleen. This reestablishment restored spleen function by improving spleen immune cells and iron metabolism. These findings are useful for the treatment of spleen vasculature-related diseases.
Collapse
|
18
|
Zhang Z, Wang X, Li F. An exploration of alginate oligosaccharides modulating intestinal inflammatory networks via gut microbiota. Front Microbiol 2023; 14:1072151. [PMID: 36778853 PMCID: PMC9909292 DOI: 10.3389/fmicb.2023.1072151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Alginate oligosaccharides (AOS) can be obtained by acidolysis and enzymatic hydrolysis. The products obtained by different methods have different structures and physiological functions. AOS have received increasing interest because of their many health-promoting properties. AOS have been reported to exert protective roles for intestinal homeostasis by modulating gut microbiota, which is closely associated with intestinal inflammation, gut barrier strength, bacterial infection, tissue injury, and biological activities. However, the roles of AOS in intestinal inflammation network remain not well understood. A review of published reports may help us to establish the linkage that AOS may improve intestinal inflammation network by affecting T helper type 1 (Th1) Th2, Th9, Th17, Th22 and regulatory T (Treg) cells, and their secreted cytokines [the hub genes of protein-protein interaction networks include interleukin-1 beta (IL-1β), IL-2, IL-4, IL-6, IL-10 and tumor necrosis factor alpha (TNF-α)] via the regulation of probiotics. The potential functional roles of molecular mechanisms are explored in this study. However, the exact mechanism for the direct interaction between AOS and probiotics or pathogenic bacteria is not yet fully understood. AOS receptors may be located on the plasma membrane of gut microbiota and will be a key solution to address such an important issue. The present paper provides a better understanding of the protecting functions of AOS on intestinal inflammation and immunity.
Collapse
Affiliation(s)
- Zhikai Zhang
- Wuzhoufeng Agricultural Science and Technology Co., Ltd., Yantai, China
| | | | | |
Collapse
|
19
|
Bao L, Fu L, Su Y, Chen Z, Peng Z, Sun L, Gonzalez FJ, Wu C, Zhang H, Shi B, Shi YB. Amino acid transporter SLC7A5 regulates Paneth cell function to affect the intestinal inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.24.524966. [PMID: 36789439 PMCID: PMC9928054 DOI: 10.1101/2023.01.24.524966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The intestine is critical for not only processing and resorbing nutrients but also protecting the organism from the environment. These functions are mainly carried out by the epithelium, which is constantly being self-renewed. Many genes and pathways can influence intestinal epithelial cell proliferation. Among them is mTORC1, whose activation increases cell proliferation. Here, we report the first intestinal epithelial cell-specific knockout ( ΔIEC ) of an amino acid transporter capable of activating mTORC1. We show that the transporter, SLC7A5, is highly expressed in mouse intestinal crypt and Slc7a5 ΔIEC reduces mTORC1 signaling. Surprisingly, Slc7a5 ΔIEC mice have increased cell proliferation but reduced secretory cells, particularly mature Paneth cells. scRNA-seq and electron microscopic analyses revealed dedifferentiation of Paneth cells in Slc7a5 ΔIEC mice, leading to markedly reduced secretory granules with little effect on Paneth cell number. We further show that Slc7a5 ΔIEC mice are prone to experimental colitis. Thus, SLC7A5 regulates secretory cell differentiation to affect stem cell niche and/or inflammatory response to regulate cell proliferation.
Collapse
|
20
|
Diego-González L, Simón-Vázquez R. Immunomodulatory properties of algae. FUNCTIONAL INGREDIENTS FROM ALGAE FOR FOODS AND NUTRACEUTICALS 2023:593-615. [DOI: 10.1016/b978-0-323-98819-3.00011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
21
|
Mavrogeni ME, Asadpoor M, Henricks PAJ, Keshavarzian A, Folkerts G, Braber S. Direct Action of Non-Digestible Oligosaccharides against a Leaky Gut. Nutrients 2022; 14:4699. [PMID: 36364961 PMCID: PMC9655944 DOI: 10.3390/nu14214699] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 10/28/2023] Open
Abstract
The epithelial monolayer is the primary determinant of mucosal barrier function, and tight junction (TJ) complexes seal the paracellular space between the adjacent epithelial cells and represent the main "gate-keepers" of the paracellular route. Impaired TJ functionality results in increased permeation of the "pro-inflammatory" luminal contents to the circulation that induces local and systemic inflammatory and immune responses, ultimately triggering and/or perpetuating (chronic) systemic inflammatory disorders. Increased gut leakiness is associated with intestinal and systemic disease states such as inflammatory bowel disease and neurodegenerative diseases such as Parkinson's disease. Modulation of TJ dynamics is an appealing strategy aiming at inflammatory conditions associated with compromised intestinal epithelial function. Recently there has been a growing interest in nutraceuticals, particularly in non-digestible oligosaccharides (NDOs). NDOs confer innumerable health benefits via microbiome-shaping and gut microbiota-related immune responses, including enhancement of epithelial barrier integrity. Emerging evidence supports that NDOs also exert health-beneficial effects on microbiota independently via direct interactions with intestinal epithelial and immune cells. Among these valuable features, NDOs promote barrier function by directly regulating TJs via AMPK-, PKC-, MAPK-, and TLR-associated pathways. This review provides a comprehensive overview of the epithelial barrier-protective effects of different NDOs with a special focus on their microbiota-independent modulation of TJs.
Collapse
Affiliation(s)
- Maria Eleni Mavrogeni
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Mostafa Asadpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Paul A. J. Henricks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Ali Keshavarzian
- Division of Gastroenterology, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
22
|
Wang Y, Song W, Yu S, Liu Y, Chen YG. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. CELL REGENERATION 2022; 11:26. [PMID: 36045190 PMCID: PMC9433512 DOI: 10.1186/s13619-022-00127-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
The intestinal epithelium is responsible for food digestion and nutrient absorption and plays a critical role in hormone secretion, microorganism defense, and immune response. These functions depend on the integral single-layered intestinal epithelium, which shows diversified cell constitution and rapid self-renewal and presents powerful regeneration plasticity after injury. Derailment of homeostasis of the intestine epithelium leads to the development of diseases, most commonly including enteritis and colorectal cancer. Therefore, it is important to understand the cellular characterization of the intestinal epithelium at the molecular level and the mechanisms underlying its homeostatic maintenance. Single-cell technologies allow us to gain molecular insights at the single-cell level. In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and intestinal disease development.
Collapse
|
23
|
Han H, Zhou Y, Xiong B, Zhong R, Jiang Y, Sun H, Tan J, Zhang B, Guan C, Schroyen M, Chen L, Zhao Y, Zhang H. Alginate oligosaccharides increase boar semen quality by affecting gut microbiota and metabolites in blood and sperm. Front Microbiol 2022; 13:982152. [PMID: 36071975 PMCID: PMC9441641 DOI: 10.3389/fmicb.2022.982152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Alginate oligosaccharides (AOS), natural polymers from brown seaweeds (such as Laminaria japonica, Undaria pinnatifida, and Sargassum fusiforme), have been reported to possess many beneficial advantages for health. In the current study, after 9 weeks of dietary supplementation, AOS 10 mg/kg group (AOS 10) group increased boar sperm motility from 87.8% to 93.5%, p < 0.05. Moreover, AOS10 increased the relative abundances of Bifidobacterium, Coprococcus, Butyricicoccus (1.3–2.3-fold; p < 0.05) to increase the beneficial blood and sperm metabolites (1.2–1.6-fold; p < 0.05), and important sperm proteins such as gelsolin, Zn-alpha2 glycoprotein, Cation Channel Sperm-Associated Protein, outer dense fiber of sperm tails, etc. (1.5–2.2-fold; p < 0.05). AOS had a long-term beneficial advantage on boar semen quality by the increase in semen volume (175 vs. 160 ml/ejaculation, p < 0.05). AOS may be used as dietary additives for improving semen quality.
Collapse
Affiliation(s)
- Hui Han
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of AgroBioChem, Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux AgroBioTech, University of Liège, Gembloux, Belgium
| | - Yexun Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of AgroBioChem, Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux AgroBioTech, University of Liège, Gembloux, Belgium
| | - Bohui Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yue Jiang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Haiqing Sun
- YangXiang Joint Stock Company, Guigang, China
| | - Jiajian Tan
- YangXiang Joint Stock Company, Guigang, China
| | - Bin Zhang
- Qingdao BZ Oligo Biotech Co., Ltd, Qingdao, China
| | - Chang Guan
- Qingdao BZ Oligo Biotech Co., Ltd, Qingdao, China
| | - Martine Schroyen
- Department of AgroBioChem, Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux AgroBioTech, University of Liège, Gembloux, Belgium
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Liang Chen,
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Yong Zhao, ;
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
24
|
Chen L, Meng Q, Li S, Jiang Y, Zhang C, Tang S, Zhong R, Tang X, Zhang S, Feng X, Zhao Y, Zhang H. Multi-Omics Uncover Neonatal Cecal Cell Development Potentials. Front Cell Dev Biol 2022; 10:840298. [PMID: 35912104 PMCID: PMC9334561 DOI: 10.3389/fcell.2022.840298] [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: 12/21/2021] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
Although, the cecum plays vital roles in absorption of water, electrolytes, and other small molecules, and harbors trillions of commensal bacteria to shape large intestine immune functions, it is unknown the cecum development potentials at single cell level during the very crucial neonatal developmental period. Using singe cell RNA-seq and proteomics, we have characterized six major types of cecal cells: undifferentiated cells; immune cells (Ims); cecumocytes (CCs); goblet, Paneth like cells (PLCs), and enteroendocrine cells (EECs) with specific markers. CCs mature with a gradual decrease in proportion of cells; however, Ims develop with a continuing increase in proportion of cells. Meanwhile, goblet and EEC cells reduced in proportion of cells from do to d14 or d21; PLCs increased in proportion of cells from d0 to d7 then decreased at d14 and d21. The cells exhibit specific development and maturation trends controlled by transcriptional factors, ligand-receptor pairs, and other factors. As piglets grow, cecal content and mucosal microbial diversity increases dramatically with population of beneficial microbiota, such as lactobacillus. Moreover, cecal mucosal-associated and cecal content microbiota are positively correlated and both show significant correlation with different types of cecal cells and plasma metabolites. This is the first presentation of neonatal cecal cell development and maturation naturally at single cell level with transcript, protein, microbiota and metabolism perspectives. Furthermore, this study provides an important tool for the determination of novel interventions in cecal drug delivery and metabolism studies.
Collapse
Affiliation(s)
- Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingshi Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shen Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yue Jiang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Cong Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shanlong Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Cornell University, Ithaca, NY, United States
| | - Xiaohui Feng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Yong Zhao, ; Hongfu Zhang,
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- *Correspondence: Yong Zhao, ; Hongfu Zhang,
| |
Collapse
|
25
|
Alginate Oligosaccharides Ameliorate DSS-Induced Colitis through Modulation of AMPK/NF-κB Pathway and Intestinal Microbiota. Nutrients 2022; 14:nu14142864. [PMID: 35889822 PMCID: PMC9321948 DOI: 10.3390/nu14142864] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023] Open
Abstract
Alginate oligosaccharides (AOS) are shown to have various biological activities of great value to medicine, food, and agriculture. However, little information is available about their beneficial effects and mechanisms on ulcerative colitis. In this study, AOS with a polymerization degree between 2 and 4 were found to possess anti-inflammatory effects in vitro and in vivo. AOS could decrease the levels of nitric oxide (NO), IL-1β, IL-6, and TNFα, and upregulate the levels of IL-10 in both RAW 264.7 and bone-marrow-derived macrophage (BMDM) cells under lipopolysaccharide (LPS) stimulation. Additionally, oral AOS administration could significantly prevent bodyweight loss, colonic shortening, and rectal bleeding in dextran sodium sulfate (DSS)-induced colitis mice. AOS pretreatment could also reduce disease activity index scores and histopathologic scores and downregulate proinflammatory cytokine levels. Importantly, AOS administration could reverse DSS-induced AMPK deactivation and NF-κB activation in colonic tissues, as evidenced by enhanced AMPK phosphorylation and p65 phosphorylation inhibition. AOS could also upregulate AMPK phosphorylation and inhibit NF-κB activation in vitro. Moreover, 16S rRNA gene sequencing of gut microbiota indicated that supplemental doses of AOS could affect overall gut microbiota structure to a varying extent and specifically change the abundance of some bacteria. Medium-dose AOS could be superior to low- or high-dose AOS in maintaining remission in DSS-induced colitis mice. In conclusion, AOS can play a protective role in colitis through modulation of gut microbiota and the AMPK/NF-kB pathway.
Collapse
|
26
|
Lu S, Na K, Wei J, Zhang L, Guo X. Alginate oligosaccharides: The structure-function relationships and the directional preparation for application. Carbohydr Polym 2022; 284:119225. [PMID: 35287920 DOI: 10.1016/j.carbpol.2022.119225] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/20/2022] [Accepted: 02/02/2022] [Indexed: 01/02/2023]
Abstract
Alginate oligosaccharides (AOS) are degradation products of alginate extracted from brown algae. With low molecular weight, high water solubility, and good biological activity, AOS present anti-inflammatory, antimicrobial, antioxidant, and antitumor properties. They also exert growth-promoting effects in animals and plants. Three types of AOS, mannuronate oligosaccharides (MAOS), guluronate oligosaccharides (GAOS), and heterozygous mannuronate and guluronate oligosaccharides (HAOS), can be produced from alginate by enzymatic hydrolysis. Thus far, most studies on the applications and biological activities of AOS have been based mainly on a hybrid form of HAOS. To improve the directional production of AOS for practical applications, systematic studies on the structures and related biological activities of AOS are needed. This review provides a summary of current understanding of structure-function relationships and advances in the production of AOS. The current challenges and opportunities in the application of AOS is suggested to guide the precise application of AOS in practice.
Collapse
Affiliation(s)
- Shuang Lu
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Kai Na
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Jiani Wei
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Li Zhang
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China
| | - Xiaohua Guo
- College of Life Science, South-Central University for Nationalities, No. 182, Minyuan Road, Hongshan District, Wuhan City, Hubei Province 430074, China.
| |
Collapse
|
27
|
Yan X, Ma X, Hao Y, Liu J, Fang H, Lu D, Shen W, Zhang H, Ge W, Zhao Y. Alginate oligosaccharides ameliorate busulfan-induced renal tubule injury. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
28
|
Improvement of ovarian insufficiency from alginate oligosaccharide in mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
Li T, Huang S, Wang J, Yin P, Liu H, Sun C. Alginate oligosaccharides protect against fumonisin B1-induced intestinal damage via promoting gut microbiota homeostasis. Food Res Int 2022; 152:110927. [PMID: 35181098 DOI: 10.1016/j.foodres.2021.110927] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022]
Abstract
Fumonisin B1 (FB1), one of the most common mycotoxins contaminating feed and food, has been shown to induce intestinal barrier degradation. However, its role on gut microbiota in this process is still unclear. Alginate oligosaccharides (AOS) have been reported to exert their anti-inflammatory and anti-apoptotic function partially via modulation the gut microbiota. However, little is known about the beneficial effect of AOS on gut microbiota upon FB1 exposure. Results show that FB1 degraded intestinal epithelial barrier function as evidenced by increased pathological epithelial cell shedding, reduced the number of goblet cells, and promoted intestinal cell apoptosis. Markedly, FB1 disturbed the cecal and fecal microbiota composition. FB1 increased the level of Lactobacillus and decreased the relative abundance of beneficial microbes. FB1 largely inhibited the production of short chain fatty acids (SCFAs). AOS greatly ameliorated FB1-induced intestinal damage, inflammation, and oxidative stress (eg., T-SOD and MDA). AOS alleviated gut microbial dysbiosis by promoting the growth of beneficial microbes such as Roseburia, Bifidobacterium, and Akkermansia, and increasing SCFAs production upon FB1 exposure. Moreover, the correlation analysis showed that FB1- and AOS-treated gut microbiota alteration is closely associated with the change of intestinal phenotype. We have thus provided a novel insight into the protective role of AOS on FB1-induced gut microbial dysbiosis.
Collapse
Affiliation(s)
- Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Jun Wang
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Peng Yin
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Hujun Liu
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China
| | - Changpo Sun
- Academy of National Food and Strategic Reserves Administration, Beijing 100037, China; Standards and Quality Center of National Food and Strategic Reserves Administration, China.
| |
Collapse
|
30
|
Wang B, Xu J, Jiang S, Wang Y, Zhu J, Zhang Y. Combined Analysis of Gut Microbiota and Plasma Metabolites Reveals the Effect of Red-Fleshed Apple Anthocyanin Extract on Dysfunction of Mice Reproductive System Induced by Busulfan. Front Nutr 2022; 8:802352. [PMID: 35096946 PMCID: PMC8789878 DOI: 10.3389/fnut.2021.802352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/14/2021] [Indexed: 12/28/2022] Open
Abstract
Busulfan is currently an indispensable anti-cancer drug, but the side effects on male reproductive system are so serious. Meanwhile, red-fleshed apples are natural products with high anthocyanin content. In this research, we analyzed the effect of red-fleshed apple anthocyanin extract (RAAE) on busulfan-treated mice. Compared with the busulfan group, main plasma biochemical indicators were significantly improved after RAAE treatment. Compared with BA0 (busulfan without RAAE) group, total antioxidant capacity(T-AOC) and the activity of superoxide dismutase (SOD) and glutathione catalase (GSH-Px) in RAAE treatment groups were obviously increased, while the activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were significantly decreased. Malondialdehyde (MDA) was significantly decreased in the RAAE groups. In addition, we found RAAE alleviated busulfan-disrupted spermatogenesis through improving genes expression which are important for spermatogenesis, such as DDX4, PGK2, and TP1. Furthermore, we found that RAAE increased beneficial bacteria Akkermansia and Lactobacillaceae, and significantly depleted harmful bacteria Erysipelotrichia. The correlation studies indicated that RAAE ameliorated busulfan-induced rise in LysoPC levels through regulating gut microbial community and their associated metabolites. In conclusion, this study extends our understanding of the alleviated effect of RAAE on busulfan-induced male reproductive dysfunction through regulating the relationships between gut microbiota and metabolites.
Collapse
Affiliation(s)
- Bin Wang
- Engineering Laboratory of Genetic Improvement of Horticultural Crops of Shandong Province, Qingdao Agricultural University, Qingdao, China.,College of Horticulture, Qingdao Agricultural University, Qingdao, China
| | - Jihua Xu
- Engineering Laboratory of Genetic Improvement of Horticultural Crops of Shandong Province, Qingdao Agricultural University, Qingdao, China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Shenhui Jiang
- College of Horticulture, Qingdao Agricultural University, Qingdao, China
| | - Yanbo Wang
- College of Horticulture, Qingdao Agricultural University, Qingdao, China
| | - Jun Zhu
- College of Horticulture, Qingdao Agricultural University, Qingdao, China
| | - Yugang Zhang
- Engineering Laboratory of Genetic Improvement of Horticultural Crops of Shandong Province, Qingdao Agricultural University, Qingdao, China.,College of Horticulture, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
31
|
Tang L, Guo E, Zhang L, Wang Y, Gao S, Bao M, Han F, Yu W. The Function of CBM32 in Alginate Lyase VxAly7B on the Activity on Both Soluble Sodium Alginate and Alginate Gel. Front Microbiol 2022; 12:798819. [PMID: 35069502 PMCID: PMC8776709 DOI: 10.3389/fmicb.2021.798819] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/15/2021] [Indexed: 11/29/2022] Open
Abstract
Carbohydrate-binding modules (CBMs), as an important auxiliary module, play a key role in degrading soluble alginate by alginate lyase, but the function on alginate gel has not been elucidated. Recently, we reported alginate lyase VxAly7B containing a CBM32 and a polysaccharide lyase family 7 (PL7). To investigate the specific function of CBM32, we characterized the full-length alginate lyase VxAly7B (VxAly7B-FL) and truncated mutants VxAly7B-CM (PL7) and VxAly7B-CBM (CBM32). Both VxAly7B-FL and native VxAly7B can spontaneously cleavage between CBM32 and PL7. The substrate-binding capacity and activity of VxAly7B-CM to soluble alginate were 0.86- and 1.97-fold those of VxAly7B-FL, respectively. Moreover, CBM32 could accelerate the expansion and cleavage of alginate gel beads, and the degradation rate of VxAly7B-FL to alginate gel beads was threefold that of VxAly7B-CM. Results showed that CBM32 is not conducive to the degradation of soluble alginate by VxAly7B but is helpful for binding and degradation of insoluble alginate gel. This study provides new insights into the function of CBM32 on alginate gel, which may inspire the application strategy of CBMs in insoluble substrates.
Collapse
Affiliation(s)
- Luyao Tang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Enwen Guo
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Lan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Ying Wang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shan Gao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Mengmeng Bao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Feng Han
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wengong Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Key Laboratory of Marine Drugs, Ministry of Education, Qingdao, China
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
32
|
Xiong LL, Xue LL, Du RL, Niu RZ, Chen L, Chen J, Hu Q, Tan YX, Shang HF, Liu J, Yu CY, Wang TH. Single-cell RNA sequencing reveals B cell-related molecular biomarkers for Alzheimer's disease. Exp Mol Med 2021; 53:1888-1901. [PMID: 34880454 PMCID: PMC8741783 DOI: 10.1038/s12276-021-00714-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 08/08/2021] [Accepted: 08/30/2021] [Indexed: 02/05/2023] Open
Abstract
In recent years, biomarkers have been integrated into the diagnostic process and have become increasingly indispensable for obtaining knowledge of the neurodegenerative processes in Alzheimer's disease (AD). Peripheral blood mononuclear cells (PBMCs) in human blood have been reported to participate in a variety of neurodegenerative activities. Here, a single-cell RNA sequencing analysis of PBMCs from 4 AD patients (2 in the early stage, 2 in the late stage) and 2 normal controls was performed to explore the differential cell subpopulations in PBMCs of AD patients. A significant decrease in B cells was detected in the blood of AD patients. Furthermore, we further examined PBMCs from 43 AD patients and 41 normal subjects by fluorescence activated cell sorting (FACS), and combined with correlation analysis, we found that the reduction in B cells was closely correlated with the patients' Clinical Dementia Rating (CDR) scores. To confirm the role of B cells in AD progression, functional experiments were performed in early-stage AD mice in which fibrous plaques were beginning to appear; the results demonstrated that B cell depletion in the early stage of AD markedly accelerated and aggravated cognitive dysfunction and augmented the Aβ burden in AD mice. Importantly, the experiments revealed 18 genes that were specifically upregulated and 7 genes that were specifically downregulated in B cells as the disease progressed, and several of these genes exhibited close correlation with AD. These findings identified possible B cell-based AD severity, which are anticipated to be conducive to the clinical identification of AD progression.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650504, Yunnan, China
| | - Lu-Lu Xue
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ruo-Lan Du
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China
| | - Rui-Ze Niu
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China
| | - Li Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jie Chen
- School of Anesthesiology , Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qiao Hu
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ya-Xin Tan
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China
| | - Hui-Fang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jia Liu
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China.
| | - Chang-Yin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - Ting-Hua Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Laboratory Animal Department, Kunming Medical University, Kunming, 650031, Yunnan, China.
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
33
|
Single-Cell Transcriptome Sequencing and Proteomics Reveal Neonatal Ileum Dynamic Developmental Potentials. mSystems 2021; 6:e0072521. [PMID: 34546071 PMCID: PMC8547457 DOI: 10.1128/msystems.00725-21] [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] [Indexed: 01/21/2023] Open
Abstract
The neonatal period is a crucial time during development of the mammalian small intestine. Moreover, neonatal development and maturation of the small intestine are exceptionally important for early growth, successful weaning, and postweaning growth and development, in order to achieve species-specific milestones. Although several publications recently characterized intestinal epithelial cell diversity at the single-cell level, it remains unclear how differentiation and molecular interactions take place between types and subtypes of epithelial cells during the neonatal period. A single-cell RNA sequencing (scRNA-seq) survey of 40,186 ileal epithelial cells and proteomics analysis of ileal samples at 6 time points in the swine neonatal period were performed. The results revealed previously unknown developmental changes: specific increases in undifferentiated cells, unique enterocyte differentiation, and time-dependent reduction in secretory cells. Moreover, we observed specific transcriptional factors, ligand-receptor pairs, G protein-coupled receptors, transforming growth factor β, bone morphogenetic protein signaling pathways, and gut mucosal microbiota playing vital roles in ileal development during the neonatal window. This work offers new comprehensive information regarding ileal development throughout the neonatal period. Reference to this data set may assist in the creation of novel interventions for inflammation-, metabolism-, and proliferation-related gut pathologies. IMPORTANCE We found previously unknown neonatal ileum developmental potentials: specific increases in undifferentiated cells, unique enterocyte differentiation, and time dependent reduction in secretory cells. Specific transcriptional factors (TFs), ligand-receptor pairs, G protein-coupled receptors, transforming growth factor β, bone morphogenetic protein signaling pathways, and the gut mucosal microbiota are involved in this process. Our results may assist in the creation of novel interventions for inflammation-, metabolism-, and proliferation-related gut pathologies.
Collapse
|
34
|
Wang M, Chen L, Zhang Z. Potential applications of alginate oligosaccharides for biomedicine - A mini review. Carbohydr Polym 2021; 271:118408. [PMID: 34364551 DOI: 10.1016/j.carbpol.2021.118408] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/23/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023]
Abstract
Extensive research on marine algae, especially on their health-promoting properties, has been conducted. Various ingredients with potential biomedical applications have been discovered and extracted from marine algae. Alginate oligosaccharides are low molecular weight alginate polysaccharides present in cell walls of brown algae. They exhibit various health benefits such as anti-inflammatory, anti-microbial, anti-oxidant, anti-tumor and immunomodulation. Their low-toxicity, non-immunogenicity, and biodegradability make them an excellent material in biomedicine. Alginate oligosaccharides can be chemically or biochemically modified to enhance their biological activity and potential in pharmaceutical applications. This paper provides a brief overview on alginate oligosaccharides characteristics, modification patterns and highlights their vital health promoting properties.
Collapse
Affiliation(s)
- Mingpeng Wang
- College of Life Science, Qufu Normal University, Qufu 273100, China
| | - Lei Chen
- College of Life Science, Qufu Normal University, Qufu 273100, China.
| | - Zhaojie Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
35
|
Huang J, Huang J, Li Y, Wang Y, Wang F, Qiu X, Liu X, Li H. Sodium Alginate Modulates Immunity, Intestinal Mucosal Barrier Function, and Gut Microbiota in Cyclophosphamide-Induced Immunosuppressed BALB/c Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7064-7073. [PMID: 34152142 DOI: 10.1021/acs.jafc.1c02294] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
This study investigated the protective effects of sodium alginate (SA) on the gut microbiota, immunity, and intestinal mucosal barrier function in cyclophosphamide-induced immunosuppressed BALB/c mice. SA alleviated spleen tissue damage and restored impaired immune functions, such as increasing the immune organ index, decreasing splenic T lymphocytes, and markedly increasing the secretion of serum immunoglobulins and cytokines in immunosuppressed mice. In addition, SA reversed the intestinal mucosal injury and increased the intestinal permeability by upregulating the expression of tight junction proteins. Moreover, SA decreased gut inflammation by reducing serum d-lactic acid (D-LA) and lipopolysaccharide (LPS) concentrations and downregulating toll-like receptor 4 (Tlr4) and mitogen-activated protein kinase (Mapk) pathway expression. Furthermore, SA significantly increased the abundance of beneficial bacteria (Lactobacillus, Roseburia, and Lachnospiraceae NK4A136) and decreased pathogenic bacteria (Helicobacter, Peptococcus, and Tyzzerella) in the intestine as determined by 16S rRNA gene high-throughput sequencing. In conclusion, our study provides a scientific basis for SA as a functional food in modulating gut microbiota and protecting against intestinal mucosal injury and indicates that SA has potential application for enhancing immunity.
Collapse
Affiliation(s)
- Juan Huang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| | - Jinli Huang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| | - Yao Li
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| | - Yilu Wang
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| | - Fahe Wang
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao Bright Moon Seaweed Group Co.,, Ltd., 266400 Qingdao, PR China
| | - Xia Qiu
- State Key Laboratory of Bioactive Seaweed Substances, Qingdao Bright Moon Seaweed Group Co.,, Ltd., 266400 Qingdao, PR China
| | - Xvliang Liu
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| | - Huajun Li
- Department of Microecology, College of Basic Medical Sciences, Dalian Medical University, 116044 Dalian, PR China
| |
Collapse
|
36
|
Han X, Zhang C, Ma X, Yan X, Xiong B, Shen W, Yin S, Zhang H, Sun Q, Zhao Y. Muscarinic acetylcholine receptor M5 is involved in spermatogenesis through the modification of cell-cell junctions. Reproduction 2021; 162:47-59. [PMID: 33970124 PMCID: PMC8183636 DOI: 10.1530/rep-21-0079] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023]
Abstract
Muscarinic acetylcholine receptor (mAChR) antagonists have been reported to decrease male fertility; however, the roles of mAChRs in spermatogenesis and the underlying mechanisms are not understood yet. During spermatogenesis, extensive remodeling between Sertoli cells and/or germ cells interfaces takes place to accommodate the transport of developing germ cells across the blood-testis barrier (BTB) and adluminal compartment. The cell–cell junctions play a vital role in the spermatogenesis process. This study used ICR male mice and spermatogonial cells (C18-4) and Sertoli cells (TM-4). shRNA of control or M5 gene was injected into 5-week-old ICR mice testes. Ten days post-viral grafting, mice were deeply anesthetized with pentobarbital and the testes were collected. One testicle was fresh frozen for RNA-seq analysis or Western blotting (WB). The second testicle was fixed for immunofluorescence staining (IHF). C18-4 or TM-4 cells were treated with shRNA of control or M5 gene. Then, the cells were collected for RNA-seq analysis, WB, or IHF. Knockdown of mAChR M5 disrupted mouse spermatogenesis and damaged the actin-based cytoskeleton and many types of junction proteins in both Sertoli cells and germ cells. M5 knockdown decreased Phldb2 expression in both germ cells and Sertoli cells which suggested that Phldb2 may be involved in cytoskeleton and cell–cell junction formation to regulate spermatogenesis. Our investigation has elucidated a novel role for mAChR M5 in the regulation of spermatogenesis through the interactions of Phldb2 and cell–cell junctions. M5 may be an attractive future therapeutic target in the treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Xiao Han
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Cong Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Xiangping Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Xiaowei Yan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Bohui Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Shen Yin
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China
| | - Qingyuan Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China.,Fertility Preservation Lab, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, People's Republic of China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, People's Republic of China.,College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| |
Collapse
|
37
|
Bai XF, Niu RZ, Liu J, Pan XD, Wang F, Yang W, Wang LQ, Sun LZ. Roles of noncoding RNAs in the initiation and progression of myocardial ischemia-reperfusion injury. Epigenomics 2021; 13:715-743. [PMID: 33858189 DOI: 10.2217/epi-2020-0359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The morbidity and mortality of myocardial ischemia-reperfusion injury (MIRI) have increased in modern society. Noncoding RNAs (ncRNAs), including lncRNAs, circRNAs, piRNAs and miRNAs, have been reported in a variety of studies to be involved in pathological initiation and developments of MIRI. Hence this review focuses on the current research regarding these ncRNAs in MIRI. We comprehensively introduce the important features of lncRNAs, circRNAs, piRNA and miRNAs and then summarize the published studies of ncRNAs in MIRI. A clarification of lncRNA-miRNA-mRNA, lncRNA-transcription factor-mRNA and circRNA-miRNA-mRNA axes in MIRI follows, to further elucidate the crucial roles of ncRNAs in MIRI. Bioinformatics analysis has revealed the biological correlation of mRNAs with MIRI. We provide a comprehensive perspective for the roles of these ncRNAs and their related networks in MIRI, providing a theoretical basis for preclinical and clinical studies on ncRNA-based gene therapy for MIRI treatment.
Collapse
Affiliation(s)
- Xiang-Feng Bai
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China.,Department of Cardiovascular Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Rui-Ze Niu
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Jia Liu
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xu-Dong Pan
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Feng Wang
- Department of Animal Zoology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Wei Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Lu-Qiao Wang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Li-Zhong Sun
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| |
Collapse
|
38
|
Single-cell sequencing technology in tumor research. Clin Chim Acta 2021; 518:101-109. [PMID: 33766554 DOI: 10.1016/j.cca.2021.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/24/2022]
Abstract
Tumor heterogeneity is a key characteristic of malignant tumors and a significant obstacle in cancer treatment and research. Although bulk tissue sequencing has wide coverage and high accuracy, it can only represent the dominant cell signal information of each sample, while masking the unique gene expression of rare cells; therefore it cannot represent genes that are unstable within a subgroup, but unchanged in a majority of cells. With the progress of genomic technology, the emergence of single-cell sequencing (SCS) has effectively solved the above problem. Genetic, transcriptomic and epigenetic sequencing at the single-cell level provides an important basis for us to correctly classify the cell subsets of heterogeneous tumor populations and to reveal the process of complex changes in tumor cells at the molecular level. Single-cell sequencing technology has been applied to the field of cancer, revealing exciting discoveries in the potential mechanisms of tumor driver gene mutation, clonal evolution, invasion and metastasis. It also provides favorable conditions for developing new tumor biomarkers and providing more accurate and individualized targeted tumor therapy. Herein, we review the steps and methods of single-cell sequencing and highlight the application of SCS in tumor diagnosis and clinical treatment.
Collapse
|
39
|
Wallentin L, Lindbäck J, Eriksson N, Hijazi Z, Eikelboom JW, Ezekowitz MD, Granger CB, Lopes RD, Yusuf S, Oldgren J, Siegbahn A. Angiotensin-converting enzyme 2 (ACE2) levels in relation to risk factors for COVID-19 in two large cohorts of patients with atrial fibrillation. Eur Heart J 2020; 41:4037-4046. [PMID: 32984892 PMCID: PMC7543499 DOI: 10.1093/eurheartj/ehaa697] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/25/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS The global COVID-19 pandemic is caused by the SARS-CoV-2 virus entering human cells using angiotensin-converting enzyme 2 (ACE2) as a cell surface receptor. ACE2 is shed to the circulation, and a higher plasma level of soluble ACE2 (sACE2) might reflect a higher cellular expression of ACE2. The present study explored the associations between sACE2 and clinical factors, cardiovascular biomarkers, and genetic variability. METHODS AND RESULTS Plasma and DNA samples were obtained from two international cohorts of elderly patients with atrial fibrillation (n = 3999 and n = 1088). The sACE2 protein level was measured by the Olink Proteomics® Multiplex CVD II96 × 96 panel. Levels of the biomarkers high-sensitive cardiac troponin T (hs-cTnT), N-terminal probrain natriuretic peptide (NT-proBNP), growth differentiation factor 15 (GDF-15), C-reactive protein, interleukin-6, D-dimer, and cystatin-C were determined by immunoassays. Genome-wide association studies were performed by Illumina chips. Higher levels of sACE2 were statistically significantly associated with male sex, cardiovascular disease, diabetes, and older age. The sACE2 level was most strongly associated with the levels of GDF-15, NT-proBNP, and hs-cTnT. When adjusting for these biomarkers, only male sex remained associated with sACE2. We found no statistically significant genetic regulation of the sACE2 level. CONCLUSIONS Male sex and clinical or biomarker indicators of biological ageing, cardiovascular disease, and diabetes are associated with higher sACE2 levels. The levels of GDF-15 and NT-proBNP, which are associated both with the sACE2 level and a higher risk for mortality and cardiovascular disease, might contribute to better identification of risk for severe COVID-19 infection.
Collapse
Affiliation(s)
- Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Johan Lindbäck
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Ziad Hijazi
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - John W Eikelboom
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | | | - Christopher B Granger
- Duke Clinical Research Institute, Duke University Medical Center, Duke Health, Durham, NC, USA
| | - Renato D Lopes
- Duke Clinical Research Institute, Duke University Medical Center, Duke Health, Durham, NC, USA
| | - Salim Yusuf
- Population Health Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, Canada
| | - Jonas Oldgren
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Ma A, Wang C, Chang Y, Brennan FH, McDermaid A, Liu B, Zhang C, Popovich PG, Ma Q. IRIS3: integrated cell-type-specific regulon inference server from single-cell RNA-Seq. Nucleic Acids Res 2020; 48:W275-W286. [PMID: 32421805 PMCID: PMC7319566 DOI: 10.1093/nar/gkaa394] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/21/2022] Open
Abstract
A group of genes controlled as a unit, usually by the same repressor or activator gene, is known as a regulon. The ability to identify active regulons within a specific cell type, i.e., cell-type-specific regulons (CTSR), provides an extraordinary opportunity to pinpoint crucial regulators and target genes responsible for complex diseases. However, the identification of CTSRs from single-cell RNA-Seq (scRNA-Seq) data is computationally challenging. We introduce IRIS3, the first-of-its-kind web server for CTSR inference from scRNA-Seq data for human and mouse. IRIS3 is an easy-to-use server empowered by over 20 functionalities to support comprehensive interpretations and graphical visualizations of identified CTSRs. CTSR data can be used to reliably characterize and distinguish the corresponding cell type from others and can be combined with other computational or experimental analyses for biomedical studies. CTSRs can, therefore, aid in the discovery of major regulatory mechanisms and allow reliable constructions of global transcriptional regulation networks encoded in a specific cell type. The broader impact of IRIS3 includes, but is not limited to, investigation of complex diseases hierarchies and heterogeneity, causal gene regulatory network construction, and drug development. IRIS3 is freely accessible from https://bmbl.bmi.osumc.edu/iris3/ with no login requirement.
Collapse
Affiliation(s)
- Anjun Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Yuzhou Chang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Faith H Brennan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Adam McDermaid
- Imagenetics, Sanford Health, Sioux Falls, SD 57104, USA.,Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| | - Bingqiang Liu
- School of Mathematics, Shandong University, Jinan 250100, China
| | - Chi Zhang
- Department of Medical & Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN 46202, USA
| | - Phillip G Popovich
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
41
|
Xiong B, Liu M, Zhang C, Hao Y, Zhang P, Chen L, Tang X, Zhang H, Zhao Y. Alginate oligosaccharides enhance small intestine cell integrity and migration ability. Life Sci 2020; 258:118085. [DOI: 10.1016/j.lfs.2020.118085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 01/27/2023]
|
42
|
Zhang P, Liu J, Xiong B, Zhang C, Kang B, Gao Y, Li Z, Ge W, Cheng S, Hao Y, Shen W, Yu S, Chen L, Tang X, Zhao Y, Zhang H. Microbiota from alginate oligosaccharide-dosed mice successfully mitigated small intestinal mucositis. MICROBIOME 2020; 8:112. [PMID: 32711581 PMCID: PMC7382812 DOI: 10.1186/s40168-020-00886-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/30/2020] [Indexed: 05/11/2023]
Abstract
BACKGROUND The increasing incidence of cancer and intestinal mucositis induced by chemotherapeutics are causing worldwide concern. Many approaches such as fecal microbiota transplantation (FMT) have been used to minimize mucositis. However, it is still unknown whether FMT from a donor with beneficial gut microbiota results in more effective intestinal function in the recipient. Recently, we found that alginate oligosaccharides (AOS) benefit murine gut microbiota through increasing "beneficial" microbes to rescue busulfan induced mucositis. RESULTS In the current investigation, FMT from AOS-dosed mice improved small intestine function over FMT from control mice through the recovery of gene expression and an increase in the levels of cell junction proteins. FMT from AOS-dosed mice showed superior benefits over FMT from control mice on recipient gut microbiotas through an increase in "beneficial" microbes such as Leuconostocaceae and recovery in blood metabolome. Furthermore, the correlation of gut microbiota and blood metabolites suggested that the "beneficial" microbe Lactobacillales helped with the recovery of blood metabolites, while the "harmful" microbe Mycoplasmatales did not. CONCLUSION The data confirm our hypothesis that FMT from a donor with superior microbes leads to a more profound recovery of small intestinal function. We propose that gut microbiota from naturally produced AOS-treated donor may be used to prevent small intestinal mucositis induced by chemotherapeutics or other factors in recipients. Video Abstract.
Collapse
Affiliation(s)
- Pengfei Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Jing Liu
- University Research Core, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Bohui Xiong
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Cong Zhang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Beining Kang
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yishan Gao
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Zengkuan Li
- College of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Wei Ge
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Shunfeng Cheng
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Yanan Hao
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Shuai Yu
- Center for Reproductive Medicine, Urology Department, Peking University Shenzhen Hospital, Shenzhen, 518036, People's Republic of China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Yong Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
| |
Collapse
|
43
|
The Lactobacillus brevis 47 f Strain Protects the Murine Intestine from Enteropathy Induced by 5-Fluorouracil. Microorganisms 2020; 8:microorganisms8060876. [PMID: 32527057 PMCID: PMC7356614 DOI: 10.3390/microorganisms8060876] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/06/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023] Open
Abstract
We report that the results of our study indicate that Lactobacillus brevis 47 f strain isolated from the faeces of a healthy individual prevents the manifestations of experimental mucositis induced by treatment of Balb/c mice with the anticancer drug 5-fluorouracil (5 FU; 100 mg/kg i.p. × 3 days). The presence of damage to the intestine and the colon was determined by a morphometric analysis of specimens including the height of villi, the amount of goblet cells and infiltrating mononuclear cells, and the expression of the proliferative Ki-67 antigen. Changes in the lipid peroxidation in the blood and the intestine were determined by severalfold increase of the concentration of malonic dialdehyde. Oral administration of L. brevis 47 f strain prior to 5 FU decreased the drug-induced morphological and biochemical changes to their respective physiological levels; the ability of intestinal epitheliocytes to express Ki-67 was partially restored. These effects of L. brevis 47 f strain were more pronounced or similar to those of the reference compound Rebamipid, a quinoline derivative known to protect the gut from drug-induced toxicity. Thus, the new lactobacilli strain attenuates the severity of 5 FU-induced enteropathy.
Collapse
|