1
|
Kim YH, Park CH, Kim JM, Yoon YC. Chitooligosaccharides suppress airway inflammation, fibrosis, and mucus hypersecretion in a house dust mite-induced allergy model. FRONTIERS IN ALLERGY 2025; 6:1533928. [PMID: 39927112 PMCID: PMC11799285 DOI: 10.3389/falgy.2025.1533928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Background Respiratory allergy is a serious respiratory disorder characterized by inflammation, mucus hypersecretion, and airway tissue sclerosis. Disruption of the T helper 1 (Th1) and T helper 2 (Th2) immune systems by stimuli induced by house dust mites (HDM) and fine particulate matter leads to the secretion of various inflammatory cytokines, resulting in immune respiratory diseases characterized by airway inflammation. Chitooligosaccharides (COS) are known for their antioxidant and anti-inflammatory properties. Methods Human airway epithelial cells (BEAS-2B) were cultured in DMEM/F12 medium containing COS at concentrations of 25-100 µg/ml for 24 h. No intracellular toxicity was observed up to 1,000 µg/ml. Cell experiments were conducted at COS concentrations below 100 µg/ml, while animal experiments were performed at concentrations below 100 mg/kg body weight for 4 weeks. Samples of right lung tissue obtained from the experimental animals were used for gene and protein expression analysis, whereas samples of contralateral lung tissue were used for immunohistochemical analysis. Results COS regulated Th1 immunity by inhibiting major cytokines, including inflammatory tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), in BEAS-2B cells. In the HDM-induced allergic respiratory model, COS suppressed the infiltration of inflammatory cells around the airways and inhibited the mRNA expression of Th1 immune cytokines in lung tissues, while also reducing the expression of nuclear factor kappa B (NF-κB)-related proteins. Furthermore, the results confirmed the suppression of the levels of immunoglobulin E (IgE) in the blood secreted by mast cells activated by HDM, which led to a reduction in allergic mucus hypersecretion and airway sclerosis. Conclusion In summary, COS are thought to improve airway resistance by alleviating inflammatory allergic respiratory diseases caused by HDM and are regarded as substances that regulate the balance of the Th1 and Th2 immune systems in epithelial cells affected by mucus hypersecretion.
Collapse
Affiliation(s)
| | | | | | - Yeo Cho Yoon
- Healthcare & Nutrition Laboratory, Amicogen, Inc., Seongnam, Republic of Korea
| |
Collapse
|
2
|
Rajabi M, Cabral JD, Saunderson S, Ali MA. 3D printing of chitooligosaccharide-polyethylene glycol diacrylate hydrogel inks for bone tissue regeneration. J Biomed Mater Res A 2023; 111:1468-1481. [PMID: 37066870 DOI: 10.1002/jbm.a.37548] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/09/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
To date, lack of functional hydrogel inks has limited 3D printing applications in tissue engineering. This study developed a series of photocurable hydrogel inks based on chitooligosaccharide (COS)-polyethylene glycol diacrylate (PEGDA) for extrusion-based 3D printing of bone tissue scaffolds. The scaffolds were prepared by aza-Michael addition of COS and PEGDA followed by photopolymerisation of unreacted PEGDA. The hydrogel inks showed sufficient shear thinning properties required for extrusion 3D printing. The printed scaffolds exhibited excellent shape fidelity and fine microstructure with a resolution of 250 μm. By increasing the COS content, the swelling ratio of the scaffolds decreased, while the compressive strength increased. 3D printed COS-PEGDA scaffolds showed high viability of human bone mesenchymal stem cells in vitro. In addition, scaffolds containing 2 wt% COS showed significantly higher alkaline phosphatase activity, calcium deposition, and bioactivity in simulated body fluid compared to the control (PEGDA). Altogether, 3D printed COS-PEGDA scaffolds represent promising candidates for bone tissue regeneration.
Collapse
Affiliation(s)
- Mina Rajabi
- Faculty of Dentistry, Division of Health Sciences, Centre for Bioengineering & Nanomedicine, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - Jaydee D Cabral
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Sarah Saunderson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - M Azam Ali
- Faculty of Dentistry, Division of Health Sciences, Centre for Bioengineering & Nanomedicine, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| |
Collapse
|
3
|
Kuo IP, Liu CS, Yang SD, Liang SH, Hu YF, Nan FH. Effects of Replacing Fishmeal with Defatted Black Soldier Fly ( Hermetia illucens Linnaeus) Larvae Meal in Japanese Eel ( Anguilla japonica) Diet on Growth Performance, Fillet Texture, Serum Biochemical Parameters, and Intestinal Histomorphology. AQUACULTURE NUTRITION 2022; 2022:1866142. [PMID: 36860438 PMCID: PMC9973222 DOI: 10.1155/2022/1866142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 06/18/2023]
Abstract
An 8-week feeding trial was conducted to investigate the effects of replacing fishmeal with defatted black soldier fly larvae meal (DBSFLM) in the diets of Japanese eel on their growth performance, fillet texture, serum biochemical parameters, and intestinal histomorphology. Six isoproteic (520 g kg-1), isolipidic (80 g kg-1), and isoenergetic (15 MJ kg-1) diets were formulated with fishmeal replacement levels of 0% (R0), 15% (R15), 30% (R30), 45% (R45), 60% (R60), and 75% (R75). The growth performance, feed utilization efficiency, survival rate, serum liver function enzymes, antioxidant ability, and lysozyme activity of fish were not affected (P > 0.05) by DBSFLM. However, the crude protein and cohesiveness of the fillet in groups R60 and R75 significantly decreased, and the fillet hardness significantly increased (P < 0.05). Additionally, the intestinal villus length significantly decreased in the R75 group, and the goblet cell densities were significantly lower in the R45, R60, and R75 groups (P < 0.05). Overall, high levels of DBSFLM did not affect growth performance and serum biochemical parameters but significantly altered fillet proximate composition and texture and intestinal histomorphology (P < 0.05). The optimal fishmeal replacement level is 30% with 184 g kg-1 DBSFLM.
Collapse
Affiliation(s)
- I-Pei Kuo
- Freshwater Aquaculture Research Center Chupei Station, Fisheries Research Institute, Council of Agriculture, No. 111, Tai-Ho, Zhubei, Hsinchu 30267, Taiwan
- Department of Aquaculture, National Taiwan Ocean University, No. 2, Beining Road, Keelung 202301, Taiwan
| | - Ching-Shuo Liu
- Freshwater Aquaculture Research Center Chupei Station, Fisheries Research Institute, Council of Agriculture, No. 111, Tai-Ho, Zhubei, Hsinchu 30267, Taiwan
| | - Shuenn-Der Yang
- Freshwater Aquaculture Research Center Chupei Station, Fisheries Research Institute, Council of Agriculture, No. 111, Tai-Ho, Zhubei, Hsinchu 30267, Taiwan
| | - Shih-Hsiang Liang
- Animal Industry Division, Livestock Research Institute, Council of Agriculture, No. 112, Farm Road, Hsinhua, Tainan, 71246, Taiwan
| | - Yeh-Fang Hu
- Department of Aquaculture, National Taiwan Ocean University, No. 2, Beining Road, Keelung 202301, Taiwan
| | - Fan-Hua Nan
- Department of Aquaculture, National Taiwan Ocean University, No. 2, Beining Road, Keelung 202301, Taiwan
| |
Collapse
|
4
|
Recent Research and Application Prospect of Functional Oligosaccharides on Intestinal Disease Treatment. Molecules 2022; 27:molecules27217622. [PMID: 36364447 PMCID: PMC9656564 DOI: 10.3390/molecules27217622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
The intestinal tract is an essential digestive organ of the human body, and damage to the intestinal barrier will lead to various diseases. Functional oligosaccharides are carbohydrates with a low degree of polymerization and exhibit beneficial effects on human intestinal health. Laboratory experiments and clinical studies indicate that functional oligosaccharides repair the damaged intestinal tract and maintain intestinal homeostasis by regulating intestinal barrier function, immune response, and intestinal microbial composition. Functional oligosaccharides treat intestinal disease such as inflammatory bowel disease (IBD) and colorectal cancer (CRC) and have excellent prospects for therapeutic application. Here, we present an overview of the recent research into the effects of functional oligosaccharides on intestinal health.
Collapse
|
5
|
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
|
6
|
Rajabi M, Cabral J, Saunderson S, Ali MA. Green synthesis of chitooligosaccharide-PEGDA derivatives through aza-Michael reaction for biomedical applications. Carbohydr Polym 2022; 295:119884. [DOI: 10.1016/j.carbpol.2022.119884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
|
7
|
Zhen H, Yan Q, Liu Y, Li Y, Yang S, Jiang Z. Chitin oligosaccharides alleviate atherosclerosis progress in ApoE-/- mice by regulating lipid metabolism and inhibiting inflammation. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.03.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
8
|
Jing B, Xia K, Zhang C, Jiao S, Zhu L, Wei J, Wang ZA, Chen N, Tu P, Li J, Du Y. Chitosan Oligosaccharides Regulate the Occurrence and Development of Enteritis in a Human Gut-On-a-Chip. Front Cell Dev Biol 2022; 10:877892. [PMID: 35557948 PMCID: PMC9086312 DOI: 10.3389/fcell.2022.877892] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/30/2022] [Indexed: 12/22/2022] Open
Abstract
Past studies on the protective effects of chitosan oligosaccharides (COS) on inflammatory bowel disease (IBD) commonly rely on animal models, because traditional cell culture systems couldn't faithfully mimic human intestinal physiology. Here a novel human gut-on-a-chip microsystem was established to further explore the regulatory effects of COS on the occurrence and development of human enteritis. By constructing an intestinal injury model caused by dextran sodium sulfate (DSS) on the chip, this study proved that COS can reduce intestinal epithelial injury by promoting the expression of the mucous layer for the first time. By establishing an inflammatory bowel disease model on the chip caused by E. coli 11775, this study demonstrated that COS can protect the intestinal epithelial barrier and vascular endothelial barrier by inhibiting the adhesion and invasion of E. coli 11775 for the first time. In addition, similar to the results in vivo, COS can decrease the inflammatory response by reducing the expression of toll-like receptor 4 protein and reducing the nuclear DNA binding rate of nuclear factor kappa-B protein on this chip. In summary, COS can be used as a potential drug to treat human IBD and the human gut-on-a-chip would be used as a platform for quick screening drugs to treat human IBD in future.
Collapse
Affiliation(s)
- Bolin Jing
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Kun Xia
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Chen Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Siming Jiao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Limeng Zhu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Jinhua Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Zhuo A. Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Nannan Chen
- China Food Industry Promotion Center, Beijing, China
| | - Pengfei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Yuguang Du
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Gu YF, Chen YP, Jin R, Wang C, Wen C, Zhou YM. Dietary chitooligosaccharide supplementation alleviates intestinal barrier damage, and oxidative and immunological stress in lipopolysaccharide-challenged laying hens. Poult Sci 2022; 101:101701. [PMID: 35150943 PMCID: PMC8844238 DOI: 10.1016/j.psj.2022.101701] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
This study aimed to investigate the effects of chitooligosaccharide (COS) on intestinal barrier, antioxidant capacity, and immunity of lipopolysaccharide (LPS)-challenged laying hens. A total of 360 Hy-line Brown laying hens (80-wk-old) were randomly divided into 5 groups with 6 replicates of 12 birds. Hens were fed a corn-soybean meal basal diet supplemented with different COS levels (0; 5; 10; 15; 20 mg/kg) for 8 wk. The results showed that 15 mg/kg COS administration elevated albumen height and Haugh unit (P < 0.05), and numerically optimized productive performance (P > 0.05), therefore, the dosage of 15 mg/kg was chosen for the subsequent experiment. Thereafter, 12 birds from non-supplemented group were randomly selected and assigned into 2 groups, and birds in each group were administered (1.5 mg/kg BW, i.p.) with saline (control group) or LPS (challenge group). Another 6 hens from 15 mg/kg COS-supplemented group were selected and injected with LPS in the same way. Compared with the control group, LPS-challenged birds exhibited elevated circulating diamine oxidase activity, and reduced jejunal villus height and ratio of villus height to crypt depth, and these indices were reversed to control levels by COS (P < 0.05). Also, LPS increased malondialdehyde accumulation and reduced several antioxidant enzyme activities in the intestinal mucosa (P < 0.05). Additionally, LPS increased jejunal secretory IgA and interferon-γ (IFN-γ), and ileal secretory IgA, IgM, and interleukin-1β (IL-1β) concentrations, whereas COS reduced jejunal IFN-γ and IL-1β, and ileal IgM levels (P < 0.05). Moreover, LPS down-regulated mRNA abundance of jejunal occludin and claudin 2, and upregulated expression of jejunal nuclear factor erythroid-2 related factor 2, superoxide dismutase 1, and IFN-γ as well as ileal IL-1β (P < 0.05). Besides, COS increased jejunal occludin and ileal claudin 2, nuclear factor erythroid-2 related factor 2, and heme oxygenase-1 expression, and decreased jejunal IFN-γ and IL-1β abundance (P < 0.05). These results suggested that COS could alleviate LPS-induced intestinal barrier impairment, and oxidative and immunological stress in laying hens.
Collapse
Affiliation(s)
- Y F Gu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Y P Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - R Jin
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - C Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - C Wen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Y M Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.
| |
Collapse
|
10
|
Asadpoor M, Varasteh S, Pieters RJ, Folkerts G, Braber S. Differential effects of oligosaccharides on the effectiveness of ampicillin against Escherichia coli in vitro. PHARMANUTRITION 2021. [DOI: 10.1016/j.phanu.2021.100264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
11
|
Zhu C, Zhao M, Fan L, Cao X, Xia Q, Zhou J, Yin H, Zhao L. Chitopentaose inhibits hepatocellular carcinoma by inducing mitochondrial mediated apoptosis and suppressing protective autophagy. BIORESOUR BIOPROCESS 2021; 8:4. [PMID: 38650195 PMCID: PMC10992246 DOI: 10.1186/s40643-020-00358-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 12/23/2020] [Indexed: 01/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and deadliest cancers. In this study, the anti-tumor effect of singular degree of polymerization (DP) chitooligosaccharides (COS) (DP 2-5) and the underlay molecular mechanisms were investigated on HCC cell line HepG2. MTT assay showed that (GlcN)5 have the best anti-proliferation effect among the different DP of COS (DP2-5). Furthermore, the administration of (GlcN)5 could decrease mitochondrial membrane potential, release cytochrome c into cytoplasm, activate the cleavage of Caspases9/3, thus inducing mitochondrial-mediated apoptosis in HepG2 cells (accounting for 24.57 ± 2.25%). In addition, (GlcN)5 treatment could increase the accumulation of autophagosomes. Further investigation showed that (GlcN)5 suppressed protective autophagy at the fusion of autophagosomes and lysosomes. Moreover, the inhibition of protective autophagy flux by (GlcN)5 could further decrease cell viability and increase the apoptosis rate. Our findings suggested that (GlcN)5 suppressed HepG2 proliferation through inducing apoptosis via the intrinsic pathway and impairing cell-protective autophagy. COS might have the potential to be an agent for lowering the risk of HCC.
Collapse
Affiliation(s)
- Chunfeng Zhu
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China
| | - Mengyao Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China.
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, 200237, China.
| | - Liqiang Fan
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, 200237, China
| | - Xuni Cao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, 200237, China
| | - Quanming Xia
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China
| | - Jiachun Zhou
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, 200237, China
| | - Hao Yin
- Organ Transplant Center, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Liming Zhao
- School of Biotechnology, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, No. 130 Meilong Road, Shanghai, 200237, China.
- School of Life Sciences, Shandong University of Technology, Zibo, 255049, China.
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology (SCICBT), Shanghai, 200237, China.
| |
Collapse
|
12
|
Yang Z, Tang Z, Cao X, Xie Q, Hu C, Zhong Z, Tan J, Zheng Y. Controlling chronic low-grade inflammation to improve follicle development and survival. Am J Reprod Immunol 2020; 84:e13265. [PMID: 32395847 DOI: 10.1111/aji.13265] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic low-grade inflammation is one cause of follicle development disturbance. Chronic inflammation exists in pathological conditions such as premature ovarian failure, physiological aging of the ovaries, and polycystic ovary syndrome. Inflammation of the whole body can affect oocytes via the follicle microenvironment, oxidative stress, and GM-CSF. Many substances without toxic side-effects extracted from natural organisms have gradually gained researchers' attention. Recently, chitosan oligosaccharide, resveratrol, anthocyanin, and melatonin have been found to contribute to an improvement in inflammation. This review discusses the interrelationships between chronic low-grade inflammation and follicle development, the underlying mechanisms, and methods that may improve follicle development by controlling the level of chronic low-grade inflammation.
Collapse
Affiliation(s)
- Ziwei Yang
- Jiangxi Medical College, Nanchang University, Nanchang, China.,The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province, Nanchang, China
| | - Zijuan Tang
- Jiangxi Medical College, Nanchang University, Nanchang, China.,The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province, Nanchang, China
| | - Xiuping Cao
- Jiangxi Medical College, Nanchang University, Nanchang, China.,The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province, Nanchang, China
| | - Qi Xie
- Jiangxi Medical College, Nanchang University, Nanchang, China.,The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province, Nanchang, China
| | - Chuan Hu
- Jiangxi Medical College, Nanchang University, Nanchang, China.,The Key Laboratory of Reproductive Physiology and Pathology of Jiangxi Province, Nanchang, China
| | - Zhisheng Zhong
- Reproductive Health Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Jun Tan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Yuehui Zheng
- Reproductive Health Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
13
|
Vigors S, O' Doherty JV, Ryan M, Sweeney T. Analysis of the basal colonic innate immune response of pigs divergent in feed efficiency and following an ex vivo lipopolysaccharide challenge. Physiol Genomics 2019; 51:443-448. [PMID: 31322475 DOI: 10.1152/physiolgenomics.00013.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
While feed efficiency is influenced by multiple physiological processes, it is not known how efficient and inefficient pigs differ in relation to their basal immune response, and particularly their innate immune response to a microbial challenge. Hence, the objective was to examine the expression of genes encoding innate immune response markers in basal colonic tissue and colonic tissue following an ex vivo lipopolysaccharide (LPS) challenge from pigs divergent in residual feed intake (RFI). Pigs that differed in RFI were selected from two different farms of origin. Colonic tissue was harvested from high RFI (HRFI) and low (LRFI) pigs, and two experimental conditions were explored: the first was basal unchallenged tissue and the second was colonic tissue following an ex vivo LPS challenge. RNA was extracted and tested on a Nanostring panel of 72 genes coding for barrier defense proteins, transmembrane receptors, kinases, transcription regulators, cytokines, and cytokine regulators. In the basal unchallenged tissue, the LRFI pigs had increased expression of AOAH, AP1, and TRAM and the cytokines TNF, IL10, and CXCL8, compared with the HRFI pigs (P < 0.05), with a significant effect of farm of origin on 31 genes (P < 0.05). In the LPS-challenged tissues, the LRFI group had higher expression of TLR1, TLR7, TLR8, GPR43/FFAR2, JAK2, and NFAM1 compared with the HRFI group (P < 0.05). In conclusion, these data suggest that LRFI pigs have an upregulated basal colonic inflammatory state and a heightened response to an LPS challenge compared with the inefficient HRFI pigs. This immune profile potentially enhances their capacity to respond to an infectious challenge.
Collapse
Affiliation(s)
- Stafford Vigors
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin, Ireland and
| | - John V O' Doherty
- School of Agriculture & Food Science, University College Dublin, Belfield, Dublin, Ireland and
| | - Marion Ryan
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
14
|
Hao S, Roesch EA, Perez A, Weiner RL, Henderson LC, Cummings L, Consiglio P, Pajka J, Eisenberg A, Yeh L, Cotton CU, Drumm ML. Inactivation of CFTR by CRISPR/Cas9 alters transcriptional regulation of inflammatory pathways and other networks. J Cyst Fibros 2019; 19:34-39. [PMID: 31126900 DOI: 10.1016/j.jcf.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/24/2019] [Accepted: 05/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Individuals with cystic fibrosis (CF) experience elevated inflammation in multiple organs, but whether this reflects an inherent feature of CF cells or is a consequence of a pro-inflammatory environment is not clear. METHOD Using CRISPR/Cas9-mediated mutagenesis of CFTR, 17 subclonal cell lines were generated from Caco-2 cells. Clonal lines with functional CFTR (CFTR+) were compared to those without (CFTR-) to directly address the role of CFTR in inflammatory gene regulation. RESULTS All lines maintained CFTR mRNA production and formation of tight junctions. CFTR+ lines displayed short circuit currents in response to forskolin, while the CFTR- lines did not. Baseline expression of cytokines IL6 and CXCL8 (IL8) was not different between the lines regardless of CFTR genotype. All lines responded to TNFα and IL1β by increasing IL6 and CXCL8 mRNA levels, but the CFTR- lines produced more CXCL8 mRNA than the CFTR+ lines. Transcriptomes of 6 CFTR- and 6 CFTR+ lines, before and after stimulation by TNFα, were compared for differential expression as a function of CFTR genotype. While some genes appeared to be differentially expressed simply because of CFTR's absence, others required stimulation for differences to be apparent. CONCLUSION Together, these data suggest cells respond to CFTR's absence by modulating transcriptional networks, some of which are only apparent when cells are exposed to different environmental contexts, such as inflammation. With regards to inflammation, these data suggest a model in which CFTR's absence leads to a poised, pro-inflammatory state of cells that is only revealed by stimulation.
Collapse
Affiliation(s)
- Shuyu Hao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Erica A Roesch
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Aura Perez
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Rebecca L Weiner
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Leigh C Henderson
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Linda Cummings
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Paul Consiglio
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Joseph Pajka
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Amy Eisenberg
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Lauren Yeh
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Calvin U Cotton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America.
| |
Collapse
|
15
|
Shi L, Fang B, Yong Y, Li X, Gong D, Li J, Yu T, Gooneratne R, Gao Z, Li S, Ju X. Chitosan oligosaccharide-mediated attenuation of LPS-induced inflammation in IPEC-J2 cells is related to the TLR4/NF-κB signaling pathway. Carbohydr Polym 2019; 219:269-279. [PMID: 31151525 DOI: 10.1016/j.carbpol.2019.05.036] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 04/16/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023]
Abstract
The protective mechanism of chitosan oligosaccharide (COS) against lipopolysaccharides (LPS) -induced inflammatory responses in IPEC-J2 and in mice with DSS dextran sulfate sodium (DSS) -induced colitis is reported. Upon exposure to LPS, the proliferation rate of IPEC-J2 cells markedly decreased, and epithelial cell integrity was compromised. However, COS pretreatment significantly reduced these changes. Low-concentration (200 μg/mL) COS up-regulated Toll-like receptor 4 (TLR4) and nuclear p65 expression, but inhibited LPS-induced expression of nuclear p65, IL-6, and IL-8. Addition of the TLR4 inhibitor reduced nuclear p65, IL-6, and IL-8 expression in IPEC-J2 cells exposed to COS or LPS alone, and a slight up-regulation in nuclear p65 was observed in COS and LPS co-treated cells. Medium-dose COS (600 mg/kg/d) protected against DSS-induced colitis, in which TLR4 and nuclear p65 expression levels were decreased. We postulate that the prevention of both LPS- and DSS -induced inflammatory responses in IPEC-J2 cells and mice by COS are related to the inhibition of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lin Shi
- Department of Animal Science, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518018, China
| | - Biao Fang
- Department of Animal Science, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Yanhong Yong
- Department of Veterinary Medicine, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Xuewen Li
- Department of Veterinary Medicine, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Dongliang Gong
- Department of Veterinary Medicine, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Junyu Li
- Department of Animal Science, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Tianyue Yu
- Department of Animal Science, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China
| | - Ravi Gooneratne
- Department of Wine, Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln 7647, New Zealand
| | - Zhenhua Gao
- Department of Animal Science, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.
| | - Sidong Li
- College of Chemistry and Environment, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China.
| | - Xianghong Ju
- Department of Veterinary Medicine, College of Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, China; Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518018, China.
| |
Collapse
|
16
|
Chitosan oligosaccharide (COS): An overview. Int J Biol Macromol 2019; 129:827-843. [PMID: 30708011 DOI: 10.1016/j.ijbiomac.2019.01.192] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/14/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023]
|
17
|
Mastrogiovanni F, Mukhopadhya A, Lacetera N, Ryan MT, Romani A, Bernini R, Sweeney T. Anti-Inflammatory Effects of Pomegranate Peel Extracts on In Vitro Human Intestinal Caco-2 Cells and Ex Vivo Porcine Colonic Tissue Explants. Nutrients 2019; 11:E548. [PMID: 30841512 PMCID: PMC6471410 DOI: 10.3390/nu11030548] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/24/2019] [Accepted: 02/26/2019] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to determine the anti-inflammatory potential of pomegranate peel extracts (PPE) prepared from waste material of pomegranate juice production both in vitro on Caco-2 cells and ex vivo using porcine colonic tissue explants. Caco-2 cells were stimulated in vitro by TNF and colonic tissue explants were stimulated ex vivo with lipopolysaccharide (LPS). Both tissues were co-treated with PPE at 0, 1.0, 2.5, 5.0, 10 and 25 μg/mL. The secretion of CXCL8 in the supernatant of both experiments was determined by enzyme linked immunosorbent assay (ELISA) and the relative expression of inflammatory cytokines were evaluated in the colonic tissue by quantitative polymerase chain reaction (QPCR). The 2.5 to 25 μg/mL of PPE suppressed CXCL8 (p < 0.001) in the Caco-2 cells, whereas CXCL8 production was suppressed by only 5 and 25 μg/mL (p < 0.01) of PPE in the colonic explants. The 5 μg/mL of PPE also suppressed the expression of IL1A (p < 0.05), IL6 (p < 0.01) and CXCL8 (p < 0.05) in LPS challenged colonic tissues compared to controls. In conclusion, the 5 μg/mL of PPE consistently elicits strong anti-inflammatory activity. These results support the potential of bioactive compounds from the waste peel of pomegranate in terms of their anti-inflammatory activity in cells and tissues of the gastrointestinal tract.
Collapse
Affiliation(s)
- Fabio Mastrogiovanni
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy.
- School of Veterinary Medicine, University College Dublin 4, D04 V1W8 Dublin, Ireland.
| | - Anindya Mukhopadhya
- School of Veterinary Medicine, University College Dublin 4, D04 V1W8 Dublin, Ireland.
| | - Nicola Lacetera
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy.
| | - Marion T Ryan
- School of Veterinary Medicine, University College Dublin 4, D04 V1W8 Dublin, Ireland.
| | - Annalisa Romani
- Department of Statistics, Computing, Applications "G. Parenti" (DISIA), PHYTOLAB, University of Florence, 50019 Florence, Italy.
| | - Roberta Bernini
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100 Viterbo, Italy.
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin 4, D04 V1W8 Dublin, Ireland.
| |
Collapse
|
18
|
Chitooligosaccharides and their biological activities: A comprehensive review. Carbohydr Polym 2018; 184:243-259. [DOI: 10.1016/j.carbpol.2017.12.067] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/10/2017] [Accepted: 12/24/2017] [Indexed: 01/11/2023]
|
19
|
Zehra S, Khambati I, Vierhout M, Mian MF, Buck R, Forsythe P. Human Milk Oligosaccharides Attenuate Antigen-Antibody Complex Induced Chemokine Release from Human Intestinal Epithelial Cell Lines. J Food Sci 2018; 83:499-508. [PMID: 29377120 DOI: 10.1111/1750-3841.14039] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/24/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022]
Abstract
There has been increased interest in the use of dietary ingredients, including prebiotics such as human-milk oligosaccharides (HMOs), as therapeutic strategies for food allergy. Understanding the mechanisms underlying the beneficial effects of HMOs is important to realizing their therapeutic potential. Here we demonstrate that the HMO, 6'-sialyllactose (6'SL) inhibited chemokine (IL-8 and CCL20) release from T-84 and HT-29 cells stimulated with antigen-antibody complex, TNFα or PGE2 ; an effect that was PPARγ dependent and associated with decreased activity of the transcription factors AP-1 and NFκB. In contrast, 2'-fucosyllactose (2'FL) selectively inhibited CCL20 release in response to antigen antibody complex in a PPARγ independent manner. This study reinforces the concept that structurally different oligosaccharides have distinct biological activities and identifies, for the first time, that the HMOs, 6'SL, and 2'FL, modulate human epithelial cell responses related to allergic disease. These findings encourage further investigation of the therapeutic potential of specific HMOs in food allergy. PRACTICAL APPLICATION This study provides evidence for direct effects of HMOs in addition to their prebiotic role and demonstrates, for the first time, modulation of Ag-IgE complex activation of human epithelial cells that may have important implications for food-allergy. The study also reinforces the concept that structurally different oligosaccharides have distinct biological activities. In determining the composition of infant formula, addition of oligosaccharides with specific structures may provide direct modulation of immune responses and potentially attenuate symptoms or development of food allergy.
Collapse
Affiliation(s)
- Sehrish Zehra
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Ibrahim Khambati
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Megan Vierhout
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - M Firoz Mian
- Dept. of Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada
| | - Rachael Buck
- Abbott Nutrition: a Division of Abbott Laboratories, Columbus, Ohio, U.S.A
| | - Paul Forsythe
- Dept. of Medicine, Pathology and Molecular Medicine, McMaster Univ., Hamilton, Ont., Canada.,The Firestone Inst. for Respiratory Research, McMaster Univ., Hamilton, Ont., Canada
| |
Collapse
|
20
|
Zhai X, Yang X, Zou P, Shao Y, Yuan S, Abd El-Aty AM, Wang J. Protective Effect of Chitosan Oligosaccharides Against Cyclophosphamide-Induced Immunosuppression and Irradiation Injury in Mice. J Food Sci 2018; 83:535-542. [PMID: 29350748 DOI: 10.1111/1750-3841.14048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/05/2017] [Accepted: 12/16/2017] [Indexed: 12/22/2022]
Abstract
Chitosan oligosaccharides (COS), hydrolyzed products of chitosan, was found to display various biological activities. Herein, we assessed the immunostimulatory activity of COS both in in vitro and in vivo studies. In vitro cytotoxicity studies to murine macrophage RAW264.7 revealed that COS is safe even at the maximum tested concentration of 1000 μg/mL. It also stimulates the production of nitric oxide (NO) and tumor necrosis factor (TNF-α) and enhances the phagocytosis in COS-stimulated RAW264.7. We have shown that the COS could significantly (P < 0.05) restore the reduced immune organs indices, phagocytic index, lymphocyte proliferation, natural killer cell activity, and antioxidant enzyme activities in a cyclophosphamide-induced immunosuppressed mice model. COS can also improve the survival rate in irradiation injury mice and significantly (P < 0.05) increased the spleen indices and up-regulates the CD4+/CD8+ ratio in splenocytes. In sum, the aforementioned results suggest that COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development. PRACTICAL APPLICATION COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development.
Collapse
Affiliation(s)
- Xingchen Zhai
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China.,the Dept. of Pharmacology and Toxicology, Beijing Inst. of Radiation Medicine, 100081 Beijing, PR China
| | - Xin Yang
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China
| | - Pan Zou
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| | - Yong Shao
- Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| | - Shoujun Yuan
- the Dept. of Pharmacology and Toxicology, Beijing Inst. of Radiation Medicine, 100081 Beijing, PR China
| | - A M Abd El-Aty
- Dept. of Pharmacology, Faculty of Veterinary Medicine, Cairo Univ., 12211 Giza, Egypt
| | - Jing Wang
- Dept. of Food Sciences and Engineering, School of Chemistry and Chemical Engineering, Harbin Inst. of Technology, 150090 Harbin, PR China.,Key Lab. of Agro-Product Quality and Safety, Inst. of Quality Standard and Testing Technology for Agro-Product, Chinese Acad. of Agricultural Sciences, 100081 Beijing, PR China
| |
Collapse
|
21
|
Zhao Y, Wang Y, Gong J, Yang L, Niu C, Ni X, Wang Y, Peng S, Gu X, Sun C, Yang Y. Chitosan degradation products facilitate peripheral nerve regeneration by improving macrophage-constructed microenvironments. Biomaterials 2017; 134:64-77. [DOI: 10.1016/j.biomaterials.2017.02.026] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022]
|
22
|
Yang JW, Tian G, Chen DW, Yao Y, He J, Zheng P, Mao XB, Yu J, Huang ZQ, Yu B. Involvement of PKA signalling in anti-inflammatory effects of chitosan oligosaccharides in IPEC-J2 porcine epithelial cells. J Anim Physiol Anim Nutr (Berl) 2017; 102:252-259. [PMID: 28299836 DOI: 10.1111/jpn.12686] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 01/05/2017] [Indexed: 12/24/2022]
Abstract
Weaning is characterized by intestinal inflammation, which is a big challenge in pig industry. Control of intestinal inflammation is important for improvement of growth performance and health. Therefore, the study was focused on the anti-inflammatory activity of low-molecular-weight chitosan oligosaccharide (LCOS) in a porcine small intestinal epithelial cell line (IPEC-J2). The results showed that TNF-α, as inflammation inducer, significantly upregulated the mRNA expression of IL-8 and MCP-1. Afterwards, LCOS significantly attenuated mRNA expression of IL-8 and MCP-1 induced by TNF-α in the cells. Mannose (MAN), as ligand of mannose receptor, had no effect on the anti-inflammatory activity of LCOS, which suggested that mannose receptor may not involve in the anti-inflammatory activity of LCOS in IPEC-J2 cells. Interestingly, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide 2HCl hydrate (H89), as PKA (protein kinase A)-specific inhibitor, reversed the mRNA expression of IL-8 when co-cultured with LCOS. Furthermore, LCOS concentration dependent downregulated the mRNA expression of claudin-1 compared with TNF-α treatment. However, the trans-epithelial electric resistance (TEER) was not affected by LCOS when co-cultured with TNF-α in 3 hr. In conclusion, LCOS have a potent anti-inflammatory activity, and as a feed additives, may be useful for the inhibition of inflammatory process in weaning period of pigs with intestinal inflammation occurring.
Collapse
Affiliation(s)
- J W Yang
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - G Tian
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - D W Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - Y Yao
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - J He
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - P Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - X B Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - J Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - Z Q Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| | - B Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Yaan, Sichuan, China
| |
Collapse
|
23
|
Shen Y, Zhou M, Yan J, Gong Z, Xiao Y, Zhang C, Du P, Chen Y. miR-200b inhibits TNF-α-induced IL-8 secretion and tight junction disruption of intestinal epithelial cells in vitro. Am J Physiol Gastrointest Liver Physiol 2017; 312:G123-G132. [PMID: 27979826 DOI: 10.1152/ajpgi.00316.2016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 01/31/2023]
Abstract
UNLABELLED Inflammatory bowel diseases (IBDs) are chronic, inflammatory disorders of the gastrointestinal tract with unclear etiologies. Intestinal epithelial cells (IECs), containing crypt and villus enterocytes, occupy a critical position in the pathogenesis of IBDs and are a major producer of immunoregulatory cytokines and a key component of the intact epithelial barrier. Previously, we have reported that miR-200b is involved in the progression of IBDs and might maintain the integrity of the intestinal epithelial barrier via reducing the loss of enterocytes. In this study, we further investigated the impact of miR-200b on intestinal epithelial inflammation and tight junctions in two distinct differentiated states of Caco-2 cells after TNF-α treatment. We demonstrated that TNF-α-enhanced IL-8 expression was decreased by microRNA (miR)-200b in undifferentiated IECs. Simultaneously, miR-200b could alleviate TNF-α-induced tight junction (TJ) disruption in well-differentiated IECs by reducing the reduction in the transepithelial electrical resistance (TEER), inhibiting the increase in paracellular permeability, and preventing the morphological redistribution of the TJ proteins claudin 1 and ZO-1. The expression levels of the JNK/c-Jun/AP-1 and myosin light chain kinase (MLCK)/phosphorylated myosin light chain (p-MLC) pathways were attenuated in undifferentiated and differentiated enterocytes, respectively. Furthermore, a dual-luciferase reporter gene detection system provided direct evidence that c-Jun and MLCK were the specific targets of miR-200b. Collectively, our results highlighted that miR-200b played a positive role in IECs via suppressing intestinal epithelial IL-8 secretion and attenuating TJ damage in vitro, which suggested that miR-200b might be a promising strategy for IBD therapy. NEW & NOTEWORTHY This was the first time that the inhibitory role of miR-200b on intestinal epithelial inflammation and paracellular permeability has been reported. Moreover, we further divided the intestinal epithelial cells (IECs) into two differentiated conditions and investigated the distinct impacts of miR-200b. Finally, we put forward and proved that myosin light chain kinase (MLCK) was a novel target of miR-200b.
Collapse
Affiliation(s)
- Yujie Shen
- Department of Gastroenterology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhou
- Department of Gastroenterology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junkai Yan
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; and
| | - Zizhen Gong
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; and
| | - Yongtao Xiao
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; and
| | - Cong Zhang
- Department of Gastroenterology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Du
- Department of Colorectal Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingwei Chen
- Department of Gastroenterology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China;
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; and
| |
Collapse
|
24
|
Muanprasat C, Chatsudthipong V. Chitosan oligosaccharide: Biological activities and potential therapeutic applications. Pharmacol Ther 2016; 170:80-97. [PMID: 27773783 DOI: 10.1016/j.pharmthera.2016.10.013] [Citation(s) in RCA: 323] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chitosan oligosaccharide (COS) is an oligomer of β-(1➔4)-linked d-glucosamine. COS can be prepared from the deacetylation and hydrolysis of chitin, which is commonly found in the exoskeletons of arthropods and insects and the cell walls of fungi. COS is water soluble, non-cytotoxic, readily absorbed through the intestine and mainly excreted in the urine. Of particular importance, COS and its derivatives have been demonstrated to possess several biological activities including anti-inflammation, immunostimulation, anti-tumor, anti-obesity, anti-hypertension, anti-Alzheimer's disease, tissue regeneration promotion, drug and DNA delivery enhancement, anti-microbial, anti-oxidation and calcium-absorption enhancement. The mechanisms of actions of COS have been found to involve the modulation of several important pathways including the suppression of nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) and the activation of AMP-activated protein kinase (AMPK). This review summarizes the current knowledge of the preparation methods, pharmacokinetic profiles, biological activities, potential therapeutic applications and safety profiles of COS and its derivatives. In addition, future research directions are discussed.
Collapse
Affiliation(s)
- Chatchai Muanprasat
- Excellent Center for Drug Discovery and Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand.
| | - Varanuj Chatsudthipong
- Excellent Center for Drug Discovery and Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand
| |
Collapse
|
25
|
Molecular Weight-Dependent Immunostimulative Activity of Low Molecular Weight Chitosan via Regulating NF-κB and AP-1 Signaling Pathways in RAW264.7 Macrophages. Mar Drugs 2016; 14:md14090169. [PMID: 27657093 PMCID: PMC5039540 DOI: 10.3390/md14090169] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 11/17/2022] Open
Abstract
Chitosan and its derivatives such as low molecular weight chitosans (LMWCs) have been found to possess many important biological properties, such as antioxidant and antitumor effects. In our previous study, LMWCs were found to elicit a strong immunomodulatory response in macrophages dependent on molecular weight. Herein we further investigated the molecular weight-dependent immunostimulative activity of LMWCs and elucidated its mechanism of action on RAW264.7 macrophages. LMWCs (3 kDa and 50 kDa of molecular weight) could significantly enhance the mRNA expression levels of COX-2, IL-10 and MCP-1 in a molecular weight and concentration-dependent manner. The results suggested that LMWCs elicited a significant immunomodulatory response, which was dependent on the dose and the molecular weight. Regarding the possible molecular mechanism of action, LMWCs promoted the expression of the genes of key molecules in NF-κB and AP-1 pathways, including IKKβ, TRAF6 and JNK1, and induced the phosphorylation of protein IKBα in RAW264.7 macrophage. Moreover, LMWCs increased nuclear translocation of p65 and activation of activator protein-1 (AP-1, C-Jun and C-Fos) in a molecular weight-dependent manner. Taken together, our findings suggested that LMWCs exert immunostimulative activity via activation of NF-κB and AP-1 pathways in RAW264.7 macrophages in a molecular weight-dependent manner and that 3 kDa LMWC shows great potential as a novel agent for the treatment of immune suppression diseases and in future vaccines.
Collapse
|
26
|
|
27
|
Mukhopadhya A, Noronha N, Bahar B, Ryan MT, Murray BA, Kelly PM, O'Loughlin IB, O'Doherty JV, Sweeney T. The anti-inflammatory potential of a moderately hydrolysed casein and its 5 kDa fraction in in vitro and ex vivo models of the gastrointestinal tract. Food Funct 2015; 6:612-621. [PMID: 25536369 DOI: 10.1039/c4fo00689e] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Bioactive peptides from milk can impart a wide range of physiological benefits without the allergies and intolerance associated with the consumption of whole milk. The objective of this study was to characterise the anti-inflammatory properties of intact sodium caseinate (NaCAS), a moderately hydrolysed NaCAS enzyme hydrolysate (EH) and its 5 kDa fraction (5kDaR), in both in vitro and ex vivo systems. In vitro, Caco-2 cells were stimulated with tumor necrosis factor (TNF) α and co-treated ± casein hydrolysates or dexamethasone (control). The inflammatory marker interleukin (IL)-8 was measured by ELISA in the supernatant at 24 h. Ex vivo, porcine colonic tissues were stimulated with lipopolysaccharide (LPS) and co-treated with casein hydrolysates for 3 h from which the relative expression of a panel of cytokines was measured in vitro. While the steroid dexamethasone brought about a 41.6% reduction in the IL-8 concentration in the supernatant, the 5kDaR reduced IL-8 by 59% (P < 0.05) when compared to the TNFα stimulated Caco-2 cells. In the ex vivo system, 5kDaR was associated with decreases in IL-1α, IL-1β, IL-8 and TGF-β expression and an increase in IL-17 expression (P < 0.05) relative to the LPS challenged tissues. We concluded, that a 5 kDa casein fraction demonstrates potent anti-inflammatory effects both in in vitro and ex vivo models of the gastrointestinal tract.
Collapse
Affiliation(s)
- A Mukhopadhya
- School of Veterinary Medicine, UCD, Belfield, Dublin.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jiang M, Guo Z, Wang C, Yang Y, Liang X, Ding F. Neural activity analysis of pure chito-oligomer components separated from a mixture of chitooligosaccharides. Neurosci Lett 2014; 581:32-6. [DOI: 10.1016/j.neulet.2014.08.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/19/2014] [Accepted: 08/05/2014] [Indexed: 10/24/2022]
|
29
|
The Beneficial Effect of Chitooligosaccharides on Cell Behavior and Function of Primary Schwann Cells is Accompanied by Up-Regulation of Adhesion Proteins and Neurotrophins. Neurochem Res 2014; 39:2047-57. [DOI: 10.1007/s11064-014-1387-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/30/2014] [Accepted: 07/11/2014] [Indexed: 01/12/2023]
|
30
|
Giusti S, Sbrana T, La Marca M, Di Patria V, Martinucci V, Tirella A, Domenici C, Ahluwalia A. A novel dual-flow bioreactor simulates increased fluorescein permeability in epithelial tissue barriers. Biotechnol J 2014; 9:1175-84. [PMID: 24756869 DOI: 10.1002/biot.201400004] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/28/2014] [Accepted: 04/21/2014] [Indexed: 12/27/2022]
Abstract
Permeability studies across epithelial barriers are of primary importance in drug delivery as well as in toxicology. However, traditional in vitro models do not adequately mimic the dynamic environment of physiological barriers. Here, we describe a novel two-chamber modular bioreactor for dynamic in vitro studies of epithelial cells. The fluid dynamic environment of the bioreactor was characterized using computational fluid dynamic models and measurements of pressure gradients for different combinations of flow rates in the apical and basal chambers. Cell culture experiments were then performed with fully differentiated Caco-2 cells as a model of the intestinal epithelium, comparing the effect of media flow applied in the bioreactor with traditional static transwells. The flow increases barrier integrity and tight junction expression of Caco-2 cells with respect to the static controls. Fluorescein permeability increased threefold in the dynamic system, indicating that the stimulus induced by flow increases transport across the barrier, closely mimicking the in vivo situation. The results are of interest for studying the influence of mechanical stimuli on cells, and underline the importance of developing more physiologically relevant in vitro tissue models. The bioreactor can be used to study drug delivery, chemical, or nanomaterial toxicity and to engineer barrier tissues.
Collapse
Affiliation(s)
- Serena Giusti
- Research Center "E. Piaggio, " University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Li Y, Xu Q, Wei P, Cheng L, Peng Q, Li S, Yin H, Du Y. Chitosan oligosaccharides downregulate the expression of E-selectin and ICAM-1 induced by LPS in endothelial cells by inhibiting MAP kinase signaling. Int J Mol Med 2013; 33:392-400. [PMID: 24336934 DOI: 10.3892/ijmm.2013.1589] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 11/25/2013] [Indexed: 11/05/2022] Open
Abstract
The expression of adhesion molecules in endothelial cells elicited by lipopolysaccharide (LPS) is involved in the adhesive interaction between endothelial cells and monocytes in inflammation. In this study, in order to characterize the anti-inflammatory effects of chitosan oligosaccharides (COS) on LPS‑induced inflammation and to elucidate the underlying mechanisms, the mRNA levels of E-selectin and intercellular adhesion molecule-1 (ICAM-1) were measured in porcine iliac artery endothelial cells (PIECs). When these cells were treated with COS, the LPS-induced mRNA expression of E-selectin and ICAM-1 was reduced through the inhibition of the signal transduction cascade, p38 mitogen‑activated protein kinase (MAPK)/extracellular regulated protein kinase 1/2 (ERK1/2) and nuclear factor-κB (NF-κB). Moreover, through the inhibition of p38 MAPK and ERK1/2, COS suppressed the LPS-induced NF-κB p65 translocation. We found that COS suppressed the phosphorylation of p38 MAPK and the translocation of NF-κB p65 into the nucleus in a dose-dependent manner, and inhibited the adhesion of U973 cells to PIECs. Based on these results, it can be concluded that COS downregulate the expression of E-selectin and ICAM-1 by inhibiting the phosphorylation of MAPKs and the activation of NF-κB in LPS-treated PIECs. Our study demonstrates the valuable anti-inflammatory properties of COS.
Collapse
Affiliation(s)
- Yu Li
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Qingsong Xu
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Peng Wei
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Likun Cheng
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Qiang Peng
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Shuguang Li
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Heng Yin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| | - Yuguang Du
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, P.R. China
| |
Collapse
|
32
|
Pang G, Xie J, Chen Q, Hu Z. How functional foods play critical roles in human health. FOOD SCIENCE AND HUMAN WELLNESS 2012. [DOI: 10.1016/j.fshw.2012.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Gray LT, Fong KK, Pavelitz T, Weiner AM. Tethering of the conserved piggyBac transposase fusion protein CSB-PGBD3 to chromosomal AP-1 proteins regulates expression of nearby genes in humans. PLoS Genet 2012; 8:e1002972. [PMID: 23028371 PMCID: PMC3459987 DOI: 10.1371/journal.pgen.1002972] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/08/2012] [Indexed: 12/01/2022] Open
Abstract
The CSB-PGBD3 fusion protein arose more than 43 million years ago when a 2.5-kb piggyBac 3 (PGBD3) transposon inserted into intron 5 of the Cockayne syndrome Group B (CSB) gene in the common ancestor of all higher primates. As a result, full-length CSB is now coexpressed with an abundant CSB-PGBD3 fusion protein by alternative splicing of CSB exons 1-5 to the PGBD3 transposase. An internal deletion of the piggyBac transposase ORF also gave rise to 889 dispersed, 140-bp MER85 elements that were mobilized in trans by PGBD3 transposase. The CSB-PGBD3 fusion protein binds MER85s in vitro and induces a strong interferon-like innate antiviral immune response when expressed in CSB-null UVSS1KO cells. To explore the connection between DNA binding and gene expression changes induced by CSB-PGBD3, we investigated the genome-wide DNA binding profile of the fusion protein. CSB-PGBD3 binds to 363 MER85 elements in vivo, but these sites do not correlate with gene expression changes induced by the fusion protein. Instead, CSB-PGBD3 is enriched at AP-1, TEAD1, and CTCF motifs, presumably through protein-protein interactions with the cognate transcription factors; moreover, recruitment of CSB-PGBD3 to AP-1 and TEAD1 motifs correlates with nearby genes regulated by CSB-PGBD3 expression in UVSS1KO cells and downregulated by CSB rescue of mutant CS1AN cells. Consistent with these data, the N-terminal CSB domain of the CSB-PGBD3 fusion protein interacts with the AP-1 transcription factor c-Jun and with RNA polymerase II, and a chimeric CSB-LacI construct containing only the N-terminus of CSB upregulates many of the genes induced by CSB-PGBD3. We conclude that the CSB-PGBD3 fusion protein substantially reshapes the transcriptome in CS patient CS1AN and that continued expression of the CSB-PGBD3 fusion protein in the absence of functional CSB may affect the clinical presentation of CS patients by directly altering the transcriptional program.
Collapse
Affiliation(s)
| | | | | | - Alan M. Weiner
- Department of Biochemistry, School of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|