1
|
Fu S, Lin X, Lu X, Qu Y, Chen H, Zheng S, Li Z, Jiao Y, Wang Q, Yang C, Deng Y. NF-κB inhibitor PDTC involved in regulating the transplantation immunity in the pearl oyster Pinctada fucata martensii. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110175. [PMID: 39909120 DOI: 10.1016/j.fsi.2025.110175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/21/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Nuclear factor kappa B (NF-κB) is involved in various important biological processes, such as inflammation, apoptosis, and cell proliferation. Here, we analyzed the function of NF-κB in transplantation immunity and pearl formation using its inhibitor, pyrrolidinedithiocarbamic acid (PDTC), in the pearl oyster Pinctada fucata martensii. The levels of pro-inflammatory factors (IL-17 and TNF-α) were lower and activity of antioxidant-related enzymes was higher in the transplanted pearl oysters pre-treated with PDTC than in transplanted pearl oysters pre-treated with phosphate-buffered saline (PBS). Transcriptomic analysis showed that PDTC pre-treatment alleviated the immune stimulation caused by transplantation, preserved normal expression of ribosome-related genes, and inhibited the activation of apoptosis and the NF-κB signaling pathway induced by transplantation. Additionally, RIG-I-like receptor, MAPK, Toll-like receptor and NOD-like receptor signaling pathways were inhibited after PDTC treatment. A 30-day pearl cultivation experiment demonstrated a significantly higher nucleus retention rate in transplanted pearl oysters that were pre-treated with PDTC compared to the control group. These results indicate that PDTC treatment suppressed immune-related pathways, thereby alleviating the immune rejection response caused by transplantation and potentially optimizing pearl production. Our results provide valuable information for optimizing pearl cultivation in P. f. martensii.
Collapse
Affiliation(s)
- Shirong Fu
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Xinyi Lin
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Xiaowen Lu
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Youmei Qu
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Hongxi Chen
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Shilin Zheng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Zhihan Li
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Yu Jiao
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China.
| | - Qingheng Wang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, China; Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Zhanjiang, 524088, China
| | - Chuangye Yang
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China
| | - Yuewen Deng
- Fishery College, Guangdong Ocean University, Zhanjiang, 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang, 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang, 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, China; Guangdong Marine Ecology Early Warning and Monitoring Laboratory, Zhanjiang, 524088, China
| |
Collapse
|
2
|
Di Mattia M, Sallese M, Lopetuso LR. Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease. Acta Physiol (Oxf) 2025; 241:e14284. [PMID: 39822064 DOI: 10.1111/apha.14284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Different physiological and pathological situations can produce alterations in the cell's endoplasmic reticulum (ER), leading to a condition known as ER stress, which can trigger an intricate intracellular signal transduction system known as the unfolded protein response (UPR). UPR is primarily tailored to restore proteostasis and ER equilibrium; otherwise, if ER stress persists, it can cause programmed cell death as a cytoprotective mechanism and drive inflammatory processes. Therefore, since intestinal cells strongly rely on UPR for their biological functions and unbalanced UPR has been linked to inflammatory, metabolic, and immune disorders, here we discussed the role of the UPR within the intestinal tract, focusing on the UPR contribution to inflammatory bowel disease development. Importantly, we also highlighted the promising potential of UPR components as therapeutic targets for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
3
|
Yang H, Wang Z, Li L, Wang X, Wei X, Gou S, Ding Z, Cai Z, Ling Q, Hoffmann PR, He J, Liu F, Huang Z. Mannose coated selenium nanoparticles normalize intestinal homeostasis in mice and mitigate colitis by inhibiting NF-κB activation and enhancing glutathione peroxidase expression. J Nanobiotechnology 2024; 22:613. [PMID: 39385176 PMCID: PMC11465824 DOI: 10.1186/s12951-024-02861-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
Impaired intestinal homeostasis is a major pathological feature of inflammatory bowel diseases (IBD). Mannose and selenium (Se) both demonstrate potential anti-inflammatory and anti-oxidative properties. However, most lectin receptors bind free monosaccharide ligands with relatively low affinity and most Se species induce side effects beyond a very narrow range of dosage. This has contributed to a poorly explored therapies for IBD that combine mannose and Se to target intestinal epithelial cells (IECs) for normalization gut homeostasis. Herein, a facile and safe strategy for ulcerative colitis (UC) treatment was developed using optimized, mannose-functionalized Se nanoparticles (M-SeNPs) encapsulated within a colon-targeted hydrogel delivery system containing alginate (SA) and chitosan (CS). This biocompatible nanosystem was efficiently taken up by IECs and led to increased expression of Se-dependent glutathione peroxidases (GPXs), thereby modulating IECs' immune response. Using a mouse model of DSS-induced colitis, (CS/SA)-embedding M-SeNPs (C/S-MSe) were found to mitigate oxidative stress and inflammation through the inhibition of the NF-kB pathway in the colon. This stabilized mucosal homeostasis of IECs and ameliorated colitis-related symptoms, thereby providing a potential new approach for treatment of IBD.
Collapse
Affiliation(s)
- Hui Yang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhiyao Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Lixin Li
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xing Wang
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xian Wei
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Shan Gou
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zimo Ding
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Zhihui Cai
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qinjie Ling
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Peter R Hoffmann
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, U.S.A
| | - Jingjun He
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Fei Liu
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Zhi Huang
- Department of Health Management of the Guangdong Second Provincial General Hospital & Postdoctoral Research Station of Basic Medicine of the School of Medicine, Jinan University, Guangzhou, 510632, China.
- Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Liu Y, Lin S, Wang C, Li T, Zheng G, Sun W, An L, Bai Y, Wu F. Sex-Specific Effects of Environmental Exposure to the Antimicrobial Agents Benzalkonium Chloride and Triclosan on the Gut Microbiota and Health of Zebrafish ( Danio rerio). ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:15450-15462. [PMID: 39141879 DOI: 10.1021/acs.est.4c03205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
The use of disinfectants containing benzalkonium chloride (BAC) has become increasingly widespread in response to triclosan (TCS) restrictions and the COVID-19 pandemic, leading to the increasing presence of BAC in aquatic ecosystems. However, the potential environmental health impacts of BAC on fish remain poorly explored. In this study, we show that BAC and TCS can induce the gut dysbiosis in zebrafish (Danio rerio), with substantial effects on health. Breeding pairs of adult zebrafish were exposed to environmentally relevant concentrations of BAC and TCS (0.4-40 μg/L) for 42 days. Both BAC and TCS exposure perturbed the gut microbiota, triggering the classical NF-κB signaling pathway and resulting in downstream pathological toxicity associated with inflammatory responses, histological damage, inhibited ingestion, and decreased survival. These effects were dose-dependent and sex-specific, as female zebrafish were more susceptible than male zebrafish. Furthermore, we found that BAC induced toxicity to a greater extent than the restricted TCS at environmentally relevant concentrations, which is particularly concerning. Our results suggest that environmental exposure to antimicrobial chemicals can have ecological consequences by perturbing the gut microbiota, a previously underappreciated target of such chemicals. Rigorous ecological analysis should be conducted before widely introducing replacement antimicrobial compounds into disinfecting products.
Collapse
Affiliation(s)
- Yueyue Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
- College of Water Science, Beijing Normal University, Beijing 100875, China
| | - Siyi Lin
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chen Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Tong Li
- Institute of Quality Standards and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Guomao Zheng
- School of Environmental Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wen Sun
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Lihui An
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Yingchen Bai
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| |
Collapse
|
5
|
Haque M, Kaminsky L, Abdulqadir R, Engers J, Kovtunov E, Rawat M, Al-Sadi R, Ma TY. Lactobacillus acidophilus inhibits the TNF-α-induced increase in intestinal epithelial tight junction permeability via a TLR-2 and PI3K-dependent inhibition of NF-κB activation. Front Immunol 2024; 15:1348010. [PMID: 39081324 PMCID: PMC11286488 DOI: 10.3389/fimmu.2024.1348010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. METHODS The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. RESULTS AND CONCLUSION Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Mohammad Haque
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Lauren Kaminsky
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Raz Abdulqadir
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Jessica Engers
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Evgeny Kovtunov
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Manmeet Rawat
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Rana Al-Sadi
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Thomas Y. Ma
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
6
|
Drif AI, Yücer R, Damiescu R, Ali NT, Abu Hagar TH, Avula B, Khan IA, Efferth T. Anti-Inflammatory and Cancer-Preventive Potential of Chamomile ( Matricaria chamomilla L.): A Comprehensive In Silico and In Vitro Study. Biomedicines 2024; 12:1484. [PMID: 39062057 PMCID: PMC11275008 DOI: 10.3390/biomedicines12071484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND AND AIM Chamomile tea, renowned for its exquisite taste, has been appreciated for centuries not only for its flavor but also for its myriad health benefits. In this study, we investigated the preventive potential of chamomile (Matricaria chamomilla L.) towards cancer by focusing on its anti-inflammatory activity. METHODS AND RESULTS A virtual drug screening of 212 phytochemicals from chamomile revealed β-amyrin, β-eudesmol, β-sitosterol, apigenin, daucosterol, and myricetin as potent NF-κB inhibitors. The in silico results were verified through microscale thermophoresis, reporter cell line experiments, and flow cytometric determination of reactive oxygen species and mitochondrial membrane potential. An oncobiogram generated through comparison of 91 anticancer agents with known modes of action using the NCI tumor cell line panel revealed significant relationships of cytotoxic chamomile compounds, lupeol, and quercetin to microtubule inhibitors. This hypothesis was verified by confocal microscopy using α-tubulin-GFP-transfected U2OS cells and molecular docking of lupeol and quercetin to tubulins. Both compounds induced G2/M cell cycle arrest and necrosis rather than apoptosis. Interestingly, lupeol and quercetin were not involved in major mechanisms of resistance to established anticancer drugs (ABC transporters, TP53, or EGFR). Performing hierarchical cluster analyses of proteomic expression data of the NCI cell line panel identified two sets of 40 proteins determining sensitivity and resistance to lupeol and quercetin, further pointing to the multi-specific nature of chamomile compounds. Furthermore, lupeol, quercetin, and β-amyrin inhibited the mRNA expression of the proinflammatory cytokines IL-1β and IL6 in NF-κB reporter cells (HEK-Blue Null1). Moreover, Kaplan-Meier-based survival analyses with NF-κB as the target protein of these compounds were performed by mining the TCGA-based KM-Plotter repository with 7489 cancer patients. Renal clear cell carcinomas (grade 3, low mutational rate, low neoantigen load) were significantly associated with shorter survival of patients, indicating that these subgroups of tumors might benefit from NF-κB inhibition by chamomile compounds. CONCLUSION This study revealed the potential of chamomile, positioning it as a promising preventive agent against inflammation and cancer. Further research and clinical studies are recommended.
Collapse
Affiliation(s)
- Assia I. Drif
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Rümeysa Yücer
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Roxana Damiescu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Nadeen T. Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Tobias H. Abu Hagar
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| | - Bharati Avula
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research (NCNPR), School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA; (B.A.); (I.A.K.)
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; (A.I.D.); (R.Y.); (R.D.); (N.T.A.)
| |
Collapse
|
7
|
Liu H, Meng H, Du M, Lv H, Wang Y, Zhang K. Chlorogenic acid ameliorates intestinal inflammation by inhibiting NF-κB and endoplasmic reticulum stress in lipopolysaccharide-challenged broilers. Poult Sci 2024; 103:103586. [PMID: 38442474 PMCID: PMC11067738 DOI: 10.1016/j.psj.2024.103586] [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: 09/09/2023] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Intestinal inflammation is a primary contributor to poor growth performance during poultry production. Chlorogenic acid (CGA) is a natural phenolic acid that exhibits superior anti-inflammatory activity and improved intestinal health. To investigate the protective effects and molecular mechanisms of CGA during intestinal inflammation in lipopolysaccharide (LPS)-challenged broilers, we randomly divided 288 one-day-old male Cobb broilers into 4 groups: a control group fed a basal diet (CON group), a basal diet + LPS group (LPS group), and 2 basal diet groups fed 500 or 750 mg/kg CGA + LPS (CGA_500 or CGA_750 groups). Broilers were injected with LPS or saline at 15, 17, 19, and 21 d old. Chlorogenic acid supplementation improved the growth performance of LPS-challenged broilers by increasing average daily gain (ADG) and reducing feed/gain (F/G) ratios (P < 0.05). CGA also improved intestinal barrier function in LPS-challenged boilers by enhancing jejunum morphology and integrity, decreasing intestinal permeability, and increasing occludin 3, zonula occludens-1, and mucin 2 expression (P < 0.05). CGA supplementation also improved systemic and jejunum antioxidant capacity by significantly enhancing glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and catalase (CAT) activities (P < 0.05), and reducing malonaldehyde (MDA) and protein carbonyl (PCO) levels (P < 0.05). Chlorogenic acid supplementation reduced systemic and jejunum pro-inflammatory cytokines (interleukin (IL)-1β, IL-6, and IL-12) and increased anti-inflammatory cytokines (IL-10) in LPS-challenged broilers (P < 0.05) by inhibiting the toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway (P < 0.05). In addition, the protective effects of CGA toward intestinal inflammation and apoptosis appeared to be correlated with inhibited endoplasmic reticulum (ER) stress (P < 0.05). In summary, CGA supplementation improved intestinal morphology and integrity by inhibiting TLR4/NF-κB and ER stress pathways, which potentially reduced oxidative stress and inflammation, and ultimately improved the growth performance of LPS-challenged broilers.
Collapse
Affiliation(s)
- Huawei Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Hongling Meng
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Mengmeng Du
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Huimin Lv
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Yang Wang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Kai Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China.
| |
Collapse
|
8
|
Ali Q, Ma S, Liu B, Niu J, Liu M, Mustafa A, Li D, Wang Z, Sun H, Cui Y, Shi Y. Supplementing Ryegrass Ameliorates Commercial Diet-Induced Gut Microbial Dysbiosis-Associated Spleen Dysfunctions by Gut-Microbiota-Spleen Axis. Nutrients 2024; 16:747. [PMID: 38474875 DOI: 10.3390/nu16050747] [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: 01/11/2024] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
The type and composition of food strongly affect the variation and enrichment of the gut microbiota. The gut-microbiota-spleen axis has been developed, incorporating the spleen's function and maturation. However, how short-chain fatty-acid-producing gut microbiota can be considered to recover spleen function, particularly in spleens damaged by changed gut microbiota, is unknown in geese. Therefore, the gut microbial composition of the caecal chyme of geese was assessed by 16S rRNA microbial genes, and a Tax4Fun analysis identified the enrichment of KEGG orthologues involved in lipopolysaccharide production. The concentrations of LPS, reactive oxygen species, antioxidant/oxidant enzymes, and immunoglobulins were measured from serum samples and spleen tissues using ELISA kits. Quantitative reverse transcription PCR was employed to detect the Kelch-like-ECH-associated protein 1-Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2), B cell and T cell targeting markers, and anti-inflammatory/inflammatory cytokines from the spleen tissues of geese. The SCFAs were determined from the caecal chyme of geese by using gas chromatography. In this study, ryegrass-enriched gut microbiota such as Eggerthellaceae, Oscillospiraceae, Rikenellaceae, and Lachnospiraceae attenuated commercial diet-induced gut microbial alterations and spleen dysfunctions in geese. Ryegrass significantly improved the SCFAs (acetic, butyric, propionic, isovaleric, and valeric acids), AMPK pathway-activated Nrf2 redox signaling cascades, B cells (B220, CD19, and IgD), and T cells (CD3, CD4, CD8, and IL-2, with an exception of IL-17 and TGF-β) to activate anti-inflammatory cytokines (IL-4 and IL-10) and immunoglobulins (IgA, IgG, and IgM) in geese. In conclusion, ryegrass-improved reprogramming of the gut microbiota restored the spleen functions by attenuating LPS-induced oxidative stress and systemic inflammation through the gut-microbiota-spleen axis in geese.
Collapse
Affiliation(s)
- Qasim Ali
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Sen Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Boshuai Liu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Jiakuan Niu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mengqi Liu
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Ahsan Mustafa
- Department of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China
| | - Defeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Zhichang Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Hao Sun
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Yalei Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| | - Yinghua Shi
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Herbage Engineering Technology Research Center, Zhengzhou 450001, China
| |
Collapse
|
9
|
Sousa A, Azevedo R, Costa VM, Oliveira S, Preguiça I, Viana S, Reis F, Almeida A, Matafome P, Dias-Pereira P, Carvalho F, Fernandes E, Freitas M. Biodistribution and intestinal inflammatory response following voluntary oral intake of silver nanoparticles by C57BL/6J mice. Arch Toxicol 2023; 97:2643-2657. [PMID: 37594589 PMCID: PMC10474984 DOI: 10.1007/s00204-023-03558-5] [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: 06/23/2023] [Accepted: 07/12/2023] [Indexed: 08/19/2023]
Abstract
Silver nanoparticles (AgNP) are among the most widely commercialized nanomaterials globally, with applications in medicine and the food industry. Consequently, the increased use of AgNP in the food industry has led to an unavoidable rise in human exposure to these nanoparticles. Their widespread use raises concerns about potential hazards to human health, specifically their intestinal pro-inflammatory effects. Thus, the main objective of this study was to evaluate the biological effects of two subacute doses of 5 nm polyvinylpyrrolidone (PVP)-AgNP in C57BL/6J mice. One mg/kg body weight or 10 mg/kg bw was provided once a day for 14 days, using a new technology (HaPILLness) that allows voluntary, stress-free, and accurate oral dosing. It was observed that after oral ingestion, while AgNP is biodistributed throughout the entire organism, most of the ingested dose is excreted in the feces. The passage and accumulation of AgNP throughout the intestine instigated a prominent inflammatory response, marked by significant histological, vascular, and cellular transformations. This response was driven by the activation of the nuclear factor-кB (NF-кB) inflammatory pathway, ultimately leading to the generation of multiple cytokines and chemokines.
Collapse
Affiliation(s)
- Adelaide Sousa
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira N.º 228, 4050-313, Porto, Portugal
| | - Rui Azevedo
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira N.º 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‑313, Porto, Portugal
- Associated Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Sara Oliveira
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Inês Preguiça
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Sofia Viana
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Flávio Reis
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
| | - Agostinho Almeida
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira N.º 228, 4050-313, Porto, Portugal
| | - Paulo Matafome
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Patrícia Dias-Pereira
- ICBAS School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‑313, Porto, Portugal
- Associated Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira N.º 228, 4050-313, Porto, Portugal.
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira N.º 228, 4050-313, Porto, Portugal.
| |
Collapse
|
10
|
Xin L, Li F, Yu H, Xiong Q, Hou Q, Meng Y. Honokiol alleviates radiation-induced premature ovarian failure via enhancing Nrf2. Am J Reprod Immunol 2023; 90:e13769. [PMID: 37766410 DOI: 10.1111/aji.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND The ovary is highly sensitive to radiation, and patients receiving radiotherapy are at significant risk of premature ovarian failure (POF). This study aimed to explore the radioprotective effect of honokiol (HKL) on ionizing radiation (IR)-induced POF. METHODS Female C57BL/6 mice were administered intraperitoneally with vehicle or HKL once daily for 7 days. On day 7, the mice in the IR and HKL+IR groups were exposed to 3.2 Gy whole-body radiation for one hour after the intraperitoneal injection and sacrificed 12 or 72 h after radiation exposure. The gonadosomatic index (GSI) was calculated. Blood samples were collected for enzyme-linked immunosorbent assay (ELISA). Ovaries were harvested for histological examination, immunohistochemistry, immunofluorescence, TUNEL, western blot, and qPCR. The fertility assessment was evaluated by calculating live offspring number. RESULTS The optimum dose of HKL against radiation was 10 mg/kg via intraperitoneal injection. POF was induced 72 h after irradiation with significantly downregulated proliferating cell nuclear antigen (PCNA). The numbers of primordial and preantral follicles decreased significantly after irradiation (p < .001), whereas the number of atretic follicles increased (p < .001). The serum levels of estradiol (E2 ) and anti-Müllerian hormone (AMH) decreased to 50% of the control group after irradiation (p < .05). Moreover, the GSI after irradiation was 27% lower than in the control group (p < .05). The number of offspring in the IR group dropped by 50% compared with the control group (p < .05). HKL pretreatment protected the animals' fertility, GSI, PCNA, serum levels of E2 and AMH, and the number of primordial and preantral follicles. Significant upregulation of apoptosis-related proteins such as Pho-P53, Bax, Cyto C, C-caspase-3, C-PARP, and pyroptosis-related proteins such as Pho-NF-κB p65, NLRP3, caspase-1, IL-1β, and IL-18 was observed after irradiation, while the expression of Bcl-2 decreased. HKL pretreatment prevented these changes. After irradiation, malondialdehyde (MDA), Nrf2, and HO-1 were upregulated. HKL treatment activated the expression of Nrf2 and HO-1 and promoted the nucleus translocation of Nrf2. Furthermore, HKL did not affect ovarian reserves under physiological conditions. CONCLUSIONS HKL ameliorated IR-induced POF by inhibiting apoptosis and pyroptosis by enhancing Nrf2 expression and translocation.
Collapse
Affiliation(s)
- Lingli Xin
- Department of Graduate Administration, General Hospital of Chinese PLA, Beijing, China
- Department of Obstetrics and Gynecology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Fengsheng Li
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Huijie Yu
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qi Xiong
- Department of Oncology, General Hospital of Chinese PLA, Beijing, China
| | - Qingxiang Hou
- Department of Obstetrics and Gynecology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yuanguang Meng
- Department of Graduate Administration, General Hospital of Chinese PLA, Beijing, China
- Department of Obstetrics and Gynecology, General Hospital of Chinese PLA, Beijing, China
| |
Collapse
|
11
|
Mweetwa MN, Haritunians T, Dube S, Chandwe K, Amadi B, Zyambo K, Liu TC, McGovern D, Kelly P. Genetic variation in environmental enteropathy and stunting in Zambian children: A pilot genome wide association study using the H3Africa chip. PLoS One 2023; 18:e0291311. [PMID: 37756315 PMCID: PMC10529557 DOI: 10.1371/journal.pone.0291311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
PURPOSE Stunting is known to be heavily influenced by environmental factors, so the genetic contribution has received little attention. Here we report an exploration of genetic influences in stunted Zambian children with environmental enteropathy. METHOD Children with stunting (LAZ < -2) were enrolled and given nutritional therapy. Those that were non-responsive to therapy were designated as cases, and children with good growth (LAZ > -1) from the same community as controls. Blood and stool samples were taken to measure biomarkers of intestinal inflammation, epithelial damage, and microbial translocation. Single nucleotide polymorphism array genotyping was carried out on saliva samples using the H3Africa consortium array. RESULTS Genome wide associations were analysed in 117 cases and 41 controls. While no significant associations with stunting were observed at P<5x10-8, likely due to the small sample size, interesting associations were observed at lower thresholds. SNPs associated with stunting were in genomic regions known to modulate neuronal differentiation and fatty acid biosynthesis. SNPs associated with increased microbial translocation were associated with non-integrin membrane ECM interactions, tight junctions, hemostasis, and G-alpha signalling events. SNPs associated with increased inflammation were associated with, ECM interactions, purine metabolism, axon guidance, and cell motility. SNPs negatively associated with inflammation overlapped genes involved in semaphoring interactions. We explored the existing coeliac disease risk HLA genotypes and found present: DQ2.5 (7.5%), DQ8 (3.5%) and DQ2.2 (3.8%); however, no children were positive for coeliac antibodies. We detected HLA-DRB:1301 and HLA-C:1802 with high odds ratios and P<0.05 in stunted children compared to controls. CONCLUSION Genetic variations associated with stunting and the enteropathy underlying it, include variants associated with multiple pathways relating to gene expression, glycosylation, nerve signalling, and sensing of the nutritional and microbiological milieu.
Collapse
Affiliation(s)
- Monica N. Mweetwa
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Physiology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Talin Haritunians
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Shishir Dube
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Kanta Chandwe
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Paediatrics, University of Zambia School of Medicine, Lusaka, Zambia
| | - Beatrice Amadi
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Department of Paediatrics, University of Zambia School of Medicine, Lusaka, Zambia
| | - Kanekwa Zyambo
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
| | - Ta-Chiang Liu
- Washington University in St. Louis (WUSTL), St. Louis, Missouri, United States of America
| | - Dermot McGovern
- F. Widjaja Foundation Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Paul Kelly
- Tropical Gastroenterology & Nutrition Group, University of Zambia School of Medicine, Lusaka, Zambia
- Washington University in St. Louis (WUSTL), St. Louis, Missouri, United States of America
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Jiang K, Wang D, Su L, Liu X, Yue Q, Zhang S, Zhao L. Tamarind Seed Polysaccharide Hydrolysate Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis via Regulating the Gut Microbiota. Pharmaceuticals (Basel) 2023; 16:1133. [PMID: 37631047 PMCID: PMC10459238 DOI: 10.3390/ph16081133] [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: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Ulcerative colitis (UC) is a disease caused by noninfectious chronic inflammation characterized by varying degrees of inflammation affecting the colon or its entire mucosal surface. Current therapeutic strategies rely on the suppression of the immune response, which is effective, but can have detrimental effects. Recently, different plant polysaccharides and their degradation products have received increasing attention due to their prominent biological activities. The aim of this research was to evaluate the mitigation of inflammation exhibited by tamarind seed polysaccharide hydrolysate (TSPH) ingestion in colitis mice. (2) Methods: TSPH was obtained from the hydrolysis of tamarind seed polysaccharide (TSP) by trifluoroacetic acid (TFA). The structure and physical properties of TSPH were characterized by ultraviolet spectroscopy (UV), thin-layer chromatography (TLC), fourier transform infrared spectroscopy (FT-IR), and High-Performance Liquid Chromatography and Electrospray Ionization Mass Spectrometry (HPLC-ESI/MS) analysis. Then, the alleviative effects of the action of TSPH on 2.5% dextran sodium sulfate (DSS)-induced colitis mice were investigated. (3) Results: TSPH restored pathological lesions in the colon and inhibited the over-secretion of pro-inflammatory cytokines in UC mice. The relative expression level of mRNA for colonic tight junction proteins was increased. These findings suggested that TSPH could reduce inflammation in the colon. Additionally, the structure of the gut microbiota was also altered, with beneficial bacteria, including Prevotella and Blautia, significantly enriched by TSPH. Moreover, the richness of Blautia was positively correlated with acetic acid. (4) Conclusions: In conclusion, TSPH suppressed colonic inflammation, alleviated imbalances in the intestinal flora and regulated bacterial metabolites. Thus, this also implies that TSPH has the potential to be a functional food against colitis.
Collapse
Affiliation(s)
- Kangjia Jiang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Duo Wang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Xinli Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Qiulin Yue
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Song Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; (K.J.); (D.W.); (L.S.); (X.L.); (Q.Y.)
- Shandong Chenzhang Biotechnology Co., Ltd., Jinan 250353, China
| |
Collapse
|
13
|
Arvanitakis K, Koufakis T, Popovic D, Maltese G, Mustafa O, Doumas M, Giouleme O, Kotsa K, Germanidis G. GLP-1 Receptor Agonists in Obese Patients with Inflammatory Bowel Disease: from Molecular Mechanisms to Clinical Considerations and Practical Recommendations for Safe and Effective Use. Curr Obes Rep 2023; 12:61-74. [PMID: 37081371 DOI: 10.1007/s13679-023-00506-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/09/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE OF REVIEW To discuss current literature and provide practical recommendations for the safe and effective use of glucagon-like peptide 1 receptor agonists (GLP-1 RA) in people with inflammatory bowel disease (IBD) and type 2 diabetes (T2D) and/or obesity. The molecular mechanisms that justify the potential benefits of GLP-1 RA in IBD and the links between IBD, obesity, and cardiovascular disease are also discussed. RECENT FINDINGS Preliminary data suggest that GLP-1 RA can modulate crucial pathways in the pathogenesis of IBD, such as chronic inflammation circuits, intestinal tight junctions, and gut microbiome dysbiosis, setting the stage for human trials to investigate the role of these agents in the treatment of IBD among people with or without diabetes and obesity. However, gastrointestinal side effects related to GLP-1 RA need appropriate clinical management to mitigate risks and maximize the benefits of therapy in people with IBD. GLP-1 RA originally emerged as drugs for the treatment of hyperglycemia and are currently licensed for the management of T2D and/or overweight/obesity. However, their wealth of pleiotropic actions soon raised expectations that they might confer benefits on non-metabolic disorders. Future studies are expected to clarify whether GLP-1 RA deserve an adjunct place in the arsenal of drugs against IBD.
Collapse
Affiliation(s)
- Konstantinos Arvanitakis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636, Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Djordje Popovic
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Centre of Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| | - Giuseppe Maltese
- Department of Diabetes and Endocrinology, Epsom & St Helier University Hospitals, Surrey, SM5 1AA, UK
- Unit for Metabolic Medicine, Cardiovascular Division, Faculty of Life Sciences & Medicine, King's College, London, UK
| | - Omar Mustafa
- Department of Diabetes, King's College Hospital NHS Foundation Trust, Denmark Hill, London, UK
- King's College London, London, UK
| | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Olga Giouleme
- Second Propedeutic Department of Internal Medicine, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism and Diabetes Centre, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Georgios Germanidis
- Division of Gastroenterology and Hepatology, First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece.
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54636, Thessaloniki, Greece.
| |
Collapse
|
14
|
Barekatain R, Chrystal PV, Nowland T, Moss AF, Howarth GS, Hao Van TT, Moore RJ. Negative consequences of reduced protein diets supplemented with synthetic amino acids for performance, intestinal barrier function, and caecal microbiota composition of broiler chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 13:216-228. [PMID: 37388459 PMCID: PMC10300400 DOI: 10.1016/j.aninu.2023.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/22/2023]
Abstract
The consequences of feeding broiler chickens with reduced protein (RP) diets for gut health and barrier function are not well understood. This study was performed to elucidate the effect of reducing dietary protein and source of protein on gut health and performance parameters. Four experimental diets included 2 control diets with standard protein levels either containing meat and bone meal (CMBM) or an all-vegetable diet (CVEG), a medium RP diet (17.5% in growers and 16.5% in finisher), and a severe RP diet (15.6% in grower and 14.6% in finisher). Off-sex Ross 308 birds were assigned to each of the 4 diets and performance measurements were taken from d 7 to 42 post-hatch. Each diet was replicated 8 times (10 birds per replicate). A challenge study was conducted on additional 96 broilers (24 birds per diet) from d 13 to 21. Half of the birds in each dietary treatment were challenged by dexamethasone (DEX) to induce a leaky gut. Feeding birds with RP diets decreased weight gain (P < 0.0001) and increased feed conversion ratio (P < 0.0001) from d 7 to 42 compared with control diets. There was no difference between CVEG and CMBM control diets for any parameter. The diet containing 15.6% protein increased (P < 0.05) intestinal permeability independent of the DEX challenge. Gene expression of claudin-3 was downregulated (P < 0.05) in birds fed 15.6% protein. There was a significant interaction between diet and DEX (P < 0.05) and both RP diets (17.5% and 15.6%) downregulated claudin-2 expression in DEX-challenged birds. The overall composition of the caecal microbiota was affected in birds fed 15.6% protein having a significantly lower richness of microbiota in both sham and DEX-injected birds. Proteobacteria was the main phylum driving the differences in birds fed 15.6% protein. At the family level, Bifidobacteriaceae, Unclassified Bifidobacteriales, Enterococcaceae, Enterobacteriaceae, and Lachnospiraceae were the main taxa in birds fed 15.6% protein. Despite supplementation of synthetic amino acids, severe reduction of dietary protein compromised performance and intestinal health parameters in broilers, evidenced by differential mRNA expression of tight junction proteins, higher permeability, and changes in caecal microbiota composition.
Collapse
Affiliation(s)
- Reza Barekatain
- South Australian Research and Development Institute, Roseworthy Campus, Roseworthy, SA, Australia
- School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Roseworthy, SA, Australia
| | - Peter V. Chrystal
- Complete Feed Solutions, Hornsby, NSW, Australia; Howick, New Zealand
| | - Tanya Nowland
- South Australian Research and Development Institute, Roseworthy Campus, Roseworthy, SA, Australia
- School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Roseworthy, SA, Australia
| | - Amy F. Moss
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| | - Gordon S. Howarth
- School of Animal and Veterinary Sciences, Roseworthy Campus, University of Adelaide, Roseworthy, SA, Australia
| | - Thi Thu Hao Van
- School of Science, RMIT University, Bundoora West Campus, Bundoora, VIC, Australia
| | - Robert J. Moore
- School of Science, RMIT University, Bundoora West Campus, Bundoora, VIC, Australia
| |
Collapse
|
15
|
Sun J, Jiao Z, Zhu W, Li X, Wang P, Wang J, Tai T, Wang Y, Wang H, Shi G. Astilbin Attenuates Cadmium-Induced Adipose Tissue Damage by Inhibiting NF-κB Pathways and Regulating the Expression of HSPs in Chicken. Biol Trace Elem Res 2023; 201:2512-2523. [PMID: 35717552 DOI: 10.1007/s12011-022-03327-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022]
Abstract
Cadmium (Cd) can damage tissues by inducing oxidative stress, lymphocyte infiltration, and inflammation in these sites. Meanwhile, astilbin (Ast) is an antioxidant agent. At present, only a few mechanisms of Cd-induced adipose tissue damage have been described. Herein, we assessed the potential protective effects and the molecular mechanism underlying the antioxidant properly of Ast after Cd intake in chicken adipose tissue. In this study, a total of 160 7-day-old roosters were randomly divided into four groups. Roosters were fed with a basic diet (C group), Ast 40 mg/kg (Ast group), CdCl2 150 mg/kg + Ast 40 mg/kg (Cd/Ast group), and CdCl2 150 mg/kg (Cd group) for 60 days. We found that Cd intake changed the morphology and structure of adipose tissues and decreased the expression of several antioxidants, including total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px), catalase (CAT), and total antioxidant capacity (T-AOC), but increased those of oxidative stress markers including malondialdehyde (MDA), inducible nitric oxide synthase (iNOS), NO, and H2O2. Cd further activated the nuclear factor kappa B (NF-κB) signaling pathway and increased the expression of the inflammation-related mediators, interleukin 1beta (IL-1β), interleukin 6 (IL-6), interleukin 8 (IL-8), interleukin 10 (IL-10), cyclooxygenase-2 (COX-2), iNOS, prostaglandin E synthase (PTGES), tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ). Cd-induced oxidative stress upregulated the expression of three heat shock proteins (HSPs), including HSP27, HSP70, and HSP90. Summarily, Cd causes oxidative stress-mediated tissue damage by activating the NF-κB pathway, promoting inflammation and upregulating the expression of HSPs. However, Ast supplementation modulates oxidative stress in adipose tissue by inhibiting inflammation mediated by the NF-κB pathway and regulating the expression of HSPs.
Collapse
Affiliation(s)
- Jianxu Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Zitao Jiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Weifeng Zhu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Xiuyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Panpan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Jiangfeng Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Tiange Tai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Yuxi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Haibin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China
| | - Guangliang Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Harbin, China.
| |
Collapse
|
16
|
Domene A, Orozco H, Rodríguez-Viso P, Monedero V, Zúñiga M, Vélez D, Devesa V. Intestinal homeostasis disruption in mice chronically exposed to arsenite-contaminated drinking water. Chem Biol Interact 2023; 373:110404. [PMID: 36791901 DOI: 10.1016/j.cbi.2023.110404] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/29/2023] [Accepted: 02/11/2023] [Indexed: 02/14/2023]
Abstract
Chronic exposure to inorganic arsenic [As(III) and As(V)] affects about 200 million people, and is linked to a greater incidence of certain types of cancer. Drinking water is the main route of exposure, so, in endemic areas, the intestinal mucosa is constantly exposed to the metalloid. However, studies on the intestinal toxicity of inorganic As are scarce. The objective of this study was to evaluate the toxicity of a chronic exposure to As(III) on the intestinal mucosa and its associated microbiota. For this purpose, BALB/c mice were exposed during 6 months through drinking water to As(III) (15 and 30 mg/L). Treatment with As(III) increased reactive oxygen species (43-64%) and lipid peroxidation (8-51%). A pro-inflammatory response was also observed, evidenced by an increase in fecal lactoferrin (23-29%) and mucosal neutrophil infiltration. As(III) also induced an increase in the colonic levels of pro-inflammatory cytokines (24-201%) and the activation of some pro-inflammatory signaling pathways. Reductions in the number of goblet cells and mucus production were also observed. Moreover, As(III) exposure resulted in changes in gut microbial alpha diversity but no differences in beta diversity. This suggested that the abundance of some taxa was significantly affected by As(III), although the composition of the population did not show significant alterations. Analysis of differential taxa agreed with this, 21 ASVs were affected in abundance or variability, especially ASVs from the family Muribaculaceae. Intestinal microbiota metabolism was also affected, as reductions in fecal concentration of short-chain fatty acids were observed. The effects observed on different components of the intestinal barrier may be responsible of the increased permeability in As(III) treated mice, evidenced by an increase in fecal albumin (48-66%). Moreover, serum levels of Lipopolysaccharide binding proteins and TNF-α were increased in animals treated with 30 mg/L of As(III), suggesting a low-level systemic inflammation.
Collapse
Affiliation(s)
- A Domene
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - H Orozco
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - P Rodríguez-Viso
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - V Monedero
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - M Zúñiga
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - D Vélez
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain
| | - V Devesa
- Instituto de Agroquímica y Tecnología de Alimentos, Calle Agustín Escardino 7, 46980, Paterna, Spain.
| |
Collapse
|
17
|
Hou W, Yang S, Lu J, Shi Y, Chen J, Chen D, Wang F, Liu L. Hypothermic machine perfusion alleviates ischemia-reperfusion injury of intestinal transplantation in pigs. Front Immunol 2023; 14:1117292. [PMID: 36926337 PMCID: PMC10011072 DOI: 10.3389/fimmu.2023.1117292] [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: 12/06/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Background Intestinal transplantation (IT) has become an important procedure for the treatment of irreversible intestinal failure. However, IT is extremely vulnerable to ischemia-reperfusion injury (IRI). Due to the limitations of static cold storage (SCS), hypothermic machine perfusion (HMP) is rapidly gaining popularity. In this study, the intestinal HMP system is established and HMP is compared with SCS. Methods An intestinal HMP system was built. Ten miniature pigs were randomly divided into the HMP and SCS groups, and their intestines were perfused using the HMP device and SCS, respectively, followed by orthotopic auto-transplantation. Analysis was done on the grafts between the two groups. Results Operation success rates of the surgery were 100% in both groups. The 7-day survival rate was 100% in the HMP group, which was significantly higher than that of the SCS group (20%, P< 0.05). The pathological results showed that fewer injuries of grafts were in the HMP group. Endotoxin (ET), IL-1, IL-6, IFN-γ and TNF-α levels in the HMP group were significantly lower than in the SCS group (P<0.05), whereas IL-10 levels were significantly higher (P<0.05).The intestinal expression levels of ZO-1 and Occludin were higher in the HMP group compared to the SCS group, whereas Toll-like receptor 4 (TLR4), nuclear factor kappa B (NFκB), and caspase-3 were lower. Conclusions In this study, we established a stable intestinal HMP system and demonstrated that HMP could significantly alleviate intestinal IRI and improve the outcome after IT.
Collapse
Affiliation(s)
- Wen Hou
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Shuang Yang
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Jiansen Lu
- First Central Clinical Institute, Tianjin Medical University, Tianjin, China
| | - Yuan Shi
- Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, Tianjin, China
| | - Jing Chen
- Organ Transplant Department, Tianjin First Central Hospital, Tianjin, China
| | - Decheng Chen
- First Central Clinical Institute, Tianjin Medical University, Tianjin, China
| | - Fei Wang
- School of Medicine, Nankai University, Tianjin, China
| | - Lei Liu
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China.,Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, Tianjin, China.,Organ Transplant Department, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
18
|
Yao W, Wang T, Huang L, Bao Z, Wen S, Huang F. Embelin alleviates weaned piglets intestinal inflammation and barrier dysfunction via PCAF/NF-κB signaling pathway in intestinal epithelial cells. J Anim Sci Biotechnol 2022; 13:139. [PMID: 36514139 PMCID: PMC9749222 DOI: 10.1186/s40104-022-00787-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Intestinal barrier plays key roles in maintaining intestinal homeostasis. Inflammation damage can severely destroy the intestinal integrity of mammals. This study was conducted to investigate the protective effects of embelin and its molecular mechanisms on intestinal inflammation in a porcine model. One hundred sixty 21-day-old castrated weaned pigs (Duroc × Landrace × Yorkshire, average initial body weight was 7.05 ± 0.28 kg, equal numbers of castrated males and females) were allotted to four groups and fed with a basal diet or a basal diet containing 200, 400, or 600 mg embelin/kg for 28 d. The growth performance, intestinal inflammatory cytokines, morphology of jejunum and ileum, tight junctions in the intestinal mucosa of piglets were tested. IPEC-1 cells with overexpression of P300/CBP associating factor (PCAF) were treated with embelin, the activity of PCAF and acetylation of nuclear factor-κB (NF-κB) were analyzed to determine the effect of embelin on PCAF/NF-κB pathway in vitro. RESULTS The results showed that embelin decreased (P < 0.05) serum D-lactate and diamine oxidase (DAO) levels, and enhanced the expression of ZO-1, occludin and claudin-1 protein in jejunum and ileum. Moreover, the expression levels of critical inflammation molecules (interleukin-1β, interleukin-6, tumor necrosis factor-α, and NF-κB) were down-regulated (P < 0.05) by embelin in jejunal and ileal mucosa. Meanwhile, the activity of PCAF were down-regulated (P < 0.05) by embelin. Importantly, transfection of PCAF siRNAs to IPEC-1 cell decreased NF-κB activities; embelin treatment downregulated (P < 0.05) the acetylation and activities of NF-κB by 31.7%-74.6% in IPEC-1 cells with overexpression of PCAF. CONCLUSIONS These results suggested that embelin ameliorates intestinal inflammation in weaned pigs, which might be mediated by suppressing the PCAF/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Tongxin Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Lu Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Zhengxi Bao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Shu Wen
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070 China
| |
Collapse
|
19
|
de Arruda Roque F, Chen J, Araujo RB, Murcio AL, de Souza Leite BG, Dias Tanaka MT, Granghelli CA, Pelissari PH, Bueno Carvalho RS, Torres D, Vázquez‐Añón M, Hancock D, Soares da Silva Araujo C, Araujo LF. Maternal supplementation of different trace mineral sources on broiler breeder production and progeny growth and gut health. Front Physiol 2022; 13:948378. [PMID: 36267581 PMCID: PMC9577897 DOI: 10.3389/fphys.2022.948378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Trace mineral minerals Zn, Cu, and Mn play important roles in breeder production and progeny performance. The objective of this study was to determine maternal supplementation of trace mineral minerals on breeder production and progeny growth and development. A total of 540 broiler breeders, Cobb 500 (Slow feathering; 0-66 weeks old) were assigned to one of three treatment groups with the same basal diet and three different supplemental trace minerals: ITM-inorganic trace minerals in sulfates: 100, 16, and 100 ppm of Zn, Cu, and Mn respectively; MMHAC -mineral methionine hydroxy analog chelate: 50, 8, and 50 ppm of bis-chelated MINTREX®Zn, Cu and Mn (Novus International, Inc.), and TMAAC - trace minerals amino acid complex: 50, 8, and 50 ppm of Zn, Cu, and Mn. At 28 weeks of age, eggs from breeder treatments were hatched for progeny trial, 10 pens with 6 males and 6 female birds per pen were fed a common diet with ITM for 45 days. Breeder production, egg quality, progeny growth performance, mRNA expression of gut health associated genes in breeder and progeny chicks were measured. Data were analyzed by one-way ANOVA; means were separated by Fisher's protected LSD test. A p-Value ≤ 0.05 was considered statistically different and 0.1 was considered numerical trend. Breeders on ITM treatment had higher (p < 0.05) body weight (BW), weight gain and lower (p < 0.05) feed conversion ratio (FCR) from 0 to 10 weeks, when compared to birds fed MMHAC. MMHAC significantly improved egg mass by 3 g (p < 0.05) and FCR by 34 points (0.05 < p < 0.1) throughout the reproductive period (26-66 weeks) in comparison to ITM. MMHAC improved (p < 0.01) egg yolk color versus (vs.) ITM and TMAAC in all periods, except 28 weeks, increased (p < 0.01) eggshell thickness and resistance vs. TMAAC at 58 weeks, and reduced (p < 0.05) jejunal NF-κB gene expression vs. TMAAC at 24 weeks. There was a significant reduction in tibial dry matter weight, Seedor index and resistance for the breeders that received MMHAC and/or TMAAC when compared to ITM at 18 weeks. Lower seedor index but numerically wider tibial circumference was seen in hens fed MMHAC at 24 weeks, and wider tibial circumference but lower tibial resistance in hens fed TMAAC at 66 weeks. Maternal supplementation of MMHAC in breeder hens increased (p < 0.0001) BW vs. ITM and TMAAC at hatching, reduced (p < 0.05) feed intake vs. ITM at d14 and d28, and improved (p < 0.01) FCR and performance index vs. TMAAC at d28, reduced (p < 0.01) NF-κB gene expression and increased (p < 0.05) A20 gene expression vs. TMAAC on d0 and vs. ITM on d14, reduced (p < 0.05) TLR2 gene expression vs. ITM on d0 and vs. TMAAC on d14, increased (p < 0.05) MUC2 gene expression vs. both ITM and TMAAC on d45 in progeny jejunum. Overall, these results suggest that supplementation with lower levels of MHA-chelated trace minerals improved breeder production and egg quality and reduced breeder jejunal inflammation while maintaining tibial development in comparison to those receiving higher inorganic mineral supplementation, and it also carried over the benefits to progeny with better growth performance, less jejunal inflammation and better innate immune response and gut barrier function in comparison to ITM and/or TMAAC.
Collapse
Affiliation(s)
| | - Juxing Chen
- Novus International Inc., St. Charles, MO, United States
| | | | | | | | | | | | | | | | - David Torres
- Novus International Inc., St. Charles, MO, United States
| | | | - Deana Hancock
- Novus International Inc., St. Charles, MO, United States
| | | | | |
Collapse
|
20
|
The role of guggulsterone on the NF-κB pathway in inflammatory bowel disease: preclinical evidence. Future Sci OA 2022; 8:FSO803. [PMID: 35909996 PMCID: PMC9327637 DOI: 10.2144/fsoa-2022-0020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
Guggulsterone, a phytosterol in the herb Commiphora wightii (Arn.) Bhandari, has been used in the treatment of diseases such as hyperlipidemia, arthritis and atherosclerosis. It has attracted significant research interest because of its pharmacological properties, especially its anti-inflammatory nature. It can be used for the treatment of inflammatory bowel disease (IBD), a disease characterized by chronic inflammation and damage in the gastrointestinal tract. In various preclinical studies, it inhibited NF-κB, an important pathway in the pathophysiology of IBD. This review summarizes the preclinical studies on this topic for providing more insights to the mechanism by which guggulsterone exerts its effect.
Collapse
|
21
|
Gori Savellini G, Anichini G, Gandolfo C, Cusi MG. Nucleopore Traffic Is Hindered by SARS-CoV-2 ORF6 Protein to Efficiently Suppress IFN-β and IL-6 Secretion. Viruses 2022; 14:1273. [PMID: 35746745 PMCID: PMC9230033 DOI: 10.3390/v14061273] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/19/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
A weak production of INF-β along with an exacerbated release of pro-inflammatory cytokines have been reported during infection by the novel SARS-CoV-2 virus. SARS-CoV-2 encodes several proteins that are able to counteract the host immune system, which is believed to be one of the most important features contributing to the viral pathogenesis and development of a severe clinical outcomes. Previous reports demonstrated that the SARS-CoV-2 ORF6 protein strongly suppresses INF-β production by hindering the RIG-I, MDA-5, and MAVS signaling cascade. In the present study, we better characterized the mechanism by which the SARS-CoV-2 ORF6 counteracts IFN-β and interleukin-6 (IL-6), which plays a crucial role in the inflammation process associated with the viral infection. In the present study, we demonstrated that the SARS-CoV-2 ORF6 protein has evolved an alternative mechanism to guarantee host IFN-β and IL-6 suppression, in addition to the transcriptional control exerted on the genes. Indeed, a block in movement through the nucleopore of newly synthetized messenger RNA encoding the immune-modulatory cytokines IFN-β and IL-6 are reported here. The ORF6 accessory protein of SARS-CoV-2 displays a multifunctional activity and may represent one of the most important virulence factors. Where conventional antagonistic strategies of immune evasion-such as the suppression of specific transcription factors (e.g., IRF-3, STAT-1/2)-would not be sufficient, the SARS-CoV-2 ORF6 protein is the trump card for the virus, also blocking the movement of IFN-β and IL-6 mRNAs from nucleus to cytoplasm. Conversely, we showed that nuclear translocation of the NF-κB transcription factor is not affected by the ORF6 protein, although inhibition of its cytoplasmic activation occurred. Therefore, the ORF6 protein exerts a 360-degree inhibition of the antiviral response by blocking as many critical points as possible.
Collapse
Affiliation(s)
- Gianni Gori Savellini
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.A.); (C.G.); (M.G.C.)
| | | | | | | |
Collapse
|
22
|
Merten D, Erman L, Marabelli GP, Leners B, Ney Y, Nasim MJ, Jacob C, Tchoumtchoua J, Cajot S, Bohn T. Potential health effects of brewers' spent grain as a functional food ingredient assessed by markers of oxidative stress and inflammation following gastro-intestinal digestion and in a cell model of the small intestine. Food Funct 2022; 13:5327-5342. [PMID: 35446320 DOI: 10.1039/d1fo03090f] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Brewer's spent grains (BSG) are a by-product of the beer-brewing industry, often employed as animal feeding stuffs. With BSG being rich not only in proteins, lipids, and dietary fiber but also in certain phytochemicals, it constitutes a potentially valuable food source that could be employed as a functional food, e.g. against chronic inflammatory diseases. Several types of bread were prepared with various amounts of BSG as flour replacement (0, 10, 20, 40, 60, 80 and 100%), either employing wet BSG or dried BSG after pressing. Total phenolics, flavonoids, insoluble dietary fiber, as well as antioxidant capacity (FRAP, ABTS) were measured in the bread, before and after simulated gastro-intestinal digestion. Furthermore, we investigated digested BSG and bread-containing BSG for their capability to alter oxidative stress (Nrf2, malondialdehyde) and inflammation (IL-6, IL-8, NO, and PGE2) in a Caco-2 cell culture model of the small intestine. Incorporation of BSG significantly and dose-dependently enhanced the amount of dietary fiber in the product, as well as total phenolics, flavonoids, and antioxidant capacity, by over 10-fold, 3-fold, 4-fold and 5-fold, respectively, when replacing all of the flour with BSG. This pattern remained after in vitro digestion. However, digesta failed to show significant antioxidant or anti-inflammatory effects on the biomarkers observed in the cell model. Consuming 150 g of such a BSG-bread (wet based) would supply the proposed RDA of 25 g d-1 dietary fiber and could be a healthy product valorizing BSG.
Collapse
Affiliation(s)
- Diane Merten
- Luxembourg Institute of Health, Department of Precision Health, Nutrition and Health Research Group, 1 A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg.
| | - Lara Erman
- Luxembourg Institute of Health, Department of Precision Health, Nutrition and Health Research Group, 1 A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg.
| | | | - Bernadette Leners
- Luxembourg Institute of Health, Department of Precision Health, Nutrition and Health Research Group, 1 A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg.
| | - Yannick Ney
- Division of Bioorganic Chemistry, School of Pharmacy, University of Saarland, 66123 Saarbruecken, Germany
| | - Muhammad Jawad Nasim
- Division of Bioorganic Chemistry, School of Pharmacy, University of Saarland, 66123 Saarbruecken, Germany
| | - Claus Jacob
- Division of Bioorganic Chemistry, School of Pharmacy, University of Saarland, 66123 Saarbruecken, Germany
| | - Job Tchoumtchoua
- CELABOR, Biomass Valorisation Platform - Extraction Department, Avenue du Parc 38, 4650 Herve, Belgium
| | - Sébastien Cajot
- CELABOR, Biomass Valorisation Platform - Extraction Department, Avenue du Parc 38, 4650 Herve, Belgium
| | - Torsten Bohn
- Luxembourg Institute of Health, Department of Precision Health, Nutrition and Health Research Group, 1 A-B, rue Thomas Edison, L-1445 Strassen, Luxembourg.
| |
Collapse
|
23
|
Hassanin TM, Fouad Y, Mohamed FE, Abdel-Hafeez EH, Hassnine A. Colonic mucosal eosinophilia and immunohistochemical expression of COX-2 and NF-kB in patients with irritable bowel syndrome. Eur J Gastroenterol Hepatol 2022; 34:512-517. [PMID: 35275879 DOI: 10.1097/meg.0000000000002363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND There is growing evidence that eosinophilic infiltration can release mediators which are harmful to the intestinal epithelium in patients with irritable bowel syndrome (IBS). Although cyclooxygenase 2 (COX-2) and nuclear factor-kappa beta (NF-kB) expression had been previously reported to increase in many inflammatory conditions, there is a paucity in data investigating their expressions in IBS. Our aim was to evaluate colonic mucosal eosinophilia and immunohistochemical expression of COX-2 and NF-kB in patients with irritable bowel syndrome. METHODS A total of 80 patients who met the inclusion criteria of IBS based on Rome IV symptoms questionnaire were subjected to abdominal ultrasound, laboratory investigations, serum immunoglobulin E (IgE) level assessment and colonoscopic examination. Immunohistochemistry was performed to detect COX-2 and NF-kB expression in colonic biopsies obtained from IBS patients. RESULTS Histopathological examination showed that 60 colonic biopsy specimens (75%) showed few mixed inflammatory cells ≤3 cells/ HPF, 12 biopsy specimens (15%) showed eosinophilic infiltration ≥25 eosinophils/HPF and 8 biopsy specimens (10%) showed severe lymphocytic infiltration and aggregation. Colonic eosinophilic infiltrate was significantly higher among patients presented with IBS-D subtype. Serum IgE was significantly higher among patients with colonic eosinophilic infiltrate than the others. In IBS-D patients, colonic mucosa showed positive expression of COX-2 and NF-kB in 52.1% and 81.25% of cases, respectively. CONCLUSION Patients with IBS -particularly IBS-D subtype- should undergo colonoscopy and biopsy to exclude underlying inflammatory pathology. Moreover, patients with positive COX-2 and NF-kB need further evaluation and follow-up.
Collapse
|
24
|
Phillippi DT, Daniel S, Nguyen KN, Penaredondo BA, Lund AK. Probiotics Function as Immunomodulators in the Intestine in C57Bl/6 Male Mice Exposed to Inhaled Diesel Exhaust Particles on a High-Fat Diet. Cells 2022; 11:cells11091445. [PMID: 35563751 PMCID: PMC9101602 DOI: 10.3390/cells11091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.
Collapse
Affiliation(s)
| | | | | | | | - Amie K. Lund
- Correspondence: ; Tel.: +1-(940)-369-8946; Fax: +1-(940)-565-4297
| |
Collapse
|
25
|
Zhang YY, Yao YD, Cheng QQ, Huang YF, Zhou H. Establishment of a High Content Image Platform to Measure NF-κB Nuclear Translocation in LPS-Induced RAW264.7 Macrophages for Screening Anti-inflammatory Drug Candidates. Curr Drug Metab 2022; 23:394-414. [PMID: 35410593 DOI: 10.2174/1389200223666220411121614] [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: 06/17/2021] [Revised: 01/19/2022] [Accepted: 01/29/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND High content image (HCI), an automatic imaging and analysis system, provides a fast drug screening method by detecting the subcellular distribution of protein in intact cells. OBJECTIVE This study established the first standardized HCI platform for lipopolysaccharide (LPS)-induced RAW264.7 macrophages to screen anti-inflammatory compounds by measuring nuclear factor-κB (NF-κB) nuclear translocation. METHOD The influence of the cell passages, cell density, LPS induction time and concentration, antibody dilution, serum, dimethyl sulfoxide and analysis parameters on NF-κB nuclear translocation and HCI data quality was optimized. The BAY-11-7085, the positive control for inhibiting NF-κB and Western blot assay were separately employed to verify the stability and reliability of the platform. Lastly, the effect of BHA on NO release, iNOS expression, IL-1β, IL-6, and TNF-α mRNA in LPS-induced RAW264.7 cells was detected. RESULTS The optimal conditions for measuring NF-κB translocation in LPS-induced RAW264.7 cells by HCI were established. Cells that do not exceed 22 passages were seeded at a density of 10 k cells/well and pretreated with compounds following 200 ng/mL LPS for 40 min. Parameters including nuclear area of 65 μm2, cell area of 80 μm2, collar of 0.9 μm and sensitivity of 25% were recommended for image segmentation algorithms in the analysis workstation. Benzoylhypaconine from aconite was screened for the first time as an anti-inflammatory candidate by the established HCI platform. The inhibitory effect of benzoylhypaconine on NF-κB translocation was verified by Western blot. Furthermore, benzoylhypaconine reduced the release of NO, inhibited the expression of iNOS, decreased the mRNA levels of IL-1β, IL-6, and TNF-α. CONCLUSION The established HCI platform could be applied to screen anti-inflammatory compounds by measuring the NF-κB nuclear translocation in LPS-induced RAW264.7 cells.
Collapse
Affiliation(s)
- Yan-Yu Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, P.R. China.,Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Yun-Da Yao
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, P.R. China.,Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Qi-Qing Cheng
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, P.R. China.,Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Yu-Feng Huang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, P.R. China.,Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Hua Zhou
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, P.R. China.,Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.,Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai City, Guangdong Province 519000, P.R. China
| |
Collapse
|
26
|
He Y, Peng X, Liu Y, Wu Q, Zhou Q, Huang Y, Liu S, Hu L, Fang Z, Lin Y, Xu S, Feng B, Li J, Jiang X, Zhuo Y, Wu D, Che L. Long-term maternal intake of inulin exacerbated the intestinal damage and inflammation of offspring rats in a DSS-induced colitis model. Food Funct 2022; 13:4047-4060. [PMID: 35315466 DOI: 10.1039/d1fo03675k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study aimed to investigate the effects of long-term maternal intake of inulin on intestinal morphology, permeability, inflammation and microbiota of offspring rats treated with dextran sulfate sodium (DSS). Sixteen female adult Sprague-Dawley rats were assigned to two groups receiving the fiber-free diet (FFD) or inulin diet (INU, 5% inulin) for three parities. The offspring weaned rats (third-parity) were fed with the same diet for four weeks until receiving 6% DSS for 7 days; the four groups were as follows: FFD, FFD + DSS, INU and INU + DSS. The results showed that maternal intake of inulin increased the histopathology score and activity of diamine oxidase (DAO) in serum, and the highest histopathology scores and activity of DAO were observed in INU + DSS rats. Maternal intake of inulin increased the activity of myeloperoxidase (MPO), mRNA expressions of inflammatory factors and protein expression of IL-1β in colonic tissues. Likewise, INU + DSS rats had the highest activity of MPO and mRNA expressions of inflammatory factors in colonic tissues. Maternal intake of inulin increased the abundances of Bacteroidetes, Bacteroides and Parasutterella, which were the highest enriched in INU + DSS rats. The level of acetate in the colonic digesta of INU + DSS rats was lower than that in FFD and INU rats. These results indicated that long-term maternal intake of inulin exacerbated the intestinal damage and inflammation of DSS-induced offspring rats, associated with the decreased level of acetate and altered intestinal microbiota.
Collapse
Affiliation(s)
- Ying He
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xie Peng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yang Liu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Qing Wu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Qiang Zhou
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yingyan Huang
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shiya Liu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Liang Hu
- College of Food Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Jian Li
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Xuemei Jiang
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - De Wu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
27
|
Paparo L, Maglio MA, Cortese M, Bruno C, Capasso M, Punzo E, Ferrucci V, Lasorsa VA, Viscardi M, Fusco G, Cerino P, Romano A, Troncone R, Zollo M. A New Butyrate Releaser Exerts a Protective Action against SARS-CoV-2 Infection in Human Intestine. Molecules 2022; 27:862. [PMID: 35164139 PMCID: PMC8838168 DOI: 10.3390/molecules27030862] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/25/2022] [Indexed: 12/12/2022] Open
Abstract
Butyrate is a major gut microbiome metabolite that regulates several defense mechanisms against infectious diseases. Alterations in the gut microbiome, leading to reduced butyrate production, have been reported in patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. A new butyrate releaser, useful for all the known applications of butyrate, presenting physiochemical characteristics suitable for easy oral administration, (N-(1-carbamoyl-2-phenyl-ethyl) butyramide (FBA), has been recently developed. We investigated the protective action of FBA against SARS-CoV-2 infection in the human small intestine and enterocytes. Relevant aspects of SARS-CoV-2 infection were assessed: infectivity, host functional receptor angiotensin-converting enzyme-2 (ACE2), transmembrane protease serine 2 (TMPRSS2), neuropilin-1 (NRP1), pro-inflammatory cytokines expression, genes involved in the antiviral response and the activation of Nf-kB nuclear factor (erythroid-derived 2-like) 2 (Nfr2) pathways. We found that FBA positively modulates the crucial aspects of the infection in small intestinal biopsies and human enterocytes, reducing the expression of ACE2, TMPRSS2 and NRP1, pro-inflammatory cytokines interleukin (IL)-15, monocyte chemoattractant protein-1 (MCP-1) and TNF-α, and regulating several genes involved in antiviral pathways. FBA was also able to reduce the number of SARS-CoV-2-infected cells, and ACE2, TMPRSS2 and NRP1 expression. Lastly, through the inhibition of Nf-kB and the up-regulation of Nfr2, it was also able to reduce the expression of pro-inflammatory cytokines IL-15, MCP-1 and TNF-α in human enterocytes. The new butyrate releaser, FBA, exerts a preventive action against SARS-CoV-2 infection. It could be considered as an innovative strategy to limit COVID-19.
Collapse
Affiliation(s)
- Lorella Paparo
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Maria Antonia Maglio
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
| | - Maddalena Cortese
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Cristina Bruno
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Mario Capasso
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
| | - Erika Punzo
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Veronica Ferrucci
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
| | - Vito Alessandro Lasorsa
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Maurizio Viscardi
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Giovanna Fusco
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Pellegrino Cerino
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| | - Alessia Romano
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
| | - Riccardo Troncone
- Dipartimento di Scienze Mediche Translazionali, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.A.M.); (M.C.); (C.B.); (E.P.); (R.T.)
| | - Massimo Zollo
- CEINGE—Advanced Biotechnologies s.c.ar.l., Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.C.); (V.F.); (V.A.L.); (A.R.); (M.Z.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy
- DAI Medicina di Laboratorio e Trasfusionale, AOU Azienda Ospedaliera, Università degli Studi di Napoli “Federico II”, 80131 Napoli, Italy; (M.V.); (G.F.); (P.C.)
| |
Collapse
|
28
|
Han NR, Kim KC, Kim JS, Park HJ, Ko SG, Moon PD. SBT (Composed of Panax ginseng and Aconitum carmichaeli) and Stigmasterol Enhances Nitric Oxide Production and Exerts Curative Properties as a Potential Anti-Oxidant and Immunity-Enhancing Agent. Antioxidants (Basel) 2022; 11:199. [PMID: 35204082 PMCID: PMC8868359 DOI: 10.3390/antiox11020199] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
Immune dysregulation is a risk factor for several diseases, including infectious diseases. Immunostimulatory agents have been used for the treatment of immune dysregulation, but deleterious adverse effects have been reported. The present study aims to establish the anti-oxidant and immunity-enhancing effects of Sambu-Tang (SBT), composed of Panax ginseng and Aconitum carmichaeli, and stigmasterol (Stig), an active compound of SBT. Immune-related factors were analyzed in RAW264.7 macrophage cells, mouse primary splenocytes, and the serum and spleen of cyclophosphamide-induced immunosuppressed mice. Results showed that the production levels of nitric oxide (NO) and expression levels of inducible NO synthase and heme oxygenase-1 were increased following SBT or Stig treatment in RAW264.7 cells. SBT or Stig increased the production levels of G-CSF, IFN-γ, IL-12, IL-2, IL-6, and TNF-α and induced the activation of NF-κB in RAW264.7 cells. SBT or Stig promoted splenic lymphocyte proliferation and increased splenic NK cell cytotoxic activity. In addition, SBT or Stig enhanced the levels of IFN-γ, IL-12, IL-2, IL-6, or TNF-α in the serum and spleen of the immunosuppressed mice. SBT or Stig increased the superoxide dismutase activity in the spleen. Collectively, SBT and Stig possess anti-oxidant and immunomodulatory activities, so they may be considered effective natural compounds for the treatment of various symptoms caused by immune dysregulation.
Collapse
Affiliation(s)
- Na-Ra Han
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Kyeoung-Cheol Kim
- Majors in Plant Resource and Environment, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea; (K.-C.K.); (J.-S.K.)
| | - Ju-Sung Kim
- Majors in Plant Resource and Environment, College of Agriculture & Life Sciences, SARI, Jeju National University, Jeju 63243, Korea; (K.-C.K.); (J.-S.K.)
| | - Hi-Joon Park
- Department of Anatomy & Information Sciences, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Seong-Gyu Ko
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea;
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Phil-Dong Moon
- Center for Converging Humanities, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
29
|
Bifidobacterium Longum: Protection against Inflammatory Bowel Disease. J Immunol Res 2021; 2021:8030297. [PMID: 34337079 PMCID: PMC8324359 DOI: 10.1155/2021/8030297] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023] Open
Abstract
The prevalence of inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), increases gradually worldwide in the past decades. IBD is generally associated with the change of the immune system and gut microbiota, and the conventional treatments usually result in some side effects. Bifidobacterium longum, as colonizing bacteria in the intestine, has been demonstrated to be capable of relieving colitis in mice and can be employed as an alternative or auxiliary way for treating IBD. Here, the mechanisms of the Bifidobacterium longum in the treatment of IBD were summarized based on previous cell and animal studies and clinical trials testing bacterial therapies. This review will be served as a basis for future research on IBD treatment.
Collapse
|
30
|
Kang C, Song CH, Kim N, Nam RH, Choi SI, Yu JE, Nho H, Choi JA, Kim JW, Na HY, Lee HN, Surh YJ. The Enhanced Inhibitory Effect of Estrogen on PD-L1 Expression Following Nrf2 Deficiency in the AOM/DSS Model of Colitis-Associated Cancer. Front Oncol 2021; 11:679324. [PMID: 34307147 PMCID: PMC8297827 DOI: 10.3389/fonc.2021.679324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a dual role in carcinogenesis. We previously reported that Nrf2 deficiency enhances the anti-tumorigenic effect of 17β-estradiol (E2) in an azoxymethane (AOM)/dextran sodium sulfate (DSS) model of colitis-associated cancer (CAC). Herein, we aimed to determine a possible explanation for our recent work and investigated the immune microenvironment represented by programmed death-ligand 1 (PD-L1) expression. One week after the AOM injection, mice were administered with DSS in drinking water for seven days; daily E2 injections were intraperitoneally administered during this period. The mice were sacrificed 16 weeks after AOM injection and analyzed for PD-L1 expression in the distal colon tissues using Western blotting and immunohistochemistry (IHC). Based on Western blotting results, PD-L1 expression was reduced in Nrf2 knockout (KO) female and E2-treated male mice when compared with their wild-type counterparts, following AOM/DSS treatment; this supports the association of PD-L1 expression with tumor progression. Additionally, this finding was in good agreement with the IHC results for PD-L1. Furthermore, we observed that PD-L1 is predominantly expressed in stromal cells rather than on epithelial cells in the colon. Western blotting revealed that PD-L1 expression in the colon positively correlates with expressions of inducible nitric oxide synthase (iNOS) (male, P = 0.002; female, P <0.001) and cyclooxygenase-2 (COX-2) (male, P <0.001; female, P <0.001). Collectively, our findings indicate that estrogen ameliorates the immune microenvironment represented by PD-L1 expression and enhances its effect in the absence of Nrf2.
Collapse
Affiliation(s)
- Changhee Kang
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Chin-Hee Song
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ryoung Hee Nam
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soo In Choi
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jeong Eun Yu
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Heewon Nho
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin A Choi
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin Won Kim
- Departments of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hee Young Na
- Departments of Pathology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ha-Na Lee
- Laboratory of Immunology, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea.,Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
31
|
Krasulova K, Illes P. Intestinal interplay of quorum sensing molecules and human receptors. Biochimie 2021; 189:108-119. [PMID: 34186126 DOI: 10.1016/j.biochi.2021.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 12/20/2022]
Abstract
Human gut is in permanent contact with microorganisms that play an important role in many physiological processes including metabolism and immunologic activity. These microorganisms communicate and manage themself by the quorum sensing system (QS) that helps to coordinate optimal growth and subsistence by activating signaling pathways that regulate bacterial gene expression. Diverse QS molecules produced by pathogenic as well as resident microbiota have been found throughout the human gut. However, even a host can by affected by these molecules. Intestinal and immune cells possess a range of molecular targets for QS. Our present knowledge on bacteria-cell communication encompasses G-protein-coupled receptors, nuclear receptors and receptors for bacterial cell-wall components. The QS of commensal bacteria has been approved as a protective factor with favourable effects on intestinal homeostasis and immunity. Signaling molecules of QS interacting with above-mentioned receptors thus parcipitate on maintaining of barrier functions, control of inflammation processes and increase of resistance to pathogen colonization in host organisms. Pathogens QS molecules can have a dual function. Host cells are able to detect the ongoing infection by monitoring the presence and changes in concentrations of QS molecules. Such information can help to set the most effective immune defence to prevent or overcome the infection. Contrary, pathogens QS signals can target the host receptors to deceive the immune system to get the best conditions for growth. However, our knowledge about communication mediated by QS is still limited and detailed understanding of molecular mechanisms of QS signaling is desired.
Collapse
Affiliation(s)
- Kristyna Krasulova
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic.
| | - Peter Illes
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Slechtitelu 27, 783 71, Olomouc, Czech Republic
| |
Collapse
|
32
|
Reyes-García J, Montaño LM, Carbajal-García A, Wang YX. Sex Hormones and Lung Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:259-321. [PMID: 34019274 DOI: 10.1007/978-3-030-68748-9_15] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inflammation is a characteristic marker in numerous lung disorders. Several immune cells, such as macrophages, dendritic cells, eosinophils, as well as T and B lymphocytes, synthetize and release cytokines involved in the inflammatory process. Gender differences in the incidence and severity of inflammatory lung ailments including asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (PF), lung cancer (LC), and infectious related illnesses have been reported. Moreover, the effects of sex hormones on both androgens and estrogens, such as testosterone (TES) and 17β-estradiol (E2), driving characteristic inflammatory patterns in those lung inflammatory diseases have been investigated. In general, androgens seem to display anti-inflammatory actions, whereas estrogens produce pro-inflammatory effects. For instance, androgens regulate negatively inflammation in asthma by targeting type 2 innate lymphoid cells (ILC2s) and T-helper (Th)-2 cells to attenuate interleukin (IL)-17A-mediated responses and leukotriene (LT) biosynthesis pathway. Estrogens may promote neutrophilic inflammation in subjects with asthma and COPD. Moreover, the activation of estrogen receptors might induce tumorigenesis. In this chapter, we summarize the most recent advances in the functional roles and associated signaling pathways of inflammatory cellular responses in asthma, COPD, PF, LC, and newly occurring COVID-19 disease. We also meticulously deliberate the influence of sex steroids on the development and progress of these common and severe lung diseases.
Collapse
Affiliation(s)
- Jorge Reyes-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico.,Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Luis M Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Abril Carbajal-García
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
33
|
Heat-Treated Bifidobacterium longum CECT-7347: A Whole-Cell Postbiotic with Antioxidant, Anti-Inflammatory, and Gut-Barrier Protection Properties. Antioxidants (Basel) 2021; 10:antiox10040536. [PMID: 33808122 PMCID: PMC8067082 DOI: 10.3390/antiox10040536] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Non-viable preparations of probiotics, as whole-cell postbiotics, attract increasing interest because of their intrinsic technological stability, and their functional properties, such as immune system modulation, gut barrier maintenance, and protection against pathogens. However, reports on Bifidobacteria-derived postbiotics remain scarce. This study aims to demonstrate the functional properties of a heat-treated (HT), non-viable, Bifidobacterium longum strain, CECT-7347, a strain previously selected for its anti-inflammatory phenotype and ability to improve biomarkers of intestinal integrity in clinical trials. The study used the nematode Caenorhabditis elegans and HT-29 cell cultures as eukaryotic model systems. Our results show that HT-CECT-7347 preserves the capacity to protect against oxidative stress damage, while it also reduces acute inflammatory response and gut-barrier disruption, and inhibits bacterial colonization, by activating pathways related to innate immune function. These findings highlight the interest of the ingredient as a novel postbiotic and pave the way to broaden the range of HT-CECT-7347 applications in gut health.
Collapse
|
34
|
Jang KB, Kim JH, Purvis JM, Chen J, Ren P, Vazquez-Anon M, Kim SW. Effects of mineral methionine hydroxy analog chelate in sow diets on epigenetic modification and growth of progeny. J Anim Sci 2020; 98:5897043. [PMID: 32841352 PMCID: PMC7507415 DOI: 10.1093/jas/skaa271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
The study was conducted to determine the effects of mineral methionine hydroxy analog chelate (MMHAC) partially replacing inorganic trace minerals in sow diets on epigenetic and transcriptional changes in the muscle and jejunum of progeny. The MMHAC is zinc (Zn), manganese (Mn), and copper (Cu) chelated with methionine hydroxy analog (Zn-, Mn-, and Cu-methionine hydroxy analog chelate [MHAC]). On day 35 of gestation, 60 pregnant sows were allotted to two dietary treatments in a randomized completed block design using parity as a block: 1) ITM: inorganic trace minerals with zinc sulfate (ZnSO4), manganese oxide (MnO), and copper sulfate (CuSO4) and 2) CTM: 50% of ITM was replaced with MMHAC (MINTREX trace minerals, Novus International Inc., St Charles, MO). Gestation and lactation diets were formulated to meet or exceed NRC requirements. On days 1 and 18 of lactation, milk samples from 16 sows per treatment were collected to measure immunoglobulins (immunoglobulin G, immunoglobulin A, and immunoglobulin M) and micromineral concentrations. Two pigs per litter were selected to collect blood to measure the concentration of immunoglobulins in the serum, and then euthanized to collect jejunal mucosa, jejunum tissues, and longissimus muscle to measure global deoxyribonucleic acid methylation, histone acetylation, cytokines, and jejunal histomorphology at birth and day 18 of lactation. Data were analyzed using Proc MIXED of SAS. Supplementation of MMHAC tended to decrease (P = 0.059) body weight (BW) loss of sows during lactation and tended to increase (P = 0.098) piglet BW on day 18 of lactation. Supplementation of MMHAC increased (P < 0.05) global histone acetylation and tended to decrease myogenic regulatory factor 4 messenger ribonucleic acid (mRNA; P = 0.068) and delta 4-desaturase sphingolipid1 (DEGS1) mRNA (P = 0.086) in longissimus muscle of piglets at birth. Supplementation of MMHAC decreased (P < 0.05) nuclear factor kappa B mRNA in the jejunum and DEGS1 mRNA in longissimus muscle and tended to decrease mucin-2 (MUC2) mRNA (P = 0.057) and transforming growth factor-beta 1 (TGF-β1) mRNA (P = 0.057) in the jejunum of piglets on day 18 of lactation. There were, however, no changes in the amounts of tumor necrosis factor-alpha, interleukin-8, TGF-β, MUC2, and myogenic factor 6 in the tissues by MMHAC. In conclusion, maternal supplementation of MMHAC could contribute to histone acetylation and programming in the fetus, which potentially regulates intestinal health and skeletal muscle development of piglets at birth and weaning, possibly leading to enhanced growth of their piglets.
Collapse
Affiliation(s)
- Ki Beom Jang
- Department of Animal Science, North Carolina State University, Raleigh, NC
| | - Jong Hyuk Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC
| | | | | | - Ping Ren
- Novus International, Inc., St. Charles, MO
| | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC
| |
Collapse
|
35
|
Arya VS, Kanthlal SK, Linda G. The role of dietary polyphenols in inflammatory bowel disease: A possible clue on the molecular mechanisms involved in the prevention of immune and inflammatory reactions. J Food Biochem 2020; 44:e13369. [PMID: 32885438 DOI: 10.1111/jfbc.13369] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/08/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD) is one of the major complications of the gastrointestinal tract, characterized by chronic inflammation, which disturbs the quality of life of the affected individuals. Genetic predisposition, immune, inflammatory, and enzyme-mediated signaling cascades are the primary mechanisms involved in the pathogenesis of the disease. Currently, the treatment strategy involves the maintenance of remission and induction of inflammation by anti-inflammatory agents and immune suppressants. Polyphenol-containing diets, including fruits and vegetables of regular use, possess anti-inflammatory, and antioxidant potential through the inhibition of major contributing pathways to IBD. This review discusses the role of these dietary polyphenols in downregulating the major signaling cascades in IBD. Our review encourages the development of nutritional strategies to improve the efficiency of current therapies for IBD and reduce the risks of side effects associated with conventional therapy. PRACTICAL APPLICATIONS: At present, almost every third person in society is under stress and having chronic disorders like diabetes, arthritis, allergy, cardiovascular disease, IBD, etc. This insists on the direct/indirect role of changes in the lifestyle for such deterioration in society. This review would emphasize the medicinal value of polyphenols present in fruits and vegetables for chronic inflammatory disorders. This concept portrays the food components which have the potential to promote health, improve general well-being, and reduce the risk of IBD. We propose to add fruits with bioactive polyphenols in the regular diet to help in preventing the immune-mediated intestinal chronic inflammatory syndrome and reduce the risks of colorectal cancer development.
Collapse
Affiliation(s)
- V S Arya
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - S K Kanthlal
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| | - Geevarghese Linda
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, Kerala, India
| |
Collapse
|
36
|
Bi Y, Ding Y, Wu J, Miao Z, Wang J, Wang F. Staphylococcus aureus induces mammary gland fibrosis through activating the TLR/NF-κB and TLR/AP-1 signaling pathways in mice. Microb Pathog 2020; 148:104427. [PMID: 32783982 DOI: 10.1016/j.micpath.2020.104427] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
To investigate the TLR-NF-κB/AP-1 pathways in S. aureus infection-induced mammary gland fibrosis, mice were infected with S. aureus isolated from the mammary glands of cows with mastitis. Lactating mice were divided into three groups: control group (CON); PBS control group (PBS) and the S. aureus-treated group (S. aureus). Pathological observations revealed that neutrophil infiltration into mammary gland tissue was obviously induced by S. aureus at the early stage of infection (1-7 d). With persistent S. aureus infection, mammary gland fibrosis developed and was characterized by infiltration and proliferation of macrophage, lymphocyte and fibroblast and ECM hyperplasia (7-21 d). Immunohistochemistry staining showed upregulation of fibrosis associated cytokines viz bFGF and PDGF-BB. Real-time qPCR and Western blot analysis revealed that transcription and translation of TLR2, TLR4, bFGF, PDGF-BB, α-SMA and COL Ⅰ α1 was significantly upregulated by S. aureus. NF-κB p65 and AP-I c-jun were translocated into the nucleus after S. aureus infection. There was no remarkable difference between the CON and PBS groups. The datas indicate that mammary gland fibrosis in mice is induced by S. aureus, which promotes cytokine release and the expression of ECM though activating the TLR/NF-κB p65 and TLR/AP-1 c-jun signaling pathways.
Collapse
Affiliation(s)
- Yannan Bi
- Department of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Zhaowuda Road 306, 010018, Hohhot, Inner Mongolia, People's Republic of China; School of Basic Medical Science and Forensic Medicine, Baotou Medical College, Jianshe Road 31, 014040, Baotou, Inner Mongolia, People's Republic of China
| | - Yulin Ding
- Department of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Zhaowuda Road 306, 010018, Hohhot, Inner Mongolia, People's Republic of China
| | - Jianmei Wu
- The Inner Mongolia Autonomous Region Comprehensive Center for Disease Control and Prevention, Erdos Street 50, 010031, Hohhot, Inner Mongolia Autonomous Region, China
| | - Zengqiang Miao
- Department of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Zhaowuda Road 306, 010018, Hohhot, Inner Mongolia, People's Republic of China
| | - Jinling Wang
- Department of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Zhaowuda Road 306, 010018, Hohhot, Inner Mongolia, People's Republic of China
| | - Fenglong Wang
- Department of Veterinary Pathology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Zhaowuda Road 306, 010018, Hohhot, Inner Mongolia, People's Republic of China.
| |
Collapse
|
37
|
Huang Y, Qiu L, Mi X, Zhang Z, Xu D, Tao X, Xing K, Wu Q, Wei H. Hot-water extract of ripened Pu-erh tea attenuates DSS-induced colitis through modulation of the NF-κB and HIF-1α signaling pathways in mice. Food Funct 2020; 11:3459-3470. [PMID: 32239008 DOI: 10.1039/c9fo02803j] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Tea consumption has been found to be associated with low incidence of inflammatory bowel disease in Asian countries. However, there is very limited knowledge of such potential protection and its underlying mechanism. Ripened Pu-erh tea (RPT) belongs to the variety of microbial fermented tea, but its function regarding anti-inflammation remains unclear. In the present study, we investigated the effects of RPT on dextran sulfate sodium (DSS)-induced colitis in mice. The results demonstrated that RPT significantly relieved the loss of body weight, disease severity and shortening of colon length, and remarkably inhibited the secretion of pro-inflammatory cytokines by lessening the infiltration of inflammatory cells. Furthermore, we found that RPT suppressed the activation of the NF-κB pathway and down-regulated the expression of HIF-1α. Thus, it was concluded that RPT attenuated the progress of colitis via suppressing the HIF-1α/NF-κB signaling pathways thus reducing inflammation. This suggests that RPT may be a potential anti-inflammatory nutraceutical for the prevention and treatment of colonic colitis.
Collapse
Affiliation(s)
- Yina Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Liang Qiu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330047, China
| | - Xuan Mi
- Wanlongshan Tea Plantation, Pingxiang, Jiangxi 337000, China
| | - Zhihong Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Di Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Keyu Xing
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| | - Qinglong Wu
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA. and Texas Children's Microbiome Center, Texas Children's Hospital, Houston, TX 77030, USA
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
38
|
Tian X, Gao J, Liu M, Lei Y, Wang F, Chen J, Chu P, Gao J, Long F, Liang M, Long X, Chu H, Liu C, Li X, Sun Q, Li G, Yang Y. Small-Molecule Antagonist Targeting Exportin-1 via Rational Structure-Based Discovery. J Med Chem 2020; 63:3881-3895. [PMID: 32223194 DOI: 10.1021/acs.jmedchem.9b01663] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Exportin-1 (also named as CRM1) plays a prominent role in autoimmune disorders and has emerged as a potential therapeutic target for colitis. Here we report on the rational structure-based discovery of a small-molecule antagonist of exportin-1, LFS-829, with low-range nanomolar activities. The co-crystallographic structure, surface plasmon resonance binding assay, and cell-based phenotypic nuclear export functional assay validated that exportin-1 is a key target of LFS-829. Moreover, we demonstrated that the C528S mutation or the knockdown on exportin-1 can abolish the cellular activities of LFS-829. Strikingly, oral administration of LFS-829 can significantly reverse the pathological features of colitis model mice. We revealed that LFS-829 can attenuate dual NF-κB signaling and the Nrf2 cytoprotection pathway via targeting exportin-1 in colitis mice. Moreover, LFS-829 has a very low risk of cardiotoxicity and acute toxicity. Therefore, LFS-829 holds great promise for the treatment of colitis and may warrant translation for use in clinical trials.
Collapse
Affiliation(s)
- Xibao Tian
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Jiali Gao
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Meishuo Liu
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Yuqin Lei
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fangjun Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Chen
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, Liaoning 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Chu
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Jiujiao Gao
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Feida Long
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Minzhi Liang
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Xiangyu Long
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| | - Huiying Chu
- Laboratory of Molecular Modeling and Design, State key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Cuixia Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xueliang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qingxiang Sun
- Department of Pathology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 457 Zhongshan Road, Dalian 116023, China
| | - Yongliang Yang
- Laboratoy of Innovative Drug Discovery, School of Bioengineering, Dalian University of Technology, Dalian 116023, China
| |
Collapse
|
39
|
Chen Z, Zhang Y, Lin R, Meng X, Zhao W, Shen W, Fan H. Cronobacter sakazakii induces necrotizing enterocolitis by regulating NLRP3 inflammasome expression via TLR4. J Med Microbiol 2020; 69:748-758. [PMID: 32209170 DOI: 10.1099/jmm.0.001181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction. Neonatal infection with Cronobacter sakazakii can cause severe intestinal damage and necrotizing enterocolitis (NEC). The inflammasome and Toll-like receptors mediate intestinal damage caused by other intestinal pathogens causing NEC, but the exact mechanism is unclear.Aim. We evaluated the molecular mechanisms underlying C. sakazakii-induced NEC.Methodology. The effects of C. sakazakii treatment on two cell lines and a Sprague-Dawley rat model of NEC were evaluated by a cell death assay, western blot and real-time PCR analyses of the NLRP3 inflammasome and downstream factors, and observation of cell and intestinal damage.Results. C. sakazakii caused cellular damage in vitro, as well as intestinal damage in an animal model. NLRP3, caspase-1, TLR4 and MyD88, as well as the downstream factor IL-1β, were upregulated in C. sakazakii-infected J774A.1 and HT-29 cells. Western blotting showed that C. sakazakii-infected J774A.1 and HT-29 cells and the NEC rat model had higher expression levels of N-terminal gasdermin D (GSDMD) compared with those in the control groups. C. sakazakii and its components promote NF-κB expression via the TLR4/MyD88 signalling pathway, thereby regulating the NLRP3 inflammasome and mediating GSDMD cleavage, resulting in pyroptosis-induced intestinal damage.Conclusion. We found that C. sakazakii upregulates NF-κB via TLR4/MyD88 to promote activation of the NLRP3 inflammasome, leading to the up-regulation of downstream caspase-1, release of IL-1β, GSDMD-mediated pyroptosis and development of NEC. These findings clarify the mechanisms by which C. sakazakii contributes to NEC.
Collapse
Affiliation(s)
- Zhenhui Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Yiduo Zhang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Sichuan, PR China.,Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Ruqin Lin
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaojing Meng
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Zhao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Shen
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Hongying Fan
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, PR China
| |
Collapse
|
40
|
Chen L, Li D, Shao Y, Wang H, Liu Y, Zhang Y. Identifying Microbiota Signature and Functional Rules Associated With Bacterial Subtypes in Human Intestine. Front Genet 2019; 10:1146. [PMID: 31803234 PMCID: PMC6872643 DOI: 10.3389/fgene.2019.01146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Gut microbiomes are integral microflora located in the human intestine with particular symbiosis. Among all microorganisms in the human intestine, bacteria are the most significant subgroup that contains many unique and functional species. The distribution patterns of bacteria in the human intestine not only reflect the different microenvironments in different sections of the intestine but also indicate that bacteria may have unique biological functions corresponding to their proper regions of the intestine. However, describing the functional differences between the bacterial subgroups and their distributions in different individuals is difficult using traditional computational approaches. Here, we first attempted to introduce four effective sets of bacterial features from independent databases. We then presented a novel computational approach to identify potential distinctive features among bacterial subgroups based on a systematic dataset on the gut microbiome from approximately 1,500 human gut bacterial strains. We also established a group of quantitative rules for explaining such distinctions. Results may reveal the microstructural characteristics of the intestinal flora and deepen our understanding on the regulatory role of bacterial subgroups in the human intestine.
Collapse
Affiliation(s)
- Lijuan Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Daojie Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Ye Shao
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Hui Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yuqing Liu
- Anhui Province Key Laboratory of Farmland Ecological Conservation and Pollution Prevention, School of Resources and Environment, Anhui Agricultural University, Hefei, China
| | - Yunhua Zhang
- Anhui Province Key Laboratory of Farmland Ecological Conservation and Pollution Prevention, School of Resources and Environment, Anhui Agricultural University, Hefei, China
| |
Collapse
|
41
|
Kou H, Fu Y, He Y, Jiang J, Gao X, Zhao H. Chronic lead exposure induces histopathological damage, microbiota dysbiosis and immune disorder in the cecum of female Japanese quails (Coturnix japonica). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 183:109588. [PMID: 31450035 DOI: 10.1016/j.ecoenv.2019.109588] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/15/2019] [Accepted: 08/18/2019] [Indexed: 06/10/2023]
Abstract
Lead (Pb) is one of the most hazardous metals to human and wildlife and it also has multiple negative impacts on birds. However, its influences on bird gut morphology and intestinal microbiota were still unclear. We used female Japanese quails (Coturnix japonica) to examine the effects of chronic lead exposure (0, 50 ppm and 1000 ppm) on cecal histology, microbial communities and immune function. The results showed 50 ppm lead exposure caused subtle damages of cecum cell structure. However, 1000 ppm lead exposure caused severe cecum histopathological changes characterized by mucosa abscission, Lieberkühn glands destruction and lymphocyte proliferation. Moreover, both lead concentrations induced ultrastructural damages featured by nucleus pyknosis, mitochondrial vacuolation and microvilli contraction. Meanwhile, microbial community structure, species diversity, taxonomic compositions and taxa abundance in the cecum were affected by lead exposure. Furthermore, the mRNA relative expression of immunity-related genes such as interleukin 2 (IL-2) and gamma interferon (IFN-γ) was significantly downregulated while that of interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and natural killer kappa B (NF-κB) was significantly upregulated in the cecum of 50 and 1000 ppm lead exposure groups. We concluded that lead exposure may cause gut health impairment of female Japanese quails by inducing cecal histopathological changes, microbiota dysbiosis and cecal immune disorder.
Collapse
Affiliation(s)
- Honghong Kou
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Yuchen Fu
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Yu He
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Junxia Jiang
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China
| | - Xuebin Gao
- Shaanxi Institute of Zoology, Xi'an, 710032, China
| | - Hongfeng Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, China. ,cn
| |
Collapse
|
42
|
Alvarenga V, Silva PTD, Bonfá ND, Pêgo B, Nanini H, Bernardazzi C, Madi K, Baetas da Cruz W, Castelo-Branco MT, de Souza HSP, Schanaider A. Protective effect of adipose tissue-derived mesenchymal stromal cells in an experimental model of high-risk colonic anastomosis. Surgery 2019; 166:914-925. [PMID: 31519305 DOI: 10.1016/j.surg.2019.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/20/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Dehiscence of intestinal anastomosis results in high morbidity and mortality. The aim of this study was to investigate the effects of locally administered adipose tissue-derived mesenchymal stromal cells in a model of high-risk colonic anastomosis in rats. METHODS Seven days after induction of colitis with 2,4,6-trinitrobenzene sulfonic acid, Wistar rats were submitted to a transection of the descending colon followed by end-to-end anastomosis and were then treated with 2×106 adipose tissue-derived mesenchymal stromal cells (from the preperitoneal fat) or an acellular culture solution instilled onto the surface of the anastomosis. At day 14, after macroscopic survey of the abdominal cavity, the anastomotic area was submitted to histologic and immunohistochemical analysis, evaluation of myeloperoxidase activity, fibrosis, epithelial integrity, NF-κ B activation, expression of inflammatory cytokines, and extracellular matrix-related genes. RESULTS Anastomotic leakage and mortality associated with high-risk anastomosis decreased with treatment with adipose tissue-derived mesenchymal stromal cells (P < .03). Application of adipose tissue-derived mesenchymal stromal cells resulted in lower histologic scores (P = .011), decreased deposition of collagen fibers (P = .003), preservation of goblet cells (P = .033), decreased myeloperoxidase activity (P = .012), decreased accumulation of CD4+ T-cells (P = .014) and macrophages (P = .011) in the lamina propria, a decrease in the number of apoptotic cells (P = .008), and the activation of NF-κ B (P = .036). Overexpression of IL-17, TNF-α , IFN-γ, and metalloproteinases in the acellular culture solution-treated, high-risk anastomosis group decreased (P < .05) to near normal values with adipose tissue-derived mesenchymal stromal cells treatment. CONCLUSION Improvements in outcomes of a high-risk colonic anastomosis with adipose tissue-derived mesenchymal stromal cells therapy reflect the immunomodulatory activity and healing effect of these cells, even after just topical administration and reinforces their use in future translational research.
Collapse
Affiliation(s)
- Valter Alvarenga
- Centro de Cirurgia Experimental, Programa de Pós-Graduação em Ciências Cirúrgicas, Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Beatriz Pêgo
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Hayandra Nanini
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Cláudio Bernardazzi
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Brazil
| | - Kalil Madi
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wagner Baetas da Cruz
- Laboratório Translacional em Fisiologia Molecular (LabTrans) do Centro de Cirurgia Experimental, Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Morgana Teixeira Castelo-Branco
- Laboratório de Imunologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Alberto Schanaider
- Centro de Cirurgia Experimental, Programa de Pós-Graduação em Ciências Cirúrgicas, Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Wang H, Zhang Q, Niu Y, Zhang X, Lu R. Surface-layer protein from Lactobacillus acidophilus NCFM attenuates tumor necrosis factor-α-induced intestinal barrier dysfunction and inflammation. Int J Biol Macromol 2019; 136:27-34. [DOI: 10.1016/j.ijbiomac.2019.06.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/15/2022]
|
44
|
SAW TY, MALIK NA, LIM KP, TEO CWL, WONG ESM, KONG SC, FONG CW, PETKOV J, YAP WN. Oral Supplementation of Tocotrienol-Rich Fraction Alleviates Severity of Ulcerative Colitis in Mice. J Nutr Sci Vitaminol (Tokyo) 2019; 65:318-327. [DOI: 10.3177/jnsv.65.318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Tzuen Yih SAW
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | - Najib Abdul MALIK
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | - Kee Pah LIM
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | - Cheryl Wei Ling TEO
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | | | - San Choon KONG
- Gastroenterology & Hepatology Department, Singapore General Hospital
| | - Chee Wai FONG
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | - Jordan PETKOV
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| | - Wei Ney YAP
- Research & Development Department, Davos Life Science
- Research & Development Department, KL-Kepong Oleomas Sdn Bhd (KLK Oleo)
| |
Collapse
|
45
|
Al Kalaldeh M, Gibson J, Lee SH, Gondro C, van der Werf JHJ. Detection of genomic regions underlying resistance to gastrointestinal parasites in Australian sheep. Genet Sel Evol 2019; 51:37. [PMID: 31269896 PMCID: PMC6609385 DOI: 10.1186/s12711-019-0479-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed at identifying genomic regions that underlie genetic variation of worm egg count, as an indicator trait for parasite resistance in a large population of Australian sheep, which was genotyped with the high-density 600 K Ovine single nucleotide polymorphism array. This study included 7539 sheep from different locations across Australia that underwent a field challenge with mixed gastrointestinal parasite species. Faecal samples were collected and worm egg counts for three strongyle species, i.e. Teladorsagia circumcincta, Haemonchus contortus and Trichostrongylus colubriformis were determined. Data were analysed using genome-wide association studies (GWAS) and regional heritability mapping (RHM). RESULTS Both RHM and GWAS detected a region on Ovis aries (OAR) chromosome 2 that was highly significantly associated with parasite resistance at a genome-wise false discovery rate of 5%. RHM revealed additional significant regions on OAR6, 18, and 24. Pathway analysis revealed 13 genes within these significant regions (SH3RF1, HERC2, MAP3K, CYFIP1, PTPN1, BIN1, HERC3, HERC5, HERC6, IBSP, SPP1, ISG20, and DET1), which have various roles in innate and acquired immune response mechanisms, as well as cytokine signalling. Other genes involved in haemostasis regulation and mucosal defence were also detected, which are important for protection of sheep against invading parasites. CONCLUSIONS This study identified significant genomic regions on OAR2, 6, 18, and 24 that are associated with parasite resistance in sheep. RHM was more powerful in detecting regions that affect parasite resistance than GWAS. Our results support the hypothesis that parasite resistance is a complex trait and is determined by a large number of genes with small effects, rather than by a few major genes with large effects.
Collapse
Affiliation(s)
- Mohammad Al Kalaldeh
- Cooperative Research Centre for Sheep Industry Innovation, Armidale, NSW, 2351, Australia. .,School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia.
| | - John Gibson
- Cooperative Research Centre for Sheep Industry Innovation, Armidale, NSW, 2351, Australia.,School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia
| | - Sang Hong Lee
- Australian Centre for Precision Health, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, SA, 5000, Australia
| | - Cedric Gondro
- School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia.,College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI, 48824, USA
| | - Julius H J van der Werf
- Cooperative Research Centre for Sheep Industry Innovation, Armidale, NSW, 2351, Australia.,School of Environmental and Rural Science, University of New England, Armidale, NSW, 2351, Australia
| |
Collapse
|
46
|
Cancer testis antigen 55 deficiency attenuates colitis-associated colorectal cancer by inhibiting NF-κB signaling. Cell Death Dis 2019; 10:304. [PMID: 30944312 PMCID: PMC6447546 DOI: 10.1038/s41419-019-1537-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022]
Abstract
Colitis-associated cancer (CAC), a prototype of inflammation-associated cancer, is one of the most common gastrointestinal tumors. As a potential cancer testis antigen (CT antigen), cancer testis antigen 55 (CT55) is expressed in different tumors and normal testes. However, its role in CAC remains unknown. Here, we identified CT55 as a new potent promoter of CAC. We discovered that Ct55 deficiency alleviated inflammatory responses, decreased cell proliferation and colitis-associated tumorigenesis in an azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model. Mechanistically, CT55 acts as an accelerator of tumor necrosis factor (TNF)-α-induced nuclear factor-κB (NF-κB) signaling. Upon stimulation with TNF-α, CT55 interacts with the IκB kinase (IKK) complex, which increases the phosphorylation of IKKα/β and activates IKK–p65 signaling, while knockout of CT55 blocks IKK–p65 signaling. Notably, inhibition of IKK abolished the positive effect of CT55 on NF-κB activation. Collectively, our findings strongly indicate that CT55 deficiency suppresses the development of CAC and that the CT55-TNF-α-induced NF-κB axis may represent a promising target for CAC therapy.
Collapse
|
47
|
Pêgo B, Martinusso CA, Bernardazzi C, Ribeiro BE, de Araujo Cunha AF, de Souza Mesquita J, Nanini HF, Machado MP, Castelo-Branco MTL, Cavalcanti MG, de Souza HSP. Schistosoma mansoni Coinfection Attenuates Murine Toxoplasma gondii-Induced Crohn's-Like Ileitis by Preserving the Epithelial Barrier and Downregulating the Inflammatory Response. Front Immunol 2019; 10:442. [PMID: 30936867 PMCID: PMC6432985 DOI: 10.3389/fimmu.2019.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/19/2019] [Indexed: 12/11/2022] Open
Abstract
Background and aims: Mice orally infected with T. gondii develop Crohn's disease (CD)-like enteritis associated with severe mucosal damage and a systemic inflammatory response, resulting in high morbidity and mortality. Previously, helminthic infections have shown therapeutic potential in experimental colitis. However, the role of S. mansoni in T. gondii-induced CD-like enteritis has not been elucidated. Our study investigated the mechanisms underlying T. gondii-induced ileitis and the potential therapeutic effect of S. mansoni coinfection. Methods: C57BL/6 mice were infected by subcutaneous injection of cercariae of the BH strain of S. mansoni, and 7-9 weeks later, they were orally infected with cysts of the ME49 strain of T. gondii. After euthanasia, the ileum was removed for histopathological analysis; staining for goblet cells; immunohistochemistry characterizing mononuclear cells, lysozyme expression, apoptotic cells, and intracellular pathway activation; and measuring gene expression levels by real-time PCR. Cytokine concentrations were measured in the serial serum samples and culture supernatants of the ileal explants, in addition to myeloperoxidase (MPO) activity. Results:T. gondii-monoinfected mice presented dense inflammatory cell infiltrates and ulcerations in the terminal ileum, with abundant cell extrusion, apoptotic bodies, and necrosis; these effects were absent in S. mansoni-infected or coinfected animals. Coinfection preserved goblet cells and Paneth cells, remarkably depleted in T. gondii-infected mice. Densities of CD4- and CD11b-positive cells were increased in T. gondii- compared to S. mansoni-infected mice and controls. MPO was significantly increased among T. gondii-mice, while attenuated in coinfected animals. In T. gondii-infected mice, the culture supernatants of the explants showed increased concentrations of TNF-alpha, IFN-gamma, and IL-17, and the ileal tissue revealed increased expression of the mRNA transcripts for IL-1 beta, NOS2, HMOX1, MMP3, and MMP9 and activation of NF-kappa B and p38 MAPK signaling, all of which were counterregulated by S. mansoni coinfection. Conclusion:S. mansoni coinfection attenuates T. gondii-induced ileitis by preserving mucosal integrity and downregulating the local inflammatory response based on the activation of NF-kappa B and MAPK. The protective function of prior S. mansoni infection suggests the involvement of innate immune mechanisms and supports a conceptually new approach to the treatment of chronic inflammatory diseases, including CD.
Collapse
Affiliation(s)
- Beatriz Pêgo
- Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Claudio Bernardazzi
- Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Hayandra F. Nanini
- Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Marta Guimarães Cavalcanti
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Infectious Diseases Clinic, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor S. P. de Souza
- Internal Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Wang F, Chen S, Deng L, Chen L, Huang Y, Tian M, Li C, Zhou X. Protective Effects of Astragaloside IV against LPS-Induced Endometritis in Mice through Inhibiting Activation of the NF-κB, p38 and JNK Signaling Pathways. Molecules 2019; 24:molecules24020373. [PMID: 30669661 PMCID: PMC6360020 DOI: 10.3390/molecules24020373] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/02/2019] [Accepted: 01/18/2019] [Indexed: 01/09/2023] Open
Abstract
Endometritis, inflammation of the endometrium, is a common reproductive obstacle disease that can lead to infertility in female animals. Astragaloside IV (AS IV), one of the major and active components of the Astragalus membranaceus (Fisch.) Bunge, is known for its anti-inflammatory effects. In the present study, the effects and mechanisms of AS IV on lipopolysaccharide (LPS)-induced endometritis were investigated using a mouse model. Female mice were prepared with AS IV (0.01 mg/g) by gavage for six days before being stimulated with LPS. The results showed that the histopathological changes, levels of inflammatory cytokines (IL-1β and TNF-α), concentration of NO, and myeloperoxidase (MPO) activity in LPS-induced uteri were attenuated significantly by pretreatment with AS IV. Furthermore, LPS-induced activations of NF-κB, p38, and JNK signal pathways were suppressed by pretreatment with AS IV. In conclusion, the data provided new evidence that AS IV effectively attenuates LPS-induced endometritis through inhibition of TLR4-mediated NF-κB, p38, and JNK signaling pathways, implying that AS IV might become a promising potential anti-inflammatory agent for endometritis and other inflammatory diseases.
Collapse
Affiliation(s)
- Fengge Wang
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Shuxiong Chen
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Liang Deng
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Lu Chen
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Yuwen Huang
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Meng Tian
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, 5333 Xian Road, Changchun, Jilin 130062, China.
| |
Collapse
|
49
|
Zhang H, Xu CN, Mine Y. Effects of a synthetic di‐phosphoserine peptide (SS‐2) on gene expression profiling against TNF‐α induced inflammation. Int J Food Sci Technol 2019. [DOI: 10.1111/ijfs.14077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hua Zhang
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| | - Cai Na Xu
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| | - Yoshinori Mine
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| |
Collapse
|
50
|
Yousefi‐Ahmadipour A, Rashidian A, Mirzaei MR, Farsinejad A, PourMohammadi‐Nejad F, Ghazi‐Khansari M, Ai J, Shirian S, Allahverdi A, Saremi J, Ebrahimi‐Barough S. Combination therapy of mesenchymal stromal cells and sulfasalazine attenuates trinitrobenzene sulfonic acid induced colitis in the rat: The S1P pathway. J Cell Physiol 2018; 234:11078-11091. [DOI: 10.1002/jcp.27944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Aliakbar Yousefi‐Ahmadipour
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Amir Rashidian
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Mohammad Reza Mirzaei
- Department of Clinical Biochemistry Faculty of Medicine Rafsanjan University of Medical Sciences Rafsanjan Iran
| | - Alireza Farsinejad
- Department of Hematology and Laboratory Sciences Faculty of Allied Medicine, Kerman University of Medical Sciences Kerman Iran
| | - Fatemeh PourMohammadi‐Nejad
- Department of Periodontics School of Dentistry, Rafsanjan University of Medical Sciences, Rafsanjan Kerman Iran
| | - Mahmoud Ghazi‐Khansari
- Department of Pharmacology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Sadegh Shirian
- Department of Pathology School of Veterinary Medicine, Shahrekord University Shahrekord Iran
| | - Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Jamileh Saremi
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Somayeh Ebrahimi‐Barough
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|