1
|
Zheng Z, Gao J, Ma Y, Hou X. Cellular and Molecular Mechanisms of Phytochemicals Against Inflammation-Associated Diseases and Viral Infection. Cell Biol Int 2025; 49:606-633. [PMID: 40091269 DOI: 10.1002/cbin.70011] [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: 12/13/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
Inflammation-associated diseases have become widespread and pose a significant threat to human health, and the therapeutic methods for diverse diseases are inadequate due to the undesirable effects of synthetic ingredients. Recently, more and more evidence indicated that phytochemicals, plant secondary metabolites, have numerous therapeutic functions against human diseases via affecting a variety of mechanisms with their distinct advantages of high efficiency and low toxicity. Here, we highlight the mechanisms of phytochemicals to hinder inflammation-associated diseases (including Inflammatory diseases, cardiovascular diseases, metabolic syndrome, neurological disorders, skin diseases, respiratory diseases, kidney diseases, gastrointestinal diseases, retinal diseases, viral infections) by regulating the crosstalk among various signal cascades (including MicroRNAs, SIRT1, DNMTs, NF-κB, NLRP3, TGF-β, the Gasdermin-mediated pyroptosis pathway), which can be considered as a novel and potential therapeutic strategy. Furthermore, phytochemicals could prevent virus infection by disturbing different targets in the virus replication cycle. However, natural plants have shown limited bioavailability due to their low water solubility, the use of adjuvants such as liposomal phytochemicals, phytochemical nanoparticles and phytochemicals-phospholipid complex promote their bioavailability to exhibit beneficial effects against various diseases. The purpose of this review is to explore the molecular mechanisms and promising applications of phytochemicals in the fields of inflammation-associated diseases and virus infection to provide some direction.
Collapse
Affiliation(s)
- Zhaodi Zheng
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| | - Junying Gao
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Yubing Ma
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| | - Xitan Hou
- College of Medical Imaging and Laboratory, Jining Medical University, Jining, China
| |
Collapse
|
2
|
Long D, Mao C, Zhang W, Zhu Y, Xu Y. Natural products for the treatment of ulcerative colitis: focus on the JAK/STAT pathway. Front Immunol 2025; 16:1538302. [PMID: 40078988 PMCID: PMC11897526 DOI: 10.3389/fimmu.2025.1538302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Ulcerative colitis (UC) is an autoimmune disease with an incompletely understood pathogenesis. The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway plays a key role in immune response and inflammation. More and more studies demonstrated that JAK/STAT signaling pathway is associated with the pathogenesis of UC. The JAK/STAT pathway affects UC in multiple ways by regulating intestinal inflammatory response, affecting intestinal mucosal barrier, modulating T cell homeostasis, and regulating macrophages. Encouragingly, natural products are promising candidates for the treatment of UC. Natural products have the advantage of being multi-targeted and rich in therapeutic modalities. This review summarized the research progress of JAK/STAT pathway-mediated UC. Furthermore, the latest studies on natural products targeting the JAK/STAT pathway for the treatment of UC were systematically summarized, including active ingredients such as arbutin, aloe polysaccharide, berberine, matrine, curcumin, Ginsenoside Rh2, and so on. The aim of this paper is to provide new ideas for drug development to regulate JAK/STAT signaling for treating UC.
Collapse
Affiliation(s)
- Dan Long
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei Zhang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yin Xu
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
3
|
Wei Z, Ni X, Cui H, Shu C, Peng Y, Liu J, Li Y. Engeletin attenuates the inflammatory response via inhibiting TLR4-NFκB signaling pathway in Crohn's disease-like colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118733. [PMID: 39181281 DOI: 10.1016/j.jep.2024.118733] [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: 07/05/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Smilax glabra rhizome has a long history been used for clinical purposes in traditional Chinese medicinal for treating various inflammatory conditions. Engeletin1 (ENG) is one of the most abundant bioactive compounds found in Smilax glabra rhizome, with anti-inflammatory, antioxidant, and ulcer-preventing activities. AIM OF THE STUDY The purpose of this study was to investigate the ability of ENG to alleviate inflammatory symptoms and improve epithelial barrier integrity utilize a 2,4,6-trinitrobenzene sulfonic acid2 (TNBS)-induced murine model in Crohn's disease3 (CD)-like colitis, and to characterize the underlying anti-inflammatory mechanisms of action. MATERIALS AND METHODS A colitis model was established in BALB/c mice and treated with ENG for 7 days. RAW264.7 macrophages were pre-treated with ENG and lipopolysaccharide4 (LPS) stimulation. The mice's weight and colon length were assessed. qPCR and Western blotting were used to analyze gene expression and TLR4-NFκB pathway. Flow cytometry was used to analyze the polarization states of the macrophages. RESULTS Treatment with ENG was sufficient to significantly alleviate symptoms of inflammation and colonic epithelial barrier integrity in treated mice. Significant inhibition of TNF-α, IL-1β, and IL-6 expression was observed following ENG treatment in vivo and in vitro. ENG was also determined to be capable of inhibiting the expression of iNOS and CD86, inhibited M1 macrophage polarization in vitro, as well as the TLR4-NFκB signaling pathway. Molecular docking showed a highly stable binding between ENG and TLR4. CONCLUSION ENG has been proven to alleviate inflammation and ameliorate the damage of epithelial barrier in CD-like colitis. ENG also suppressed the M1 macrophages polarization and the inhibited inflammatory cytokines. TLR4-NFκB signaling pathway, especially TLR4, may be the target of ENG. These data offer a new insight into the therapeutic mechanisms of ENG.
Collapse
Affiliation(s)
- Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - He Cui
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Chang Shu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China
| | - Jieyu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, PR China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China.
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, PR China.
| |
Collapse
|
4
|
Zhu Y, Wang Y, Zuo X, Liu S, Cao L, Wang J, Yang Q, Huang Q, Huang Q, Tian M, Ping Y, Wu Q. Inhibition SIRT1 to regulate FOXP3 or RORγt can restore the balance of Treg/Th17 axis in ulcerative colitis and enhance the anti-inflammatory effect of moxibustion. Front Immunol 2025; 15:1525469. [PMID: 39867884 PMCID: PMC11757129 DOI: 10.3389/fimmu.2024.1525469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is a chronic inflammatory disease. Patients with UC typically exhibit disruption of the Treg/Th17 immune axis, but its exact mechanism is still unclear. Methods This study first analyzed RNA- seq data from public databases of humans and mice, and in vitro cytology experiments were conducted to induce or inhibit the expression of SIRT1. In vivo, UC mice were treated with moxibustion and SIRT1 inhibitor EX-527 to confirm the changes in the transcription factors identified through analysis of the datasets. Results The results show that Treg/Th17 axis disruption is an important feature of UC. Differential gene expression and immune infiltration analysis showed that upstream transcription factors, including Forkhead box P3 (FOXP3), were significantly disrupted. In vitro cytology experiments, the results indicate that SIRT1 is activated in LPS induced inflammation, subsequently perturbing the Treg/Th17 immune balance axis. Finally, in vivo studies, the results have shown that administering EX-527 to inhibit SIRT1 leads to an increasing in FOXP3 expression and a decreasing in RORγt expression in UC colon tissue. In addition, the results indicate that traditional Chinese moxibustion can down regulate the expression of SIRT1, directly affecting the balance of Th17/Treg axis, and the combined use of EX-527 further improves the therapeutic effect of moxibustion. Conclusion Our research shows that inhibition SIRT1 can regulate Treg and Th17 immune balance axis. This finding indicates a new important potential target for the treatment of UC.
Collapse
MESH Headings
- Sirtuin 1/antagonists & inhibitors
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Colitis, Ulcerative/therapy
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/metabolism
- Moxibustion
- Animals
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Humans
- Mice
- Disease Models, Animal
- Male
- Carbazoles
Collapse
Affiliation(s)
- Yuanbing Zhu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yuemei Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaotong Zuo
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuqing Liu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Lishuang Cao
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Junmeng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qingqing Yang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhui Huang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qin Huang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Muqiu Tian
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yanling Ping
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaofeng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Acupuncture & Chronobiology Key Laboratory of Sichuan Province (Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan, China
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of Traditional Chinese Medicine), Ministry of Education, Chengdu, Sichuan, China
- Institute of Acupuncture and Homeostasis Regulation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Giammona A, Galuzzi BG, Imperia E, Gervasoni C, Remedia S, Restaneo L, Nespoli M, De Gara L, Tani F, Cicala M, Guarino MPL, Porro D, Cerasa A, Lo Dico A, Altomare A, Bertoli G. Chronic Gastrointestinal Disorders and miRNA-Associated Disease: An Up-to-Date. Int J Mol Sci 2025; 26:413. [PMID: 39796266 PMCID: PMC11720538 DOI: 10.3390/ijms26010413] [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: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic gastrointestinal disorders such as inflammatory bowel diseases (IBDs) and irritable bowel syndrome (IBS) impose significant health burdens globally. IBDs, encompassing Crohn's disease and ulcerative colitis, are multifactorial disorders characterized by chronic inflammation of the gastrointestinal tract. On the other hand, IBS is one of the principal gastrointestinal tract functional disorders and is characterized by abdominal pain and altered bowel habits. Although the precise etiopathogenesis of these disorders remains unclear, mounting evidence suggests that non-coding RNA molecules play crucial roles in regulating gene expression associated with inflammation, apoptosis, oxidative stress, and tissue permeability, thus influencing disease progression. miRNAs have emerged as possible reliable biomarkers, as they can be analyzed in the biological fluids of patients at a low cost. This review explores the roles of miRNAs in IBDs and IBS, focusing on their involvement in the control of disease hallmarks. By an extensive literature review and employing bioinformatics tools, we identified the miRNAs frequently studied concerning these diseases. Ultimately, specific miRNAs could be proposed as diagnostic biomarkers for IBDs and IBS. Their ability to be secreted into biofluids makes them promising candidates for non-invasive diagnostic tools. Therefore, understanding molecular mechanisms through the ways in which they regulate gastrointestinal inflammation and immune responses could provide new insights into the pathogenesis of IBDs and IBS and open avenues for miRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Alessandro Giammona
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Bruno Giovanni Galuzzi
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Elena Imperia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Clarissa Gervasoni
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Sofia Remedia
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Dipartimento di Scienze della Terra e del Mare (DISTEM), Università di Palermo, Via Archirafi, 22, 90123 Palermo, Italy
| | - Laura Restaneo
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Martina Nespoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Laura De Gara
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Flaminia Tani
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Michele Cicala
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
- Unit of Gastroenterology, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Michele Pier Luca Guarino
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
- Unit of Gastroenterology, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Danilo Porro
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, 20126 Milan, Italy
| | - Antonio Cerasa
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Alessia Lo Dico
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Annamaria Altomare
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
| | - Gloria Bertoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
6
|
Yang Q, Wang Z, Su J, Yang G, Liu H, Liu B, Cheng J, Dong B, Jafari H, Wang H, Zhang Y, Lei C, Dang R, Yu J. Comprehensive omics analysis of the fecal microbiome and serum metabolome in Dezhou donkey foals at the end of weaning and after weaning. Anim Sci J 2025; 96:e70021. [PMID: 39815660 DOI: 10.1111/asj.70021] [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: 10/10/2023] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 01/18/2025]
Abstract
Weaning is essential for foal growth and development. We determined the intestinal flora structure of donkey foals at the end of weaning (PreW_4d) and three stages after weaning (PostW_4d, PostW_8d, and PostW_15d) to explore the effects of weaning on intestinal development of donkey foals. The results showed that the main microbial flora in the gut of the donkey foal were Firmicutes and Bacteroides, and the proportion of Firmicutes gradually increased with weaning, which was an important reflection of the donkey foal's adaptability to the transition from lactose liquid feed to plant fiber solid feed. We also identified important microorganisms that maintain intestinal stability and boost immune, such as oscillospiraceae, Firmicutes, and lachnospiraceae. The metabolome showed that serum metabolites were mainly enriched in arachidonic acid metabolism and the tricarboxylic acid cycle (TCA cycle), which can influence energy metabolism, growth, and immunity in weaned donkey foals. We also found that the metabolite resveratrol was positively correlated with g_NK4A214_group and lactobacillus, which may have important implications for the prevention of diseases such as colon-inflammation in donkey foals. In summary, we provide a theoretical basis for studying the mechanism of intestinal microbiome and serum metabolite changes in weaning and postweaning donkey foals.
Collapse
Affiliation(s)
- Qiwen Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Zhaofei Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Jiangtian Su
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Ge Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Haibing Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Bing Liu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Jie Cheng
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Boying Dong
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Halima Jafari
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Hua Wang
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Youxin Zhang
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| | - Chuzhao Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Ruihua Dang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang, Shaanxi Province, China
| | - Jie Yu
- National Engineering Research Center for Gelatin-based Traditional Chinese Medicine, Dong-E-E-Jiao Co. Ltd, Dong-E Country, Shandong Province, China
| |
Collapse
|
7
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-w] [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: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
8
|
Sun D, Zhang Z, Xue J. MiRNAs: a new target for Chinese medicine to repair the intestinal barrier in the treatment of ulcerative colitis. Front Pharmacol 2024; 15:1446554. [PMID: 39185319 PMCID: PMC11341499 DOI: 10.3389/fphar.2024.1446554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic nonspecific inflammatory bowel disease whose pathogenesis remains unclear. Dysfunction of the intestinal mucosal barrier is closely related to the pathogenesis of UC, which is characterised by damage to the colon epithelial barrier, disruption of immune homeostasis, and persistent inflammatory cell infiltration. MicroRNAs (miRNAs) exhibit specific or differential expression in both UC animal models and patients, implicating their involvement in the pathogenesis of UC. In recent years there has been progress in using Traditional Chinese medicine (TCM) to regulate miRNA expression for repairing the intestinal mucosal barrier in UC, as demonstrated in animal and cell experiments. However, it has not been applied in a clinical setting and its underlying molecular mechanisms require further investigation. Therefore, this study systematically described the role of miRNAs in UC-induced intestinal barrier damage and the application of TCM to repair this intestinal barrier by regulating miRNA expression, offering new therapeutic targets for UC treatment.
Collapse
Affiliation(s)
- Dajuan Sun
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhongtao Zhang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jingwei Xue
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| |
Collapse
|
9
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in the role of resveratrol and its mechanism of action in common gynecological tumors. Front Pharmacol 2024; 15:1417532. [PMID: 39086397 PMCID: PMC11288957 DOI: 10.3389/fphar.2024.1417532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
The incidence of common gynecological malignancies remains high, with current treatments facing multiple limitations and adverse effects. Thus, continuing the search for safe and effective oncologic treatment strategies continues. Resveratrol (RES), a natural non-flavonoid polyphenolic compound, is widely found in various plants and fruits, such as grapes, Reynoutria japonica Houtt., peanuts, and berries. RES possesses diverse biological properties, including neuroprotective, antitumor, anti-inflammatory, and osteoporosis inhibition effects. Notably, RES is broadly applicable in antitumor therapy, particularly for treating gynecological tumors (cervical, endometrial, and ovarian carcinomas). RES exerts antitumor effects by promoting tumor cell apoptosis, inhibiting cell proliferation, invasion, and metastasis, regulating tumor cell autophagy, and enhancing the efficacy of antitumor drugs while minimizing their toxic side effects. However, comprehensive reviews on the role of RES in combating gynecological tumors and its mechanisms of action are lacking. This review aims to fill this gap by examining the RES antitumor mechanisms of action in gynecological tumors, providing valuable insights for clinical treatment.
Collapse
Affiliation(s)
- Qian Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dandan Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingchen Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
10
|
Gu Y, Lou Y, Zhou Z, Zhao X, Ye X, Wu S, Li H, Ji Y. Resveratrol for inflammatory bowel disease in preclinical studies: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1411566. [PMID: 38948464 PMCID: PMC11211549 DOI: 10.3389/fphar.2024.1411566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
Background: Inflammatory bowel disease (IBD) is a chronic condition that can be managed with treatment, but it is challenging to get IBD cured. Resveratrol, a non-flavonoid polyphenolic organic compound derived from various plants, has a potential effect on IBD. The current research was set out to investigate the therapeutic effects of resveratrol on animal models of IBD. Methods: A comprehensive search of PubMed, Embase, Web of Science, and Chinese databases was performed. The literature search process was completed independently by two people and reviewed by a third person. The risk of bias in the included literature was assessed using the Collaborative Approach to Meta Analysis and Review of Animal Data from Experimental Stroke (CAMARADES) 10-point quality checklist. The meta-analysis utilized Review Manager 5.4 software to evaluate the efficacy of resveratrol, with histopathological index as the primary outcome measure. Subgroup analysis was conducted based on this indicator. Additionally, meta-analyses were carried out on different outcomes reported in the literature, including final disease activity index, final body weight change, colon length, splenic index, and inflammatory factors. Results: After conducting a thorough literature search and selection process, a total of 28 studies were ultimately included in the analysis. It was found that over half of the selected studies had more than five items with low risk of bias in the bias risk assessment. Relevant datas from included literature indicated that the histopathological index of the resveratrol group was significantly lower than that of the control group (WMD = -2.58 [-3.29, -1.87]). Subgroup analysis revealed that higher doses of resveratrol (>80 mg/kg) had a better efficacy (WMD = -3.47 [-4.97, -1.98]). Furthermore, The data summary and quantitative analysis results of SI and colon length also showed that resveratrol was effective in alleviating intestinal mucosal pathological injury of IBD. In terms of biochemical indicators, the summary analysis revealed that resveratrol affected interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), interferon-γ (IFN-γ), malondialdehyde (MDA), myeloperoxidase (MPO), superoxide dismutase (SOD), and prostaglandin E2 (PGE2) significantly. These effects may be attributed to the mechanism of resveratrol in regulating immune response and inhibiting oxidative stress. Conclusion: This review suggests that resveratrol demonstrated a notable therapeutic impact in preclinical models of IBD, particularly at doses exceeding 80 mg/kg. This efficacy is attributed to the protective mechanisms targeting the intestinal mucosa involved in the pathogenesis of IBD through various pathways. As a result, resveratrol holds promising prospects for potential clinical use in the future.
Collapse
Affiliation(s)
- Yuting Gu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yijie Lou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhanyi Zhou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuan Zhao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolu Ye
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuwen Wu
- Department of Acupuncture and Moxibustion, Zhejiang Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhenjiang, China
| | - Haitao Li
- Department of Digestive System, Jinhua Municipal Hospital of Traditional Chinese Medicine, Jinhua, China
| | - Yunxi Ji
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Department of General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Yu X, Wang Y, Xu Y, Li X, Zhang J, Su Y, Guo L. Resveratrol attenuates intestinal epithelial barrier dysfunction via Nrf2/HO-1 pathway in dextran sulfate sodium-induced Caco-2 cells. Immun Inflamm Dis 2024; 12:e1193. [PMID: 38372468 PMCID: PMC10875904 DOI: 10.1002/iid3.1193] [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: 10/14/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/20/2024] Open
Abstract
INTRODUCTION The intestinal tract serves as an innate barrier, safeguarding the internal milieu from microorganisms and toxins. Various intestinal inflammatory diseases have a strong association with intestinal barrier dysfunction. The primary functional cells within the intestinal tract, intestinal epithelial cells (IECs) and their tight junctions (TJs), are crucial in preserving the integrity of this mechanical barrier. Resveratrol (Res), a plant-derived phenolic compound, exhibits a range of health-promoting benefits attributed to its anti-inflammatory properties. This study aims to examine Res's efficacy in bolstering IECs barrier function. METHODS Dextran sulfate sodium (DSS) was employed to induce barrier dysfunction in IECs. Inflammatory cytokines in supernatants (interleukin [IL]-6, IL-1β, tumor necrotic factor [TNF]-α, and IL-10) were quantified via enzyme-linked immunosorbent assay (ELISA). Then we assessed monolayer integrity using transepithelial electrical resistance (TEER). TJ protein expression (zonula occludens [ZO]-1 and Occludin) in IECs was evaluated through immunofluorescence and Western blot analysis. Network pharmacology helped identify the biological processes, signaling pathways, and key targets involved in Res's mitigation of DSS-induced IECs barrier dysfunction. The efficacy of the primary target was further corroborated using Western blot. RESULTS Res was shown to increase cell viability and IL-10 expression while reducing TNF-α, IL-6, and IL-1β levels, thus mitigating the inflammatory response. It enhanced TEER values and upregulated TJ protein expression (ZO-1 and Occludin). Network pharmacology revealed that Res potentially targets the NFE2L2 (nuclear factor erythroid-2-related factor 2, Nrf2), a vital antioxidant factor. Significantly, Res augmented Nrf2 and heme oxygenase 1 (HO-1) protein levels, counteracting oxidative stress in the IECs barrier dysfunction model. CONCLUSION Overall, our findings suggested that Res ameliorated DSS-induced IECs barrier dysfunction by activating Nrf2/HO-1 pathway, showcasing significant therapeutic potential in the early stages of colitis.
Collapse
Affiliation(s)
- Xinya Yu
- Department of Medical Microbiology and Immunology, School of Basic Medical SciencesDali UniversityDaliYunnanPeople's Republic of China
| | - Yazhi Wang
- Department of Medical Microbiology and Immunology, School of Basic Medical SciencesDali UniversityDaliYunnanPeople's Republic of China
| | - Yunchun Xu
- Department of Medical Microbiology and Immunology, School of Basic Medical SciencesDali UniversityDaliYunnanPeople's Republic of China
| | - Xiaoxi Li
- Department of General Surgery, School of Clinical MedicineDali UniversityDaliYunnanPeople's Republic of China
| | - Junhua Zhang
- Department of General Surgery, School of Clinical MedicineDali UniversityDaliYunnanPeople's Republic of China
| | - Yunpeng Su
- Department of Medical Microbiology and Immunology, School of Basic Medical SciencesDali UniversityDaliYunnanPeople's Republic of China
| | - Le Guo
- Department of Medical Microbiology and Immunology, School of Basic Medical SciencesDali UniversityDaliYunnanPeople's Republic of China
| |
Collapse
|
12
|
Wen C, Wang J, Sun Z, Zhong R, Li M, Shen X, Ye Q, Qin K, Peng X. Dietary Zinc Ameliorates TNBS-Induced Colitis in Mice Associated with Regulation of Th1/Th2/Th17 Balance and NF-κB/NLRP3 Signaling Pathway. Biol Trace Elem Res 2024; 202:659-670. [PMID: 37249802 DOI: 10.1007/s12011-023-03715-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, are chronic relapsing inflammatory gastrointestinal tract diseases of uncertain origin, which are frequently associated with zinc deficiency. Animal models have a considerable value in elucidating the process of IBD. In this study, 50 male C57BL/6 J mice were randomly assigned to five groups: control group (Con), 2,4,6-trinitrobenzenesulfonic acid (TNBS) group, and three zinc supplementation groups, namely 160 ppm group, 400 ppm group, and 1000 ppm group. The results showed that supplementation of dietary zinc with zinc oxide could effectively relieve the severity of ulcerative colitis induced by TNBS in mice. We demonstrate that the protective mechanism involves the immunomodulation of dietary zinc by increasing CD3+, CD3+CD8+, and Th2 cells, suppressing Th1 and Th17 cells, and decreasing the production of serum IL-1β and IL-18. The dietary zinc oxide seems to be able to suppress the NF-κB/NLRP3 signaling pathway by downregulating the mRNA and protein expression of NIK, IKK, NF-κB, and NLRP3. The results suggest that dietary supplementation of zinc oxide may protect against colitis, and proper daily zinc supplementation may reduce the risk of IBD.
Collapse
Affiliation(s)
- Changlin Wen
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Jiayu Wang
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Zhenhua Sun
- Department of Technology, Sichuan Youngster Technology Co., Ltd., No. 733, Furong Avenue, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Rao Zhong
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China
| | - Mengjie Li
- Department of Technology, Sichuan Youngster Technology Co., Ltd., No. 733, Furong Avenue, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Xuemei Shen
- Department of Technology, Sichuan Youngster Technology Co., Ltd., No. 733, Furong Avenue, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Qiaobo Ye
- School of Basic Medicine Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Kaihua Qin
- School of Basic Medicine Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Xi Peng
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, Sichuan, China.
| |
Collapse
|
13
|
Qiu C, Zhang JZ, Wu B, Xu CC, Pang HH, Tu QC, Lu YQ, Guo QY, Xia F, Wang JG. Advanced application of nanotechnology in active constituents of Traditional Chinese Medicines. J Nanobiotechnology 2023; 21:456. [PMID: 38017573 PMCID: PMC10685519 DOI: 10.1186/s12951-023-02165-x] [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: 09/01/2023] [Accepted: 10/16/2023] [Indexed: 11/30/2023] Open
Abstract
Traditional Chinese Medicines (TCMs) have been used for centuries for the treatment and management of various diseases. However, their effective delivery to targeted sites may be a major challenge due to their poor water solubility, low bioavailability, and potential toxicity. Nanocarriers, such as liposomes, polymeric nanoparticles, inorganic nanoparticles and organic/inorganic nanohybrids based on active constituents from TCMs have been extensively studied as a promising strategy to improve the delivery of active constituents from TCMs to achieve a higher therapeutic effect with fewer side effects compared to conventional formulations. This review summarizes the recent advances in nanocarrier-based delivery systems for various types of active constituents of TCMs, including terpenoids, polyphenols, alkaloids, flavonoids, and quinones, from different natural sources. This review covers the design and preparation of nanocarriers, their characterization, and in vitro/vivo evaluations. Additionally, this review highlights the challenges and opportunities in the field and suggests future directions for research. Nanocarrier-based delivery systems have shown great potential in improving the therapeutic efficacy of TCMs, and this review may serve as a comprehensive resource to researchers in this field.
Collapse
Affiliation(s)
- Chong Qiu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun Zhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Bo Wu
- Department of Traditional Chinese Medical Science, Sixth Medical Center of the Chinese PLA General Hospital, Beijing, 100037, China
| | - Cheng Chao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huan Huan Pang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qing Chao Tu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yu Qian Lu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qiu Yan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Fei Xia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ji Gang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-Di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
14
|
Marangoni K, Dorneles G, da Silva DM, Pinto LP, Rossoni C, Fernandes SA. Diet as an epigenetic factor in inflammatory bowel disease. World J Gastroenterol 2023; 29:5618-5629. [PMID: 38077158 PMCID: PMC10701328 DOI: 10.3748/wjg.v29.i41.5618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/24/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) has as a main characteristic the exacerbation of the immune system against enterocytes, compromising the individual's intestinal microbiota. This inflammatory cascade causes several nutritional deficiencies, which further compromise immunological functioning and, as a result, worsen the prognosis. This vicious cycle can be interrupted as the patient's dietary pattern meets their needs according to their clinical condition, acting directly on the inflammatory process of IBD through the interaction of food, intestinal microbiota, and epigenome. Specific nutritional intervention for IBD has a crucial role in preventing and managing disease activity. This review addresses epigenetic modifications through dietary compounds as a mechanism for modulating the intestinal microbiota of patients with IBD.
Collapse
Affiliation(s)
- Karina Marangoni
- Egas Moniz School of Health and Science, Caparica - Almada, Portugal, Caparica 2820-062, Portugal
- National Institute of Sciences and Technology - Theranostics and Nanobiotechnology, Federal University of Uberlandia - MG, Brazil, Uberlândia 38400-902, Brazil
| | - Gilson Dorneles
- Corporate Social Responsibility, Hospital Moinhos de Vento, Porto Alegre 90035-004, Brazil
| | - Daniella Miranda da Silva
- Postgraduate Program in Gastroenterology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91540-000, Brazil
- Department of Nutrition, Uniasselvi - Group Vitru, Santa Catarina 89082-262, Brazil
| | - Letícia Pereira Pinto
- Postgraduate Program in Hepatology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre 90050-170, Brazil
| | - Carina Rossoni
- Faculty of Medicine, Institute of Environmental Health, University of Lisbon, Lisboa 1649-026, Portugal
- Master in Physical Activity and Health, Polytechnic Institute of Beja, Beja 7800-000, Portugal
- Degree in Nutrition Sciences, Lusófona University, Lisboa 1749-024, Portugal
| | - Sabrina Alves Fernandes
- Postgraduate Program in Hepatology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre 90050-170, Brazil
| |
Collapse
|
15
|
Datta N, Johnson C, Kao D, Gurnani P, Alexander C, Polytarchou C, Monaghan TM. MicroRNA-based therapeutics for inflammatory disorders of the microbiota-gut-brain axis. Pharmacol Res 2023; 194:106870. [PMID: 37499702 DOI: 10.1016/j.phrs.2023.106870] [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: 03/11/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
An emerging but less explored shared pathophysiology across microbiota-gut-brain axis disorders is aberrant miRNA expression, which may represent novel therapeutic targets. miRNAs are small, endogenous non-coding RNAs that are important transcriptional repressors of gene expression. Most importantly, they regulate the integrity of the intestinal epithelial and blood-brain barriers and serve as an important communication channel between the gut microbiome and the host. A well-defined understanding of the mode of action, therapeutic strategies and delivery mechanisms of miRNAs is pivotal in translating the clinical applications of miRNA-based therapeutics. Accumulating evidence links disorders of the microbiota-gut-brain axis with a compromised gut-blood-brain-barrier, causing gut contents such as immune cells and microbiota to enter the bloodstream leading to low-grade systemic inflammation. This has the potential to affect all organs, including the brain, causing central inflammation and the development of neurodegenerative and neuropsychiatric diseases. In this review, we have examined in detail miRNA biogenesis, strategies for therapeutic application, delivery mechanisms, as well as their pathophysiology and clinical applications in inflammatory gut-brain disorders. The research data in this review was drawn from the following databases: PubMed, Google Scholar, and Clinicaltrials.gov. With increasing evidence of the pathophysiological importance for miRNAs in microbiota-gut-brain axis disorders, therapeutic targeting of cross-regulated miRNAs in these disorders displays potentially transformative and translational potential. Further preclinical research and human clinical trials are required to further advance this area of research.
Collapse
Affiliation(s)
- Neha Datta
- School of Medicine, University of Nottingham, Nottingham, UK
| | - Charlotte Johnson
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Pratik Gurnani
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Christos Polytarchou
- Department of Biosciences, John van Geest Cancer Research Centre, School of Science & Technology, Nottingham Trent University, Nottingham, UK.
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
16
|
Brockmueller A, Buhrmann C, Shayan P, Shakibaei M. Resveratrol induces apoptosis by modulating the reciprocal crosstalk between p53 and Sirt-1 in the CRC tumor microenvironment. Front Immunol 2023; 14:1225530. [PMID: 37575245 PMCID: PMC10413256 DOI: 10.3389/fimmu.2023.1225530] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction P53 represents a key player in apoptosis-induction in cancers including colorectal cancer (CRC) that ranks third worldwide in cancer prevalence as well as mortality statistics. Although a pro-apoptotic effect of resveratrol has been repeatedly proven in CRC cells, its pathway mechanisms are not completely understood, as there are controversial statements in the literature regarding its activation or inhibition of the counteracting proteins Sirt-1 and p53. Methods CRC cells as wild-type (HCT-116 WT) or p53-deficient (HCT-116 p53-/-) were cultured using multicellular tumor microenvironment (TME) cultures containing T-lymphocytes and fibroblasts to elucidate the role of p53/Sirt-1 modulation in resveratrol's concentration-dependent, pro-apoptotic, and thus anti-cancer effects. Results Resveratrol dose-dependently inhibited viability, proliferation, plasticity as well as migration, and induced apoptosis in HCT-116 WT more effectively than in HCT-116 p53-/- cells. Moreover, resveratrol stimulated Sirt-1 expression when administered at low concentrations (<5µM) but suppressed it when added at high concentrations (>10µM) to CRC-TME. In parallel, similar to the knockdown of Sirt-1 at the mRNA level, treatment with high-concentration resveratrol boosted the acetylation of p53, the expression of p21, Bax, cytochrome C, caspase-3, and ultimately induced apoptosis in CRC WT but not in CRC p53-/- cells. Notably, increasing concentrations of resveratrol were found to promote hyperacetylation of p53 and FOXO3a as post-translational substrates of Sirt-1, indicating a negative regulatory loop between Sirt-1 and p53. Discussion These results demonstrate for the first time, a negative reciprocal crosstalk between the regulatory circuits of p53 and Sirt-1, consequently, apoptosis induction by higher resveratrol concentrations in CRC-TME.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Constanze Buhrmann
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Parviz Shayan
- Department of Parasitology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
17
|
Lv J, Zhang R, Li D, Liu Y. Resveratrol plays an anti-fibrotic and anti-autophagy role by stimulating miR-192-5p expression in urethral fibrosis. Funct Integr Genomics 2023; 23:241. [PMID: 37450096 DOI: 10.1007/s10142-023-01173-2] [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: 03/14/2023] [Revised: 06/25/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Resveratrol (RSV) exerts anti-fibrotic effects on various fibrotic diseases. Whereas the biological role of RSV on urethral fibrosis remains to be elucidated. This study aimed to determine the mechanisms by which RSV affects urethral fibrosis and autophagy. METHODS Sprague‒Dawley rats and primary fibroblasts were treated with transforming growth factor-β1 (TGFβ1) to generate in vivo and in vitro fibrosis models. Then, those were treated with RSV, and autophagy and fibrosis-related indicators were tested. RESULTS Firstly, we found that RSV reversed the upregulation of indicators related to TGFβ1-induced fibrosis (TGFβ1, α-smooth muscle actin, collagen type I, and collagen type III), autophagy (TFEB and LC3), and TGFβR1/Smad4 pathway, as well as the downregulation of p62 and miR-192-5p expression both in vivo and in vitro. Overexpression of miR-192-5p suppressed the upregulation of fibrosis-related markers expression, as well as TFEB and LC3 expression, induced by TGFβ1, while the expression trend of p62 was the opposite. Inhibiting miR-192-5p reversed the effects of RSV on the model group cells. It was also shown that RSV combined with sh-Smad4 inhibited autophagy more effectively than RSV alone. CONCLUSION These results suggest that RSV inhibits urinary fibrosis and autophagy via the miR-192-5p/TGFβR1/Smad4 pathway. RAV may be a potential drug for alleviating urethral fibrosis.
Collapse
Affiliation(s)
- Jin Lv
- Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - Rui Zhang
- Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - DaoYuan Li
- Department of Urology, Hainan Afliated Hospital of Hainan Medical University, Haikou, China
- Department of Urology, Hainan General Hospital, Haikou, China
| | - Yan Liu
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
18
|
Airola C, Severino A, Porcari S, Fusco W, Mullish BH, Gasbarrini A, Cammarota G, Ponziani FR, Ianiro G. Future Modulation of Gut Microbiota: From Eubiotics to FMT, Engineered Bacteria, and Phage Therapy. Antibiotics (Basel) 2023; 12:antibiotics12050868. [PMID: 37237771 DOI: 10.3390/antibiotics12050868] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The human gut is inhabited by a multitude of bacteria, yeasts, and viruses. A dynamic balance among these microorganisms is associated with the well-being of the human being, and a large body of evidence supports a role of dysbiosis in the pathogenesis of several diseases. Given the importance of the gut microbiota in the preservation of human health, probiotics, prebiotics, synbiotics, and postbiotics have been classically used as strategies to modulate the gut microbiota and achieve beneficial effects for the host. Nonetheless, several molecules not typically included in these categories have demonstrated a role in restoring the equilibrium among the components of the gut microbiota. Among these, rifaximin, as well as other antimicrobial drugs, such as triclosan, or natural compounds (including evodiamine and polyphenols) have common pleiotropic characteristics. On one hand, they suppress the growth of dangerous bacteria while promoting beneficial bacteria in the gut microbiota. On the other hand, they contribute to the regulation of the immune response in the case of dysbiosis by directly influencing the immune system and epithelial cells or by inducing the gut bacteria to produce immune-modulatory compounds, such as short-chain fatty acids. Fecal microbiota transplantation (FMT) has also been investigated as a procedure to restore the equilibrium of the gut microbiota and has shown benefits in many diseases, including inflammatory bowel disease, chronic liver disorders, and extraintestinal autoimmune conditions. One of the most significant limits of the current techniques used to modulate the gut microbiota is the lack of tools that can precisely modulate specific members of complex microbial communities. Novel approaches, including the use of engineered probiotic bacteria or bacteriophage-based therapy, have recently appeared as promising strategies to provide targeted and tailored therapeutic modulation of the gut microbiota, but their role in clinical practice has yet to be clarified. The aim of this review is to discuss the most recently introduced innovations in the field of therapeutic microbiome modulation.
Collapse
Affiliation(s)
- Carlo Airola
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Severino
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Serena Porcari
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - William Fusco
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London W2 1NY, UK
- Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Antonio Gasbarrini
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
19
|
Lv L, Chen Z, Bai W, Hao J, Heng Z, Meng C, Wang L, Luo X, Wang X, Cao Y, He J. Taurohyodeoxycholic acid alleviates trinitrobenzene sulfonic acid induced ulcerative colitis via regulating Th1/Th2 and Th17/Treg cells balance. Life Sci 2023; 318:121501. [PMID: 36801213 DOI: 10.1016/j.lfs.2023.121501] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/21/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
AIMS Taurohyodeoxycholic acid (THDCA), a natural 6α-hydroxylated bile acid, exhibits intestinal anti-inflammatory effects. This study aimed to explore the efficacy of THDCA on ulcerative colitis and to reveal its mechanisms of action. MAIN METHODS Colitis was induced by intrarectal administration of trinitrobenzene sulfonic acid (TNBS) to mice. Mice in the treatment group were gavage THDCA (20, 40, and 80 mg/kg/day) or sulfasalazine (500 mg/kg/day) or azathioprine (10 mg/kg/day). The pathologic markers of colitis were comprehensively assessed. The levels of Th1-/Th2-/Th17-/Treg-related inflammatory cytokines and transcription factors were detected by ELISA, RT-PCR, and Western blotting. The balance of Th1/Th2 and Th17/Treg cells was analyzed by Flow cytometry. KEY FINDINGS THDCA significantly alleviated colitis by improving the body weight, colon length, spleen weight, histological characteristics, and MPO activity of colitis mice. THDCA reduced the secretion of Th1-/Th17-related cytokines (IFN-γ, IL-12p70, IL-6, IL-17A, IL-21, IL-22, and TNF-α) and the expressions of transcription factors (T-bet, STAT4, RORγt, and STAT3), but increase the production of Th2-/Treg-related cytokines (IL-4, IL-10, and TGF-β1) and the expressions of transcription factors (GATA3, STAT6, Foxp3, and Smad3) in the colon. Meanwhile, THDCA inhibited the expressions of IFN-γ, IL-17A, T-bet, and RORγt, but improved the expression of IL-4, IL-10, GATA3, and Foxp3 in the spleen. Furthermore, THDCA restored the proportion of Th1, Th2, Th17, and Treg cells, and balanced the Th1/Th2 and Th17/Treg immune response of colitis mice. SIGNIFICANCE THDCA can alleviate TNBS-induced colitis via regulating Th1/Th2 and Th17/Treg balance, which may represent a promising treatment for patients with colitis.
Collapse
Affiliation(s)
- Le Lv
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Ziyang Chen
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Wenhui Bai
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Jiahui Hao
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Zhengang Heng
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Caijin Meng
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Lin Wang
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Xianglan Luo
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Xinmiao Wang
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China
| | - Yanjun Cao
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, China
| | - Jiao He
- Biomedicine Key Laboratory of Shaanxi Province, College of Life Sciences, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, China.
| |
Collapse
|
20
|
Alghetaa H, Mohammed A, Singh N, Wilson K, Cai G, Putluri N, Nagarkatti M, Nagarkatti P. Resveratrol attenuates staphylococcal enterotoxin B-activated immune cell metabolism via upregulation of miR-100 and suppression of mTOR signaling pathway. Front Pharmacol 2023; 14:1106733. [PMID: 36909201 PMCID: PMC9999031 DOI: 10.3389/fphar.2023.1106733] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is triggered by a variety of insults, such as bacterial and viral infections, including SARS-CoV-2, leading to high mortality. In the murine model of ARDS induced by Staphylococcal enterotoxin-B (SEB), our previous studies showed that while SEB triggered 100% mortality, treatment with Resveratrol (RES) completely prevented such mortality by attenuating inflammation in the lungs. In the current study, we investigated the metabolic profile of SEB-activated immune cells in the lungs following treatment with RES. RES-treated mice had higher expression of miR-100 in the lung mononuclear cells (MNCs), which targeted mTOR, leading to its decreased expression. Also, Single-cell RNA-seq (scRNA seq) unveiled the decreased expression of mTOR in a variety of immune cells in the lungs. There was also an increase in glycolytic and mitochondrial respiration in the cells from SEB + VEH group in comparison with SEB + RES group. Together these data suggested that RES alters the metabolic reprogramming of SEB-activated immune cells, through suppression of mTOR activation and its down- and upstream effects on energy metabolism. Also, miR-100 could serve as novel potential therapeutic molecule in the amelioration of ARDS.
Collapse
Affiliation(s)
- Hasan Alghetaa
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Amira Mohammed
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Baghdad, Baghdad, Iraq
| | - Narendra Singh
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kiesha Wilson
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Goushuai Cai
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States
| | - Nagireddy Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
21
|
Chuang YT, Tang JY, Shiau JP, Yen CY, Chang FR, Yang KH, Hou MF, Farooqi AA, Chang HW. Modulating Effects of Cancer-Derived Exosomal miRNAs and Exosomal Processing by Natural Products. Cancers (Basel) 2023; 15:318. [PMID: 36612314 PMCID: PMC9818271 DOI: 10.3390/cancers15010318] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer-derived exosomes exhibit sophisticated functions, such as proliferation, apoptosis, migration, resistance, and tumor microenvironment changes. Several clinical drugs modulate these exosome functions, but the impacts of natural products are not well understood. Exosome functions are regulated by exosome processing, such as secretion and assembly. The modulation of these exosome-processing genes can exert the anticancer and precancer effects of cancer-derived exosomes. This review focuses on the cancer-derived exosomal miRNAs that regulate exosome processing, acting on the natural-product-modulating cell functions of cancer cells. However, the role of exosomal processing has been overlooked in several studies of exosomal miRNAs and natural products. In this study, utilizing the bioinformatics database (miRDB), the exosome-processing genes of natural-product-modulated exosomal miRNAs were predicted. Consequently, several natural drugs that modulate exosome processing and exosomal miRNAs and regulate cancer cell functions are described here. This review sheds light on and improves our understanding of the modulating effects of exosomal miRNAs and their potential exosomal processing targets on anticancer treatments based on the use of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Hsueh-Wei Chang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
22
|
Su T, Zhang Z, Han X, Yang F, Wang Z, Cheng Y, Liu H. Systematic Insight of Resveratrol Activated SIRT1 Interactome through Proximity Labeling Strategy. Antioxidants (Basel) 2022; 11:antiox11122330. [PMID: 36552538 PMCID: PMC9774693 DOI: 10.3390/antiox11122330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022] Open
Abstract
SIRT1 functions by regulating the modification of proteins or interacting with other proteins to form complexes. It has been widely studied and found to play significant roles in various biological processes and diseases. However, systematic studies on activated-SIRT1 interactions remain limited. Here, we present a comprehensive SIRT1 interactome under resveratrol stimulation through proximity labeling methods. Our results demonstrated that RanGap1 interacted with SIRT1 in HEK 293T cells and MCF-7 cells. SIRT1 regulated the protein level of RanGap1 and had no obvious effect on RanGap1 transcription. Moreover, the overexpression of Rangap1 increased the ROS level in MCF-7 cells, which sensitized cells to resveratrol and reduced the cell viability. These findings provide evidence that RanGap1 interacts with SIRT1 and influences intracellular ROS, critical signals for mitochondrial functions, cell proliferation and transcription. Additionally, we identified that the SIRT1-RanGap1 interaction affects downstream signals induced by ROS. Overall, our study provides an essential resource for future studies on the interactions of resveratrol-activated SIRT1. There are conflicts about the relationship between resveratrol and ROS in previous reports. However, our data identified the impact of the resveratrol-SIRT1-RanGap1 axis on intracellular ROS.
Collapse
Affiliation(s)
- Tian Su
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zhengyi Zhang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xiao Han
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Fei Yang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zhen Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Ying Cheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Correspondence: (Y.C.); (H.L.)
| | - Huadong Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
- Correspondence: (Y.C.); (H.L.)
| |
Collapse
|
23
|
Vaghari-Tabari M, Alemi F, Zokaei M, Moein S, Qujeq D, Yousefi B, Farzami P, Hosseininasab SS. Polyphenols and inflammatory bowel disease: Natural products with therapeutic effects? Crit Rev Food Sci Nutr 2022; 64:4155-4178. [PMID: 36345891 DOI: 10.1080/10408398.2022.2139222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Inflammatory bowel disease (IBD) is a long-life disease with periods of recurrence and relief. Oxidative stress plays an important role in the pathogenesis of this disease. Recent years' studies in the field of IBD treatment mostly have focused on targeting cytokines and immune cell trafficking using antibodies and inhibitors, altering the composition of intestinal bacteria in the line of attenuation of inflammation using probiotics and prebiotics, and attenuating oxidative stress through antioxidant supplementation. Studies in animal models of IBD have shown that some polyphenolic compounds including curcumin, quercetin, resveratrol, naringenin, and epigallocatechin-3-gallate can affect almost all of the above aspects and are useful compounds in the treatment of IBD. Clinical studies performed on IBD patients have also confirmed the findings of animal model studies and have shown that supplementation with some of the above-mentioned polyphenolic compounds has positive effects in reducing disease clinical and endoscopic activity, inducing and maintaining remission, and improving quality of life. In this review article, in addition to a detailed reviewing the effects of the above-mentioned polyphenolic compounds on the events involved in the pathogenesis of IBD, the results of these clinical studies will also be reviewed.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Soheila Moein
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Payam Farzami
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
24
|
Li Z, Lin M, Li Y, Shao J, Huang R, Qiu Y, Liu Y, Chen L. Total flavonoids of Sophora flavescens and kurarinone ameliorated ulcerative colitis by regulating Th17/Treg cell homeostasis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115500. [PMID: 35863614 DOI: 10.1016/j.jep.2022.115500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is relevant to dysregulation of inflammation and immune processes. Sophora flavescens Aiton is a classic medicine widely used in the treatment of UC in ancient and modern China, alkaloids and flavonoids are the main components. Previous studies reveal that Sophora flavescens Aiton total flavonoids extracts (SFE) exert an anti-UC effect by regulating the intestinal microbe structure and restoring the balance of the "host-microbe" co-metabolic network in UC mice. However, whether SFE influences immune inflammation remains unclear, which is the core link to UC disease. It also remains to be verified flavonoids are the material basis that plays a role in SFE. AIM OF THE STUDY To identify the action mechanism of the immune-inflammatory regulation of SFE and its main active component Kurarinone against UC. METHODS This study constructed UC mice and abnormal immune RAW 264.7 cell models, and subsequently used western blotting and flow cytometry (FCM) to evaluate the effects of SFE on the NF-κB pathway and the regulation of immunity in UC mice. Kurarinone was screened from flavonoid compounds of SFE by lipopolysaccharide (LPS)-induced RAW 264.7 cells, and its effect was subsequently investigated in UC mice. Western blotting, ELISA, FCM, and RT-PCR were used to determine the regulation of Kurarinone on the Th17/Treg differentiation and the JAK2/STAT3 signaling pathway. RESULTS SFE regulated the differentiation of Th17/Treg in peripheral blood and inhibited immune-inflammatory response to treat UC. Various flavonoid components in SFE inhibited the synthesis of IL-6 and TNF-α in RAW 264.7 cells, among which Kurarinone had better effect. This study revealed the therapeutic effects of Kurarinone in UC mice for the first time. Kurarinone promoted the secretion of SIgA to improve the regulation of the intestinal mucosal barrier and resistance to pathogens. It also regulated the transcription level of RORγt and Foxp3 in colon, decreased the expression of pro-inflammatory factor IL-17A and up-regulated the expression of immunosuppressive factors TGF-β1 and IL-10 in colon. Furthermore, Kurarinone restored intestinal immune system homeostasis by down-regulating the JAK2/STAT3 signaling pathway and regulating the balance of Th17/Treg cell differentiation in UC. CONCLUSIONS SFE, especially the flavonoid ingredients represented by Kurarinone, has significant effects on immunoregulation against UC. And their mechanism of effect is related to inhibiting the activation of JAK2/STAT3 signaling pathway and regulating differentiation of Th17/Treg cells. KEYWORK Immunoregulatory; Kurarinone; Th17 cells; Treg cells; Ulcerative colitis.
Collapse
Affiliation(s)
- Zhaocheng Li
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Minling Lin
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yadi Li
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Shao
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ruiting Huang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongyi Qiu
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Lei Chen
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medical of State Administration of TCM, China, Engineering & Technology Research Center for Chines Materia Medical Quality of Guangdong Province, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
25
|
Caban M, Lewandowska U. Polyphenols and the potential mechanisms of their therapeutic benefits against inflammatory bowel diseases. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
26
|
Vieujean S, Caron B, Haghnejad V, Jouzeau JY, Netter P, Heba AC, Ndiaye NC, Moulin D, Barreto G, Danese S, Peyrin-Biroulet L. Impact of the Exposome on the Epigenome in Inflammatory Bowel Disease Patients and Animal Models. Int J Mol Sci 2022; 23:7611. [PMID: 35886959 PMCID: PMC9321337 DOI: 10.3390/ijms23147611] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that encompass two main phenotypes, namely Crohn's disease and ulcerative colitis. These conditions occur in genetically predisposed individuals in response to environmental factors. Epigenetics, acting by DNA methylation, post-translational histones modifications or by non-coding RNAs, could explain how the exposome (or all environmental influences over the life course, from conception to death) could influence the gene expression to contribute to intestinal inflammation. We performed a scoping search using Medline to identify all the elements of the exposome that may play a role in intestinal inflammation through epigenetic modifications, as well as the underlying mechanisms. The environmental factors epigenetically influencing the occurrence of intestinal inflammation are the maternal lifestyle (mainly diet, the occurrence of infection during pregnancy and smoking); breastfeeding; microbiota; diet (including a low-fiber diet, high-fat diet and deficiency in micronutrients); smoking habits, vitamin D and drugs (e.g., IBD treatments, antibiotics and probiotics). Influenced by both microbiota and diet, short-chain fatty acids are gut microbiota-derived metabolites resulting from the anaerobic fermentation of non-digestible dietary fibers, playing an epigenetically mediated role in the integrity of the epithelial barrier and in the defense against invading microorganisms. Although the impact of some environmental factors has been identified, the exposome-induced epimutations in IBD remain a largely underexplored field. How these environmental exposures induce epigenetic modifications (in terms of duration, frequency and the timing at which they occur) and how other environmental factors associated with IBD modulate epigenetics deserve to be further investigated.
Collapse
Affiliation(s)
- Sophie Vieujean
- Hepato-Gastroenterology and Digestive Oncology, University Hospital CHU of Liège, 4000 Liege, Belgium;
| | - Bénédicte Caron
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| | - Vincent Haghnejad
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| | - Jean-Yves Jouzeau
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Patrick Netter
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Anne-Charlotte Heba
- NGERE (Nutrition-Genetics and Exposure to Environmental Risks), National Institute of Health and Medical Research, University of Lorraine, F-54000 Nancy, France; (A.-C.H.); (N.C.N.)
| | - Ndeye Coumba Ndiaye
- NGERE (Nutrition-Genetics and Exposure to Environmental Risks), National Institute of Health and Medical Research, University of Lorraine, F-54000 Nancy, France; (A.-C.H.); (N.C.N.)
| | - David Moulin
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
| | - Guillermo Barreto
- CNRS (French National Centre for Scientific Research), Laboratoire IMoPA, Université de Lorraine, UMR 7365, F-54000 Nancy, France; (J.-Y.J.); (P.N.); (D.M.); (G.B.)
- Lung Cancer Epigenetics, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Universidad de la Salud del Estado de Puebla, Puebla 72000, Mexico
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, 20132 Milan, Italy;
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology NGERE (INSERM U1256), Nancy University Hospital, University of Lorraine, Vandœuvre-lès-Nancy, F-54052 Nancy, France; (B.C.); (V.H.)
| |
Collapse
|
27
|
Resveratrol and resveratrol nano-delivery systems in the treatment of inflammatory bowel disease. J Nutr Biochem 2022; 109:109101. [PMID: 35777588 DOI: 10.1016/j.jnutbio.2022.109101] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 04/04/2022] [Accepted: 06/08/2022] [Indexed: 12/22/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder associated with the inflammation in the digestive tract. The exact cause of IBD is unknown; nevertheless, in IBD, the homeostasis of key regulatory factors involved in intestinal immunity has been documented to be disrupted. Despite the lack of a viable treatment for IBD, synthetic drugs and monoclonal antibodies are currently used to treat it. However, these treatments have side effects, and the high relapse rate limits their usage. Dietary polyphenols constitute a great variety of compounds and have shown an array of biological properties. Resveratrol is a natural polyphenol found in grapevines and berries. The therapeutic ability of resveratrol against IBD is amply demonstrated in many in vivo studies. Resveratrol can interact with several molecular targets (Nf-kB, SIRT1, mTOR, HIF-1α, miRNAs, and TNF-α) and effectively prevent/ alleviate IBD symptoms with promising results. Although resveratrol has profound anti-inflammatory properties against IBD, its therapeutic employment is limited due to its low water solubility, less chemical stability, less bioavailability, and rapid metabolism in vivo. Hence, resveratrol encapsulation using different carries and its controlled release has become a promising strategy to overcome limitations. Herein, we meticulously review, talk-over the anti-inflammatory effect and mechanisms of resveratrol in IBD. We further provide the latest information on resveratrol formulations and nano-delivery systems used in oral delivery of resveratrol for the treatment of IBD and offer our view on future research on resveratrol in IBD treatment.
Collapse
|
28
|
Xiao X, Mao X, Chen D, Yu B, He J, Yan H, Wang J. miRNAs Can Affect Intestinal Epithelial Barrier in Inflammatory Bowel Disease. Front Immunol 2022; 13:868229. [PMID: 35493445 PMCID: PMC9043318 DOI: 10.3389/fimmu.2022.868229] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/18/2022] [Indexed: 11/17/2022] Open
Abstract
The most obvious pathological characterization of inflammatory bowel disease (IBD) is intestinal epithelium erosion and severe inflammation invasion. Micro-ribonucleic acids (miRNA or microRNA), single-stranded noncoding RNAs of ~22 nucleotides, have been considered as the potential therapeutic targets in the pathogenesis of IBD. Many previous studies have focused on the mechanisms that miRNAs use to regulate inflammation, immunity, and microorganisms in IBD. The review highlights in detail the findings of miRNAs in the intestinal epithelial barrier of IBD, and focuses on their gene targets, signaling pathways associated with IBD, and some potential therapies. It will be beneficial for the elucidation of the interaction between miRNAs and the intestinal epithelial barrier in IBD and provide a theoretical reference for preventing and treating IBD in the future.
Collapse
Affiliation(s)
- Xiangjun Xiao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| | - Jianping Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Key Laboratory of Animal Disease-Resistant Nutrition and Feed of China Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Chengdu, China
| |
Collapse
|
29
|
Li M, Li P, Tang R, Lu H. Resveratrol and its derivates improve inflammatory bowel disease by targeting gut microbiota and inflammatory signaling pathways. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Xie Z, Chen X. Healthy benefits and edible delivery systems of resveratrol: a review. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.2013873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Zhenfeng Xie
- State Key Laboratory of Food Science and Technology, Jiangnan University, 214122, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 214122, Wuxi, Jiangsu, China
| | - Xing Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, 214122, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 214122, Wuxi, Jiangsu, China
| |
Collapse
|
31
|
Roberts LB, Kapoor P, Howard JK, Shah AM, Lord GM. An update on the roles of immune system-derived microRNAs in cardiovascular diseases. Cardiovasc Res 2021; 117:2434-2449. [PMID: 33483751 PMCID: PMC8562329 DOI: 10.1093/cvr/cvab007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of human death worldwide. Over the past two decades, the emerging field of cardioimmunology has demonstrated how cells of the immune system play vital roles in the pathogenesis of CVD. MicroRNAs (miRNAs) are critical regulators of cellular identity and function. Cell-intrinsic, as well as cell-extrinsic, roles of immune and inflammatory cell-derived miRNAs have been, and continue to be, extensively studied. Several 'immuno-miRNAs' appear to be specifically expressed or demonstrate greatly enriched expression within leucocytes. Identification of miRNAs as critical regulators of immune system signalling pathways has posed the question of whether and how targeting these molecules therapeutically, may afford opportunities for disease treatment and/or management. As the field of cardioimmunology rapidly continues to advance, this review discusses findings from recent human and murine studies which contribute to our understanding of how leucocytes of innate and adaptive immunity are regulated-and may also regulate other cell types, via the actions of the miRNAs they express, in the context of CVD. Finally, we focus on available information regarding miRNA regulation of regulatory T cells and argue that targeted manipulation of miRNA regulated pathways in these cells may hold therapeutic promise for the treatment of CVD and associated risk factors.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Puja Kapoor
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jane K Howard
- School of Life Course Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- Faculty of Biology, Medicine and Health, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
32
|
Targeting miRNA by Natural Products: A Novel Therapeutic Approach for Nonalcoholic Fatty Liver. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6641031. [PMID: 34426744 PMCID: PMC8380168 DOI: 10.1155/2021/6641031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 08/02/2021] [Indexed: 02/07/2023]
Abstract
The increasing prevalence of nonalcoholic fatty liver disease (NAFLD) as multifactorial chronic liver disease and the lack of a specific treatment have begun a new era in its treatment using gene expression changes and microRNAs. This study aimed to investigate the potential therapeutic effects of natural compounds in NAFLD by regulating miRNA expression. MicroRNAs play essential roles in regulating the cell's biological processes, such as apoptosis, migration, lipid metabolism, insulin resistance, and adipocyte differentiation, by controlling the posttranscriptional gene expression level. The impact of current NAFLD pharmacological management, including drug and biological therapies, is uncertain. In this context, various dietary fruits or medicinal herbal sources have received worldwide attention versus NAFLD development. Natural ingredients such as berberine, lychee pulp, grape seed, and rosemary possess protective and therapeutic effects against NAFLD by modifying the gene's expression and noncoding RNAs, especially miRNAs.
Collapse
|
33
|
Nunes AM, Ramirez M, Jones TI, Jones PL. Identification of candidate miRNA biomarkers for facioscapulohumeral muscular dystrophy using DUX4-based mouse models. Dis Model Mech 2021; 14:dmm049016. [PMID: 34338285 PMCID: PMC8405850 DOI: 10.1242/dmm.049016] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by misexpression of DUX4 in skeletal myocytes. As DUX4 is the key therapeutic target in FSHD, surrogate biomarkers of DUX4 expression in skeletal muscle are critically needed for clinical trials. Although no natural animal models of FSHD exist, transgenic mice with inducible DUX4 expression in skeletal muscles rapidly develop myopathic phenotypes consistent with FSHD. Here, we established a new, more-accurate FSHD-like mouse model based on chronic DUX4 expression in a small fraction of skeletal myonuclei that develops pathology mimicking key aspects of FSHD across its lifespan. Utilizing this new aged mouse model and DUX4-inducible mouse models, we characterized the DUX4-related microRNA signatures in skeletal muscles, which represent potential biomarkers for FSHD. We found increased expression of miR-31-5p and miR-206 in muscles expressing different levels of DUX4 and displaying varying degrees of pathology. Importantly, miR-206 expression is significantly increased in serum samples from FSHD patients compared with healthy controls. Our data support miR-31-5p and miR-206 as new potential regulators of muscle pathology and miR-206 as a potential circulating biomarker for FSHD. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | - Takako I. Jones
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Peter L. Jones
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| |
Collapse
|
34
|
Chang Y, Zhai L, Peng J, Wu H, Bian Z, Xiao H. Phytochemicals as regulators of Th17/Treg balance in inflammatory bowel diseases. Biomed Pharmacother 2021; 141:111931. [PMID: 34328111 DOI: 10.1016/j.biopha.2021.111931] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/09/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disorder that is difficult to cure and characterized by periods of relapse. To face the challenges of limited treatment strategies and drawbacks of conventional medications, developing new and promising strategies as well as safe and effective drugs for treatment of IBD has become an urgent demand for clinics. The imbalance of Th17/Treg is a crucial event for the development of IBD, and studies have verified that correcting the imbalance of Th17/Treg is an effective strategy for preventing and treating IBD. Recently, a growing body of studies has indicated that phytochemicals derived from natural products are potent regulators of Th17/Treg, and exert preferable protective benefits against colonic inflammation. In this review, the great potential of anti-colitis agents derived from natural products through targeting Th17/Treg cells and their action mechanisms for the treatment or prevention of IBD in recent research is summarized, which may help further the development of new drugs for IBD treatment.
Collapse
Affiliation(s)
- Yaoyao Chang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Lixiang Zhai
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Jiao Peng
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, China
| | - Haiqiang Wu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
35
|
Ganesan K, Quiles JL, Daglia M, Xiao J, Xu B. Dietary phytochemicals modulate intestinal epithelial barrier dysfunction and autoimmune diseases. FOOD FRONTIERS 2021. [DOI: 10.1002/fft2.102] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Kumar Ganesan
- Food Science and Technology Program BNU–HKBU United International College Zhuhai China
- The School of Chinese Medicine The University of Hong Kong Hong Kong China
| | - José L. Quiles
- Institute of Nutrition and Food Technology “José Mataix Verdú,” Department of Physiology Biomedical Research Center University of Granada Granada Spain
| | - Maria Daglia
- Department of Pharmacy University of Naples Federico II Naples Italy
- International Research Center for Food Nutrition and Safety Jiangsu University Zhenjiang China
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology University of Vigo Vigo Pontevedra E‐36310 Spain
| | - Baojun Xu
- Food Science and Technology Program BNU–HKBU United International College Zhuhai China
| |
Collapse
|
36
|
Zhao Y, Luan H, Jiang H, Xu Y, Wu X, Zhang Y, Li R. Gegen Qinlian decoction relieved DSS-induced ulcerative colitis in mice by modulating Th17/Treg cell homeostasis via suppressing IL-6/JAK2/STAT3 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153519. [PMID: 33640781 DOI: 10.1016/j.phymed.2021.153519] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/05/2021] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Gegen Qinlian decoction (GQ) is a traditional Chinese herbal prescription that has been widely used for the treatment of bacterial dysentery and enteric typhoid fever. Recently, GQ has been clinically reported to be a potential candidate for the treatment of ulcerative colitis (UC). However, the immunoregulatory function of GQ in the treatment of UC has not been fully elucidated. PURPOSE This study focused on the role of immune imbalance in the pathogenesis of UC and the immunomodulatory effect of GQ in the treatment of UC. METHODS The UC model was established by treating female mice with 3.0% dextran sulfate sodium (DSS) for 7 days, and GQ was orally administered at dosages of 1.5 and 7.5 g/kg/day. Inflammatory factors were detected by ELISA and qRT-PCR. Treg and Th17 cell dysregulation was analyzed by qRT-PCR, immunohistochemistry and flow cytometry. Proteins related to IL-6/JAK2/STAT3 signaling were detected by western blotting. RESULTS GQ significantly alleviated the symptoms of UC mice and suppressed the activity of myeloperoxidase (MPO). Furthermore, the production of proinflammatory factors, such as IL-1β, TNF-α and IL-6, was dramatically reduced after GQ administration. Furthermore, GQ improved the infiltration of Treg and Th17 cells into the colons and decreased the expression of inflammatory factors, such as TGF-β1 and IL-17. The frequencies of Treg and Th17 cells in the Peyer's patches and spleen were reduced by GQ administration; however, GQ had no significant regulatory effect on normal mice. The western blotting results showed that GQ markedly suppressed the phosphorylation of JAK2 and STAT3 and decreased the transcription function of phosphorylated STAT3. CONCLUSIONS Taken together, these results indicated that GQ alleviated DSS-induced UC by suppressing IL-6/JAK2/STAT3 signaling to restore Treg and Th17 cell homeostasis in colonic tissue.
Collapse
Affiliation(s)
- Yaxing Zhao
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Haofan Luan
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Hua Jiang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Yingmei Xu
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Xiaojun Wu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yubin Zhang
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China.
| | - Ruiyan Li
- State Key Laboratory of Natural Medicines, Department of Biochemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China.
| |
Collapse
|
37
|
Li X, Zhang ZH, Zabed HM, Yun J, Zhang G, Qi X. An Insight into the Roles of Dietary Tryptophan and Its Metabolites in Intestinal Inflammation and Inflammatory Bowel Disease. Mol Nutr Food Res 2021; 65:e2000461. [PMID: 33216452 DOI: 10.1002/mnfr.202000461] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/14/2020] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel disease (IBD) is complex, chronic, and relapsing gastrointestinal inflammatory disorders, which includes mainly two conditions, namely ulcerative colitis (UC) and Crohn's disease (CD). Development of IBD in any individual is closely related to his/her autoimmune regulation, gene-microbiota interactions, and dietary factors. Dietary tryptophan (Trp) is an essential amino acid for intestinal mucosal cells, and it is associated with the intestinal inflammation, epithelial barrier, and energy homeostasis of the host. According to recent studies, Trp and its three major metabolic pathways, namely kynurenine (KYN) pathway, indole pathway, and 5-hydroxytryptamine (5-HT) pathway, have vital roles in the regulation of intestinal inflammation by acting directly or indirectly on the pro/anti-inflammatory cytokines, functions of various immune cells, as well as the intestinal microbial composition and homeostasis. In this review, recent advances in Trp- and its metabolites-associated intestinal inflammation are summarized. It further discusses the complex mechanisms and interrelationships of the three major metabolic pathways of Trp in regulating inflammation, which could elucidate the value of dietary Trp to be used as a nutrient for IBD patients.
Collapse
Affiliation(s)
- Xiaolan Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zhi-Hong Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Hossain M Zabed
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Junhua Yun
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Guoyan Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xianghui Qi
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
38
|
Zhou X, Zhang J, Li Y, Cui L, Wu K, Luo H. Astaxanthin inhibits microglia M1 activation against inflammatory injury triggered by lipopolysaccharide through down-regulating miR-31-5p. Life Sci 2021; 267:118943. [PMID: 33359248 DOI: 10.1016/j.lfs.2020.118943] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 12/06/2020] [Accepted: 12/15/2020] [Indexed: 01/31/2023]
Abstract
AIMS Astaxanthin is a natural carotenoid, can readily cross the blood-brain barrier and exerts a powerful neuroprotective effect. In this study, experiments were performed to explore the underlying molecular mechanisms of which Astaxanthin inhibiting the microglia M1 activation. MAIN METHODS BV2 cells and mice were pre-treated with Astaxanthin and treated by Lipopolysaccharide (LPS). The expressions of M1-related factors (pro-inflammatory cytokines and M1 markers) were measured by RT-qPCR and western blot. The target association between miR-31-5p and Numb was explored via luciferase activity assay. MiR-31-5p mimic was transfected into BV2 cells, then the cells were treated with Astaxanthin in combination with LPS. The expression of M1-related factors and Notch pathway-related molecules were measured via RT-qPCR, western blot and immunofluorescence assay. KEY FINDINGS Precondition of BV2 cells with Astaxanthin inhibited the expression of M1-related factors triggered by LPS. In addition, Astaxanthin decreased the number of Iba1-positive microglia and downregulated the levels of M1-related factors in hippocampus in LPS-treated mice. Further investigation revealed that Astaxanthin-mediated suppression of M1-related factors levels was reversed by miR-31-5p mimic in BV2 cells stimulated by LPS. Subsequently, we verified that miR-31-5p repressed Numb expression by binding to the 3'-UTR of Numb mRNA. Also, Astaxanthin suppressed the expression of Notch1, Hes1 and Hes5 and improved the expression of Numb in BV2 cells challenged by LPS, but this alteration can be reversed by miR-31-5p mimic. SIGNIFICANCE Our study demonstrated that down-regulating miR-31-5p by Astaxanthin could be a potential therapeutic approach to suppress neuroinflammation via regulating microglia M1 activation.
Collapse
Affiliation(s)
- Xin Zhou
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| | - Junyu Zhang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Yuxin Li
- Department of Pharmacology, Guangdong Medical University, Zhanjiang 524023, China
| | - Liao Cui
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, China
| | - Kefeng Wu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, China.
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China.
| |
Collapse
|
39
|
Huang MC, White KL, Elmore SA, Guo TL, Germolec D. Immunotoxicity studies of trans-resveratrol in male B 6C 3F 1/N mice. J Immunotoxicol 2020; 17:194-201. [PMID: 33213203 PMCID: PMC7961840 DOI: 10.1080/1547691x.2020.1833113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/21/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022] Open
Abstract
Resveratrol is a naturally occurring polyphenol that is being investigated to treat and prevent various diseases, both experimentally and in the clinic. Despite increased use and interest in resveratrol due to its immunomodulatory properties, there is a lack of studies evaluating potential toxicities, particularly immunotoxicity, associated with resveratrol use. A previous 2-week study found decreasing thymus weight in male B6C3F1/N mice with increasing exposure to trans-resveratrol. This study is a follow-up on those findings by evaluating immune function. Male adult B6C3F1/N mice were given trans-resveratrol (0, 156, 312, 625, 1250, 2500 mg/kg/day) via oral gavage for 28 days and functional immune tests and histopathology were evaluated. There were no treatment-related effects on body weight during the study. Humoral, cell-mediated, and innate immune function were not altered after 28 days of trans-resveratrol treatment. There were also no changes in organ weight or microscopic alterations in immune organs. Overall, under the conditions of this study, there was no evidence of immunotoxicity or improvements in immune function associated with oral exposure to trans-resveratrol in male mice. Importantly, the immunomodulatory benefits of resveratrol may require a prerequisite level of inflammatory activity and may not be observable in healthy individuals.
Collapse
Affiliation(s)
- Madelyn C Huang
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Kimber L White
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA (retired)
| | - Susan A Elmore
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Dori Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
40
|
Ashrafizadeh M, Najafi M, Orouei S, Zabolian A, Saleki H, Azami N, Sharifi N, Hushmandi K, Zarrabi A, Ahn KS. Resveratrol Modulates Transforming Growth Factor-Beta (TGF-β) Signaling Pathway for Disease Therapy: A New Insight into Its Pharmacological Activities. Biomedicines 2020; 8:E261. [PMID: 32752069 PMCID: PMC7460084 DOI: 10.3390/biomedicines8080261] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Resveratrol (Res) is a well-known natural product that can exhibit important pharmacological activities such as antioxidant, anti-diabetes, anti-tumor, and anti-inflammatory. An evaluation of its therapeutic effects demonstrates that this naturally occurring bioactive compound can target different molecular pathways to exert its pharmacological actions. Transforming growth factor-beta (TGF-β) is an important molecular pathway that is capable of regulating different cellular mechanisms such as proliferation, migration, and angiogenesis. TGF-β has been reported to be involved in the development of disorders such as diabetes, cancer, inflammatory disorders, fibrosis, cardiovascular disorders, etc. In the present review, the relationship between Res and TGF-β has been investigated. It was noticed that Res can inhibit TGF-β to suppress the proliferation and migration of cancer cells. In addition, Res can improve fibrosis by reducing inflammation via promoting TGF-β down-regulation. Res has been reported to be also beneficial in the amelioration of diabetic complications via targeting the TGF-β signaling pathway. These topics are discussed in detail in this review to shed light on the protective effects of Res mediated via the modulation of TGF-β signaling.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Sima Orouei
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Negar Azami
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Negin Sharifi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956 Istanbul, Turkey
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
41
|
Magrone T, Magrone M, Russo MA, Jirillo E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants (Basel) 2019; 9:E35. [PMID: 31906123 PMCID: PMC7022464 DOI: 10.3390/antiox9010035] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/20/2022] Open
Abstract
In this review, special emphasis will be placed on red grape polyphenols for their antioxidant and anti-inflammatory activities. Therefore, their capacity to inhibit major pathways responsible for activation of oxidative systems and expression and release of proinflammatory cytokines and chemokines will be discussed. Furthermore, regulation of immune cells by polyphenols will be illustrated with special reference to the activation of T regulatory cells which support a tolerogenic pathway at intestinal level. Additionally, the effects of red grape polyphenols will be analyzed in obesity, as a low-grade systemic inflammation. Also, possible modifications of inflammatory bowel disease biomarkers and clinical course have been studied upon polyphenol administration, either in animal models or in clinical trials. Moreover, the ability of polyphenols to cross the blood-brain barrier has been exploited to investigate their neuroprotective properties. In cancer, polyphenols seem to exert several beneficial effects, even if conflicting data are reported about their influence on T regulatory cells. Finally, the effects of polyphenols have been evaluated in experimental models of allergy and autoimmune diseases. Conclusively, red grape polyphenols are endowed with a great antioxidant and anti-inflammatory potential but some issues, such as polyphenol bioavailability, activity of metabolites, and interaction with microbiota, deserve deeper studies.
Collapse
Affiliation(s)
- Thea Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, 70124 Bari, Italy; (M.M.); (E.J.)
| | - Manrico Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, 70124 Bari, Italy; (M.M.); (E.J.)
| | - Matteo Antonio Russo
- MEBIC Consortium, San Raffaele Open University of Rome and IRCCS San Raffaele Pisana of Rome, 00166 Rome, Italy;
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, School of Medicine, University of Bari, 70124 Bari, Italy; (M.M.); (E.J.)
| |
Collapse
|