1
|
Lee MJ, Lee J, Ha J, Kim G, Kim HJ, Lee S, Koo BK, Park Y. Long-term three-dimensional high-resolution imaging of live unlabeled small intestinal organoids via low-coherence holotomography. Exp Mol Med 2024; 56:2162-2170. [PMID: 39349827 PMCID: PMC11541879 DOI: 10.1038/s12276-024-01312-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 10/03/2024] Open
Abstract
Organoids, which are miniature in vitro versions of organs, possess significant potential for studying human diseases and elucidating their underlying mechanisms. Live imaging techniques play a crucial role in organoid research and contribute to elucidating the complex structure and dynamic biological phenomena of organoids. However, live, unlabeled high-resolution imaging of native organoids is challenging, primarily owing to the complexities of sample handling and optical scattering inherent in three-dimensional (3D) structures. Additionally, conventional imaging methods fail to capture the real-time dynamic processes of growing organoids. In this study, we introduce low-coherence holotomography as an advanced, label-free, quantitative imaging modality designed to overcome several technical obstacles for long-term live imaging of 3D organoids. We demonstrate the efficacy of low-coherence holotomography by capturing high-resolution morphological details and dynamic activities within mouse small intestinal organoids at subcellular resolution. Moreover, our approach facilitates the distinction between viable and nonviable organoids, significantly enhancing its utility in organoid-based research. This advancement underscores the critical role of live imaging in organoid studies, offering a more comprehensive understanding of these complex systems.
Collapse
Affiliation(s)
- Mahn Jae Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea
| | | | - Jeongmin Ha
- Center for Genome Engineering, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Geon Kim
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea
- Department of Physics, KAIST, Daejeon, 34141, Republic of Korea
| | | | - Sumin Lee
- Tomocube Inc., Daejeon, Republic of Korea
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon, 34126, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
| | - YongKeun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- KAIST Institute for Health Science and Technology, Daejeon, 34141, Republic of Korea.
- Tomocube Inc., Daejeon, Republic of Korea.
- Department of Physics, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
2
|
Mao J, Tan L, Tian C, Wang W, Zou Y, Zhu Z, Li Y. Systematically investigate the mechanism underlying the therapeutic effect of Astragalus membranaceus in ulcerative colitis. Am J Med Sci 2024:S0002-9629(24)01355-7. [PMID: 39009282 DOI: 10.1016/j.amjms.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Whether Astragalus membranaceus is an effective drug in treatment of ulcerative colitis (UC) and how it exhibit activity effect on UC is unclear. METHODS TCMSP, GeneCards, String, and DAVID database were used to screening target genes construct PPI network and performed for GO and KEGG pathway enrichment analysis respectively. Molecular docking and animal experiment were performed. The body weight and disease activity index (DAI) of mice were recorded. ELISA kits were used to detect the levels of CAT, SOD, MDA and IL-6, IL-10, TNF-α in the blood of mice. Western blot kits were utilized to measured the expressions of MAPK14, RB1, MAPK1, JUN, ATK1, and IL2 proteins. RESULTS The active components of Astragalus membranaceus mainly including 7-O-methylisomucronulatol, quercetin, kaempferol, formononetin and isrhamnetin. Astragalus membranaceus may inhibited the expression of TNF-α, IL-6, MDA, and promoted the expression of CAT, SOD, IL-10. The expression levels of MAPK14, RB1, MAPK1, JUN and ATK1 proteins were significantly decreased while IL2 protein increased administrated with Astragalus membranaceus. CONCLUSIONS Astragalus membranaceus is an effective drug in treatment of UC according to related to above targets that may exhibits the anti-UC effect via its antioxidant pathway and regulating the balance of pro-inflammatory and anti-inflammatory factors.
Collapse
Affiliation(s)
- Jingxin Mao
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lihong Tan
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Cheng Tian
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Wenxiang Wang
- College of pharmacy, Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - YanLin Zou
- College of pharmacy, Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Zhaojing Zhu
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Yan Li
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China.
| |
Collapse
|
3
|
Guo P, Li X, Xue Y, Lu Q, Liu Y, Xiong J, Wu Z, Fu S, Ye C, Wang X, Qiu Y. Using network pharmacology and molecular docking to uncover the mechanism by which quercetin alleviates deoxynivalenol-induced porcine intestinal injury. Toxicon 2024; 243:107709. [PMID: 38615996 DOI: 10.1016/j.toxicon.2024.107709] [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: 11/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Deoxynivalenol is a widespread feed contaminant that leads to vomit, which results in serious symptom such as increased intestinal permeability and even intestinal mucosal necrosis. Recent studies have reported the role of quercetin in alleviating deoxynivalenol-induced intestinal injury; however, the mechanisms and targets remain unclear. Thus, we aimed to identify the mechanisms of action by using a combination of network pharmacology and molecular docking. We identified 151 quercetin targets, 235 deoxynivalenol targets and 47 porcine intestinal injury targets by searching compound database and PubMed database, among which there were two common targets. The PPI network showed that the key proteins involved are NQO1 and PPAR-γ. The PPI network showed that the key proteins involved were NQO1 and PPARG. GO analysis found that genes were enriched primarily in response to oxidative stress. The PPI network showed that the key proteins involved are NQO1 and PPAR-γ. The genes are enriched primarily in response to oxidative stress. KEGG analysis showed enrichment of the HIF, reactive oxygen species and other signaling pathways. The molecular docking results indicated key binding activity between NQO1-quercetin and PPAR-γ-quercetin. By using network pharmacology, we have revealed the potential molecular mechanisms by which quercetin alleviates deoxynivalenol-induced porcine intestinal injury, which lays the foundation for the development of drugs to treat deoxynivalenol-induced intestinal injury in pigs.
Collapse
Affiliation(s)
- Pu Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xuemin Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yunda Xue
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qirong Lu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Jianglin Xiong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Zhongyuan Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
4
|
Gao F, Zhu F, Shuai B, Wu M, Wei C, Yuan Y, Gui Y, Tian Y, Fan H, Wu H. Quercetin ameliorates ulcerative colitis by restoring the balance of M2/M1 and repairing the intestinal barrier via downregulating cGAS‒STING pathway. Front Pharmacol 2024; 15:1351538. [PMID: 38774206 PMCID: PMC11106451 DOI: 10.3389/fphar.2024.1351538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Macrophage polarization is closely associated with the pathogenesis of ulcerative colitis (UC). Quercetin, a flavonoid, has shown promise as a treatment for inflammatory diseases, but its specific mechanism of action remains unclear. This study investigates whether quercetin can regulate intestinal macrophage polarization and promote intestinal tissue repair via the cGAS-STING pathway for the treatment of UC. In vivo, mice with 3% DSS-induced UC were intraperitoneally injected with quercetin and RU.521 for 7 days, following which their general conditions and corresponding therapeutic effects were assessed. The impact of interferon-stimulated DNA (ISD) and quercetin on macrophage polarization and the cGAS-STING pathway was investigated using RAW264.7 cells and bone marrow-derived macrophages (BMDMs) in vitro. The results demonstrated that ISD induced M1 macrophage polarization and activated the cGAS-STING pathway in vitro, while quercetin reversed ISD's inflammatory effects. In vivo, quercetin suppressed the cGAS-STING pathway in the intestinal macrophages of DSS-induced UC mice, which reduced M1 macrophage polarization, increased M2 polarization, and facilitated intestinal barrier repair in UC. Taken together, these findings provide new insights into the mechanisms via which quercetin could be used to treat UC.
Collapse
Affiliation(s)
- Fei Gao
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Shuai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Wu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunzhu Wei
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuyi Yuan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Gui
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yushi Tian
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Liu B, Zhang J, Wang X, Ye W, Yao J. Exploration of the Mechanisms Underlying Yu's Enema Formula in Treating Ulcerative Colitis by Blocking the RhoA/ROCK Pathway based on Network Pharmacology, High-performance Liquid Chromatography Analysis, and Experimental Verification. Curr Pharm Des 2024; 30:1085-1102. [PMID: 38523541 DOI: 10.2174/0113816128290586240315071044] [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: 11/28/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND The traditional Chinese medicine formula, Yu's Enema Formula (YEF), has demonstrated potential in the treatment of Ulcerative Colitis (UC). OBJECTIVE This study aimed to unveil the anti-UC mechanisms of YEF. METHODS Utilizing public databases, we obtained YEF and UC-related targets. GO and KEGG analyses were conducted via clusterProfiler and Reactome. The STRING database facilitated the construction of the PPI network, and hub targets were selected using cytoHubba. We used R software for differential expression and correlation analyses, and molecular docking was performed with PyMOL and AutoDock. HPLC analysis identified the compounds in YEF. For in vivo validation, a UC rat model was employed. RESULTS AND DISCUSSION 495 YEF-UC overlapping targets were identified. GO and KEGG analyses indicated enrichment in exogenous stimuli response, peptide response, positive MAPK cascade regulation, interleukin- related signaling, and the TLR4 cascade. Hub targets included CTNNB1, JUN, MAPK1, MAPK3, SRC, STAT3, TLR4, TP53, and RELA, which were often interconnected. Molecular docking revealed quercetin's strong binding affinity with CTNNB1, MAPK1, MAPK3, SRC, STAT3, TLR4, and TP53, consistent with HPLC analysis. In vivo experiments suggested that YEF has the potential to alleviate UC symptoms and protect the intestinal mucosal barrier by inhibiting the RhoA/ROCK pathway. CONCLUSION YEF may safeguard the intestinal mucosal barrier in UC by targeting CTNNB1, MAPK1, MAPK3, SRC, STAT3, TLR4, and TP53, while blocking the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Binbin Liu
- Department of Digestion, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Zhang
- Department of Digestion, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaoqi Wang
- Department of Digestion, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wei Ye
- Department of Digestion, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiaming Yao
- Department of Digestion, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Cui Y, Hu J, Li Y, Au R, Fang Y, Cheng C, Xu F, Li W, Wu Y, Zhu L, Shen H. Integrated Network Pharmacology, Molecular Docking and Animal Experiment to Explore the Efficacy and Potential Mechanism of Baiyu Decoction Against Ulcerative Colitis by Enema. Drug Des Devel Ther 2023; 17:3453-3472. [PMID: 38024534 PMCID: PMC10680469 DOI: 10.2147/dddt.s432268] [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: 09/09/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Background Baiyu Decoction (BYD), a clinical prescription of traditional Chinese medicine, has been proven to be valuable for treating ulcerative colitis (UC) by enema. However, the mechanism of BYD against UC remains unclear. Purpose A combination of bioinformatics methods including network pharmacology and molecular docking and animal experiments were utilized to investigate the potential mechanism of BYD in the treatment of UC. Materials and Methods Firstly, the representative compounds of each herb in BYD were detected by liquid chromatography-mass spectrometry. Subsequently, we predicted the core targets and potential pathways of BYD for treating UC through network pharmacology. And rat colitis model was established with dextran sodium sulfate. UC rats were subjected to BYD enema administration, during which we recorded body weight changes, disease activity index, and colon length to assess the effectiveness of BYD. Besides, quantitative real-time PCR, western blotting, ELISA and immunofluorescence were used to detect intestinal inflammatory factors, intestinal barrier biomarkers and TOLL-like receptor pathway in rats. Finally, the core components and targets of BYD were subjected to molecular docking so as to further validate the results of network pharmacology. Results A total of 41 active compositions and 203 targets related to BYD-UC were subjected to screening. The results of bioinformatics analysis showed that quercetin and kaempferol may be the main compounds. Additionally, AKT1, IL-6, TP53, TNF and IL-1β were regarded as potential therapeutic targets. KEGG results explained that TOLL-like receptor pathway might play a pivotal role in BYD protecting against UC. In addition, animal experiments and molecular docking validated the network pharmacology results. BYD enema treatment can reduce body weight loss, lower disease activity index score, reverse colon shortening, relieve intestinal inflammation, protect intestinal barrier, and inhibit TOLL-like receptor pathway in UC rats. Besides, molecular docking suggested that quercetin and kaempferol docked well with TLR4, AKT1, IL-6, TP53. Conclusion Utilizing network pharmacology, animal studies, and molecular docking, enema therapy with BYD was confirmed to have anti-UC efficacy by alleviating intestinal inflammation, protecting the intestinal barrier, and inhibiting the TOLL-like receptor pathway. Researchers should focus not only on oral medications but also on the rectal administration of medications in furtherance of the cure of ulcerative colitis.
Collapse
Affiliation(s)
- Yuan Cui
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jingyi Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanan Li
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ryan Au
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yulai Fang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Cheng Cheng
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Feng Xu
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Weiyang Li
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yuguang Wu
- Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Lei Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
7
|
Dembogurski DSDO, Bonfá IS, Candeloro L, Parisotto EB, Toffoli Kadri MC, Silva DB. Infusion from Miconia albicans (Melastomataceae) leaves exhibits anti-inflammatory and anti-hyperalgesic activities without toxicity. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116251. [PMID: 36791930 DOI: 10.1016/j.jep.2023.116251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The leaves of Miconia albicans have been extensively used as a traditional medicine to treat inflammation, infection, arthritis, joint pain, and analgesia, which can be purchased easily. Nevertheless, the scientific evidence of chemical profile identification and toxicity investigation is meager. AIM OF THE STUDY This study aimed to determine the chemical profile of Miconia albicans aqueous extract (MAAE), to investigate its anti-inflammatory and hyperalgesic effects, and toxicity (acute and repeated-dose oral) in vivo studies. MATERIALS AND METHODS MAAE was obtained by infusion method and its chemical constituents were analyzed and annotated by LC-DAD-MS. The in vivo tests were performed with male and female Swiss mice. Toxicity studies were examined by acute (2000 mg/kg) and repeated-dose oral assays (51.2; 256; 1280 mg/kg); anti-inflammatory evaluation was performed by paw edema and leukocyte migration, and anti-hyperalgesic properties were analyzed by abdominal writhing induced by acetic acid and formalin. The animals were treated by oral means with 51.2, 256, and 1280 mg/kg of MAAE. RESULTS Twenty-four compounds were annotated from MAAE by LC-DAD-MS, such as ellagitannins, ellagic acid derivatives, flavan-3-ol, and O-glycosylated compounds, including flavonols, triterpenes, and megastigmanes. MAAE induced no significant toxicological effects in the acute and repeated-dose oral assays at lower doses and no histological changes were observed. Hematological and biochemical showed no significant alterations. The oral administration of MAAE 256 mg/kg inhibited the edematogenic effect and reduced the leukocyte migration. In addition, MAAE decreased the abdominal writhings induced by acetic acid and the paw-licking time by formalin assay. CONCLUSION MAAE showed a significant reduction in inflammatory levels and leukocyte migration, revealing anti-hyperalgesic properties. Additionally, MAAE revealed no acute and repeated-doses toxicities.
Collapse
Affiliation(s)
- Djaceli Sampaio de Oliveira Dembogurski
- Laboratory of Natural Products and Mass Spectrometry (LaPNEM), Faculty of Pharmaceutical Sciences, Food and Nutrition (FACFAN), Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Iluska Senna Bonfá
- Laboratory of Pharmacology and Inflammation, FACFAN/ Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Luciane Candeloro
- Laboratory of Histology, Biosciences Institute (INBIO), Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Eduardo Benedetti Parisotto
- Laboratory of Hematology, Faculty of Pharmaceutical Sciences, Food and Nutrition (FACFAN), Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Mônica Cristina Toffoli Kadri
- Laboratory of Pharmacology and Inflammation, FACFAN/ Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil
| | - Denise Brentan Silva
- Laboratory of Natural Products and Mass Spectrometry (LaPNEM), Faculty of Pharmaceutical Sciences, Food and Nutrition (FACFAN), Federal University of Mato Grosso do Sul, Mato Grosso do Sul, Brazil.
| |
Collapse
|
8
|
Gong X, Huang Y, Ma Q, Jiang M, Zhan K, Zhao G. Quercetin Alleviates Lipopolysaccharide-Induced Cell Damage and Inflammation via Regulation of the TLR4/NF-κB Pathway in Bovine Intestinal Epithelial Cells. Curr Issues Mol Biol 2022; 44:5234-5246. [PMID: 36354668 PMCID: PMC9688721 DOI: 10.3390/cimb44110356] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 09/28/2023] Open
Abstract
Acute diarrhoea and intestinal inflammation represent one of the most prevalent clinical disorders of milk production, resulting in enormous annual financial damage for the dairy sector. In the context of an unsatisfactory therapeutic effect of antibiotics, the natural products of plants have been the focus of research. Quercetin is an important flavonoid found in a variety of plants, including fruits and vegetables, and has strong anti-inflammatory effects, so it has received extensive attention as a potential anti-inflammatory antioxidant. However, the underlying basis of quercetin on inflammatory reactions and oxidative tension generated by lipopolysaccharide (LPS) in bovine intestinal epithelial cells (BIECs) is currently unexplained. This research aimed to determine the influence of quercetin on LPS-induced inflammatory reactions, oxidative tension, and the barrier role of BIECs. Our findings demonstrated that BIEC viability was significantly improved in LPS-treated BIEC with 80 μg/mL quercetin compared with the control group. Indicators of oxidative overload and genes involved in barrier role revealed that 80 μg/mL quercetin efficiently rescued BIECs from oxidative and barrier impairment triggered by 5 μg/mL LPS. In addition, the mRNA expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, as well as chemokines CXCL2, CXCL5, CCL5, and CXCL8, was diminished in LPS-treated BIECs with 80 μg/mL quercetin compared with LPS alone. Furthermore, the mRNA expression of toll-like receptor 4 (TLR4), CD14, myeloid differential protein-2 (MD2), and myeloid differentiation primary response protein (MyD88) genes associated with the TLR4 signal mechanism was markedly reduced by the addition of quercetin to LPS-modulated BIECs, indicating that quercetin can suppress the TLR4 signal mechanism. We performed Western blotting on the NF-κB signalling mechanism and compared it with immunofluorescence to further corroborate this conclusion. The LPS treatment enhanced the proportions of p-IκBα/GAPDH and p-p65/GAPDH. Compared with the LPS-treated group, quercetin administration decreased the proportions of p-IκBα/GAPDH and p-p65/GAPDH. In addition, immunofluorescence demonstrated that quercetin greatly reduced the LPS-induced nuclear translocation of NF-κB p65 in BIECs. The benefits of quercetin on inflammatory reactions in LPS-induced BIECs may be a result of its capacity to inhibit the TLR4-mediated NF-κB signalling mechanism. These findings suggest that quercetin can be used as an anti-inflammatory reagent to treat intestinal inflammation induced by LPS release.
Collapse
Affiliation(s)
- Xiaoxiao Gong
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yinghao Huang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Qianbo Ma
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Maocheng Jiang
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kang Zhan
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Guoqi Zhao
- Institute of Animal Culture Collection and Application, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
9
|
Liu Z, Shao J, Lai S, Wang J, Zhao K, Tang T, Wang M. The Use of Metabolomics as a Tool to Compare the Regulatory Mechanisms in the Cecum, Ileum, and Jejunum in Healthy Rabbits and with Diarrhea. Animals (Basel) 2022; 12:ani12182438. [PMID: 36139297 PMCID: PMC9495174 DOI: 10.3390/ani12182438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary The problems caused by antibiotic abuse have swept the world, and the Chinese government has responded to calls for a comprehensive ban on antibiotics. However, not using antibiotics also challenges China’s existing livestock industry. Based on this, we carried out a nontargeted metabolomics analysis of the jejunum, ileum, and cecum of diarrhea rabbits and normal rabbits fed with antibiotic-free diets, respectively, to find out the mechanism of action of each intestinal segment group and between different intestinal segments. The screened differential metabolites were mostly related to intestinal barrier, intestinal inflammation, and autophagy after a KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis. In this paper, we analyzed the metabolic pathways that were significantly different between different intestinal segments and illustrated the mechanism and potential connections of the screened differential metabolites in different intestinal segments in the form of charts. Abstract For many years, antibiotics in feed have been an effective and economical means to promote growth and disease resistance in livestock production. However, the rampant abuse of antibiotics has also brought very serious harm to human health and the environment. Therefore, the Chinese government promulgated laws and regulations on 1 July 2020, to prohibit the use of antibiotics in feed. To improve the effect of antibiotic-free feeding on China’s existing rabbit industry, we used the nontargeted metabolomics method to detect the differences between diarrhea rabbits (Dia) and normal rabbits (Con) on an antibiotic-free diet. A total of 1902 different metabolites were identified. A KEGG analysis showed that in the cecum, metabolites were mainly concentrated in bile secretion, antifolate resistance, aldosterone synthesis, and secretion pathways. The ileal metabolites were mainly concentrated in tyrosine metabolism, phenylalanine, tyrosine and tryptophan biosynthesis, steroid hormone biosynthesis, alanine, aspartate, and glutamate metabolism. The metabolites in the jejunum were mainly rich in panquinone and other terpenoid compound quinone biosynthesis, AMPK (adenosine 5′-monophosphate (AMP)-activated protein kinase) signal, inositol phosphate metabolism, and pentose phosphate pathway. After a deep excavation of the discovered differential metabolites and metabolic pathways with large differences between groups, it was found that these metabolic pathways mainly involved intestinal inflammation, intestinal barrier, and autophagy. The results showed that panquinone and other terpenoids could increase AMPK activity to promote cell metabolism and autophagy, thus trying to prevent inflammation and alleviate intestinal disease symptoms. In addition, we discussed the possible reasons for the changes in the levels of seven intestinal endogenous metabolites in rabbits in the diarrhea group. The possibility of improving diarrhea by adding amino acids to feed was discussed. In addition, the intermediate products produced by the pentose phosphate pathway and coenzyme Q had a positive effect on steroid hormone biosynthesis to combat intestinal inflammation.
Collapse
Affiliation(s)
- Zheliang Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Chengdu Campus, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence:
| | - Jie Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Kaisen Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Meigui Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
10
|
The Role of Diet in Regulation of Macrophages Functioning. Biomedicines 2022; 10:biomedicines10092087. [PMID: 36140188 PMCID: PMC9495355 DOI: 10.3390/biomedicines10092087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 12/05/2022] Open
Abstract
The great importance of diet for health and high life-expectancy is established. The impact of nutrients on immune system is a point of growing research interest. Recent studies have found pro- and anti-inflammatory properties of some diet patterns and nutrients that can be used from the bench to the bedside for chronic low-grade inflammatory status correction. In this regard, the assessment of potential effects of nutrition on macrophage differentiation, proliferation, and functioning in health and disease is highly demanded. In this review, we present current data on the effects of nutrients on the macrophage functioning.
Collapse
|
11
|
Calabriso N, Scoditti E, Massaro M, Maffia M, Chieppa M, Laddomada B, Carluccio MA. Non-Celiac Gluten Sensitivity and Protective Role of Dietary Polyphenols. Nutrients 2022; 14:2679. [PMID: 35807860 PMCID: PMC9268201 DOI: 10.3390/nu14132679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022] Open
Abstract
Pathogenetically characterized by the absence of celiac disease and wheat allergy, non-celiac gluten sensitivity (NCGS) is a clinical entity triggered by the consumption of gluten-containing foods that relieved by a gluten-free diet. Since it is very difficult to maintain a complete gluten-free diet, there is a high interest in discovering alternative strategies aimed at reducing gluten concentration or mitigating its toxic effects. Plant-based dietary models are usually rich in bioactive compounds, such as polyphenols, recognized to prevent, delay, or even reverse chronic diseases, including intestinal disorders. However, research on the role of polyphenols in mitigating the toxicity of gluten-containing foods is currently limited. We address the metabolic fate of dietary polyphenols, both as free and bound macromolecule-linked forms, with particular reference to the gastrointestinal compartment, where the concentration of polyphenols can reach high levels. We analyze the potential targets of polyphenols including the gluten peptide bioavailability, the dysfunction of the intestinal epithelial barrier, intestinal immune response, oxidative stress and inflammation, and dysbiosis. Overall, this review provides an updated overview of the effects of polyphenols as possible dietary strategies to counteract the toxic effects of gluten, potentially resulting in the improved quality of life of patients with gluten-related disorders.
Collapse
Affiliation(s)
- Nadia Calabriso
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Egeria Scoditti
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (M.M.); (M.C.)
| | - Marcello Chieppa
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy; (M.M.); (M.C.)
| | - Barbara Laddomada
- Institute of Sciences of Food Production (ISPA), National Research Council (CNR), 73100 Lecce, Italy
| | - Maria Annunziata Carluccio
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy; (N.C.); (E.S.); (M.M.)
| |
Collapse
|
12
|
Phenotyping of Fecal Microbiota of Winnie, a Rodent Model of Spontaneous Chronic Colitis, Reveals Specific Metabolic, Genotoxic, and Pro-inflammatory Properties. Inflammation 2022; 45:2477-2497. [PMID: 35732858 DOI: 10.1007/s10753-022-01706-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/27/2022] [Accepted: 06/08/2022] [Indexed: 11/05/2022]
Abstract
Winnie, a mouse carrying a missense mutation in the MUC2 mucin gene, is a valuable model for inflammatory bowel disease (IBD) with signs and symptoms that have multiple similarities with those observed in patients with ulcerative colitis. MUC2 mucin is present in Winnie, but is not firmly compacted in a tight inner layer. Indeed, these mice develop chronic intestinal inflammation due to the primary epithelial defect with signs of mucosal damage, including thickening of muscle and mucosal layers, goblet cell loss, increased intestinal permeability, enhanced susceptibility to luminal inflammation-inducing toxins, and alteration of innervation in the distal colon. In this study, we show that the intestinal environment of the Winnie mouse, genetically determined by MUC2 mutation, selects an intestinal microbial community characterized by specific pro-inflammatory, genotoxic, and metabolic features that could imply a direct involvement in the pathogenesis of chronic intestinal inflammation. We report results obtained by using a variety of in vitro approaches for fecal microbiota functional characterization. These approaches include Caco-2 cell cultures and Caco-2/THP-1 cell co-culture models for evaluation of geno-cytotoxic and pro-inflammatory properties using a panel of 43 marker RNAs assayed by RT-qPCR, and cell-based phenotypic testing for metabolic profiling of the intestinal microbial communities by Biolog EcoPlates. While adding a further step towards understanding the etiopathogenetic mechanisms underlying IBD, the results of this study provide a reliable method for phenotyping gut microbial communities, which can complement their structural characterization by providing novel functional information.
Collapse
|
13
|
Wu T, Yang X, Xu B, Zhu H, Guo J, Zhou Y, Liang G, Sun H. Using Network Pharmacology and Molecular Docking Technology to Explore the Mechanism of Modified Pulsatilla Decoction in the Treatment of Ulcerative Colitis. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221098850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: Using network pharmacology and molecular docking technology, our aim was to clarify the biological activity, key targets, and potential pharmacological mechanisms of modified Pulsatilla decoction (MPD) in the treatment of ulcerative colitis (UC). Materials and Methods: The main active ingredients of MPD were screened using the traditional Chinese medicine systems pharmacology platform. UC targets were obtained from the GeneCard, OMIM, DisGeNET, PharmGkb, and DrugBank databases. The common genes of MPD in the treatment of UC were identified by Venn diagram. The visual interactive network diagram of “active ingredient-target-disease” was constructed using the software Cytoscape. We used the STRING database to construct a protein–protein interaction network and analyze the correlation in protein interaction. We conducted gene ontology enrichment analysis and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis for common genes using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) database and R software. Subsequently, the molecular docking verification of ingredients and targets was conducted through Discovery Studio. Last, in vivo experiments were conducted to further verify the findings. Results: A total of 51 active ingredients were screened, involving 141 common genes. The top 5 ingredients in MPD were quercetin, β-sitosterol, luteolin, kaempferol, and stigmasterol. Pathways involved in the treatment of UC include the advanced glycation end products-receptor for advanced glycation end products (AGE-RAGE) signaling pathway, the interleukin-17 (IL-17) signaling pathway, the tumor necrosis factor (TNF) signaling pathway, viral infection-related signaling pathways, and some cancer pathways. Molecular docking showed that the important ingredients of MPD were well docked with mitogen-activated protein kinase 1 (MAPK1), mitogen-activated protein kinase 8 (MAPK8), RAC-alpha serine (AKT1), vascular endothelial growth factor-A (VEGFA), transcription factor AP-1 (JUN), and interleukin-6 (IL-6). Animal experiments showed that MPD could ameliorate the injury and colitis in dextran sulfate sodium (DSS)-induced colitic rats. MPD inhibited the expression of p-p38A and p-MLC in UC rats. Conclusions: MPD has the characteristics of a multisystem, multi-ingredient, and multitarget in the treatment of UC. The possible mechanisms include inhibition of inflammation, apoptosis, oxidation, and tumor gene transcription. MPD may have a protective effect in the treatment of UC.
Collapse
Affiliation(s)
- Tingting Wu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xin Yang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
- The Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Bo Xu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Huiping Zhu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Jinwei Guo
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yu Zhou
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Guoqiang Liang
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
- Suzhou Academy of Wumen Chinese Medicine, Suzhou, China
| | - Hongwen Sun
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| |
Collapse
|
14
|
Mechanism of Fructus Mume Pills Underlying Their Protective Effects in Rats with Acetic Acid-Inducedulcerative Colitis via the Regulation of Inflammatory Cytokines and the VEGF-PI3K/Akt-eNOS Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4621131. [PMID: 35620404 PMCID: PMC9129976 DOI: 10.1155/2022/4621131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/04/2022] [Accepted: 03/22/2022] [Indexed: 12/19/2022]
Abstract
Background Fructus mume pills (FMPs) have been clinically proven to be effective for treating ulcerative colitis (UC). However, the therapeutic and protective mechanisms have not been fully studied. Aim We aimed to explore the mechanism of FMPs in an acetic acid (AA)-induced ulcerative colitis rat model. Methods The targets, GO terms, and KEGG pathways for the FMPs and UC were screened and constructed using network pharmacology. A possible mechanism was verified in a 4% AA-induced colitis rat model. Colitis activity and state were evaluated using the disease activity index, and colon ulceration and intestinal mucosal damage were determined by histopathological observation through HE, AB-PAS, and Masson pathological staining. The concentrations of TNF-α, IL-6, IL-8, IL-10, MPO, MMP9, CXCR1, eNOS, and VEGF were measured to evaluate vascular permeability effects. Results The network pharmacology results showed 108 active compounds, and 139 FMP-related targets were identified. Twenty-nine targets were identified for FMPs against UC, which included MMP9, MMP3, ESR1, PTGS1, PPARA, MPO, and NOS2. A total of 1,536 GO terms and 41 pathways were associated with FMP treatment of UC. The pharmacological evaluation showed that FMPs attenuated inflammation in AA-induced colitis by reducing the serum concentrations of TNF-α, IL-6, IL-8, and IL-10 and the colonic concentrations of MPO, MMP9, and CXCR1. FMPs ameliorated hyperpermeability by reducing the colonic VEGF and eNOS concentrations. FMPs also significantly decreased the VEGFA, VEGFR2, Src, and eNOS protein expressions in colon tissue through the VEGF-PI3K/Akt-eNOS signaling pathway. Conclusion These results suggest that FMPs control UC inflammation by regulating inflammatory cytokine concentrations. FMPs alleviate AA-induced UC by regulating microvascular permeability through the VEGF-PI3K/Akt-eNOS signaling pathway.
Collapse
|
15
|
Molecular mechanisms of Huanglian jiedu decoction on ulcerative colitis based on network pharmacology and molecular docking. Sci Rep 2022; 12:5526. [PMID: 35365737 PMCID: PMC8972650 DOI: 10.1038/s41598-022-09559-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Huanglian jiedu decoction (HLJDD) is a heat-clearing and detoxifying agent composed of four kinds of Chinese herbal medicine. Previous studies have shown that HLJDD can improve the inflammatory response of ulcerative colitis (UC) and maintain intestinal barrier function. However, its molecular mechanism is not completely clear. In this study, we verified the bioactive components (BCI) and potential targets of HLJDD in the treatment of UC using network pharmacology and molecular docking, and constructed the pharmacological network and PPI network. Then the core genes were enriched by GO and KEGG. Finally, the bioactive components were docked with the key targets to verify the binding ability between them. A total of 54 active components related to UC were identified. Ten genes are very important to the PPI network. Functional analysis showed that these target genes were mainly involved in the regulation of cell response to different stimuli, IL-17 signal pathway and TNF signal pathway. The results of molecular docking showed that the active components of HLJDD had a good binding ability with the Hub gene. This study systematically elucidates the “multi-component, multi-target, multi-pathway” mechanism of anti-UC with HLJDD for the first time, suggesting that HLJDD or its active components may be candidate drugs for the treatment of ulcerative colitis.
Collapse
|
16
|
Shou X, Wang Y, Zhang X, Zhang Y, Yang Y, Duan C, Yang Y, Jia Q, Yuan G, Shi J, Shi S, Cui H, Hu Y. Network Pharmacology and Molecular Docking Analysis on Molecular Mechanism of Qingzi Zhitong Decoction in the Treatment of Ulcerative Colitis. Front Pharmacol 2022; 13:727608. [PMID: 35237152 PMCID: PMC8883437 DOI: 10.3389/fphar.2022.727608] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is a disease with complex pathological mechanisms. We explored the potential molecular mechanisms behind the therapeutic functions of Qingzi Zhitong decoction (QZZTD) in the treatment of UC by network pharmacology and molecular docking. QZZTD is a formula of Chinese traditional medicine consisting of 10 herbs. The potential active ingredients of QZZTD and their target genes were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database, and UC-related target genes were obtained from GeneCards and OMIM databases. A total of 138 co-identified target genes were obtained by plotting the intersection target Venn diagram, and then the STRING database and Cytoscape software were used to establish protein-protein interaction networks and herb-ingredient-target networks. Four key active compounds and nine key proteins were identified. Then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses showed that the biological functions of potential target genes were associated with DNA transcription, signaling receptor and ligand activity, cytokine activity, cellular autophagy, and antioxidant pathways, with related pathways involving the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway, advanced glycosylation end product (AGE)-RAGE signaling pathway, tumor necrosis factor (TNF) signaling pathway, and IL-17 signaling pathway. Moreover, the binding activities of key target genes and essential active compounds of Chinese herbal medicines in QZZTD were further validated by molecular docking. This demonstrated that quercetin, luteolin, hyndarin, and beta-sitosterol had good binding to eight key proteins, and Akt1 was the target protein with the best binding activity, suggesting that Akt1 could be the essential mediator responsible for signaling transduction after QZZTD administration. The rat experiment verified that QZZTD inhibited PI3K-Akt pathway activation and reduced inflammation in UC. In conclusion, our study suggested four potential key active components, including quercetin, were identified in QZZTD, which could interact with Akt1 and modulate the activation of the PI3K-Akt pathway. The other three pathways may also be involved in the signaling transduction induced by QZZTD in the treatment of UC.
Collapse
Affiliation(s)
- Xintian Shou
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yumeng Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Xuesong Zhang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yanju Zhang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yan Yang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chenglin Duan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Qiulei Jia
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Yuan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Jingjing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Shuqing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Hanming Cui
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yuanhui Hu
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| |
Collapse
|
17
|
Reveals of quercetin's therapeutic effects on oral lichen planus based on network pharmacology approach and experimental validation. Sci Rep 2022; 12:1162. [PMID: 35064144 PMCID: PMC8782947 DOI: 10.1038/s41598-022-04769-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/31/2021] [Indexed: 01/03/2023] Open
Abstract
Oral lichen planus (OLP) is a localized autoimmune disease of the oral mucosa, with an incidence of up to 2%. Although corticosteroids are the first-line treatment, they cause several adverse effects. Quercetin, a naturally occurring compound, has fewer side-effects and provides long-term benefits. Besides, it has powerful anti‑inflammatory activities. Here, we combined network pharmacology with experimental verification to predict and verify the key targets of quercetin against OLP. First, 66 quercetin-OLP common targets were analyzed from various databases. The protein–protein interaction (PPI) network was constructed. Topology analysis and MCODE cluster analysis of common targets were conducted to identify 12 key targets including TP53, IL-6 and IFN-γ and their connections. Gene functions and key signaling pathways, including reactive oxygen species metabolism, IL-17 pathway and AGE-RAGE pathway, were enriched by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Then, in vitro experiments showed that quercetin interfered with Th1/Th2 balance by acting on IL-6 and IFN-γ to modulate the immune system in treating OLP. Quercetin considerably affected the apoptosis and migration of T lymphocytes in OLP patients. Our study reveals the potential therapeutic targets and signaling pathways of quercetin associated with OLP, and establishes the groundwork for future clinical applications.
Collapse
|
18
|
Rees WD, Telkar N, Lin DTS, Wong MQ, Poloni C, Fathi A, Kobor M, Zachos NC, Steiner TS. An in vitro chronic damage model impairs inflammatory and regenerative responses in human colonoid monolayers. Cell Rep 2022; 38:110283. [PMID: 35045294 DOI: 10.1016/j.celrep.2021.110283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/20/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Acute damage to the intestinal epithelium can be repaired via de-differentiation of mature intestinal epithelial cells (IECs) to a stem-like state, but there is a lack of knowledge on how intestinal stem cells function after chronic injury, such as in inflammatory bowel disease (IBD). We developed a chronic-injury model in human colonoid monolayers by repeated rounds of air-liquid interface and submerged culture. We use this model to understand how chronic intestinal damage affects the ability of IECs to (1) respond to microbial stimulation, using the Toll-like receptor 5 (TLR5) agonist FliC and (2) regenerate and protect the epithelium from further damage. Repeated rounds of damage impair the ability of IECs to regrow and respond to TLR stimulation. We also identify mRNA expression and DNA methylation changes in genes associated with IBD and colon cancer. This methodology results in a human model of recurrent IEC injury like that which occurs in IBD.
Collapse
Affiliation(s)
- William D Rees
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada; Division of Hematology, Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Nikita Telkar
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada; BC Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - David T S Lin
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada
| | - May Q Wong
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada
| | - Chad Poloni
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada
| | - Ayda Fathi
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada
| | - Michael Kobor
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada
| | - Nicholas C Zachos
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Theodore S Steiner
- BC Children's Hospital Research Institute, University of British Columbia, Rm. C328 HP East, VGH, Vancouver, BC V5Z 3J5, Canada.
| |
Collapse
|
19
|
Nutritional Regimes Enriched with Antioxidants as an Efficient Adjuvant for IBD Patients under Infliximab Administration, a Pilot Study. Antioxidants (Basel) 2022; 11:antiox11010138. [PMID: 35052642 PMCID: PMC8773281 DOI: 10.3390/antiox11010138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Antioxidants are privileged candidates for the development of adjuvants able to improve the efficiency of pharmacological therapies, particularly for chronic inflammatory syndromes. During the last 20 years, anti-TNFα (tumor necrosis factor alpha) monoclonal antibodies infusion has been the biological therapy most frequently administered but there is still large space for improvement in disease remission rates and maintenance. In this context, nutritional bioactive compounds contained in dietary patterns or included as supplements, may act as adjuvants for the induction and maintenance of IBD (inflammatory bowel diseases) remission. To verify this possibility, a single-center preliminary study (SI-CURA, Soluzioni Innovative per la gestione del paziente e il follow up terapeutico della Colite UlceRosA) was designed and carried out to evaluate whether a daily administration of purple corn supplement could improve the response to Infliximab (IFX) infusion of IBD patients with both Crohn’s disease (CD) and ulcerative colitis (UC). A cohort of 47 patients was enrolled in the study. Biological samples were collected before the first and the third IFX infusion. All patients received nutritional guidelines, 27 of them received commercial red fruit tea with low anthocyanins content, while 20 received a purple corn supplement with a high anthocyanin content. Results show that the administration of an antioxidant-enriched purple corn supplement could improve IFX-mediated disease remission in terms of circulating inflammatory markers. Comparison between CD and UC patients revealed that, at this anthocyanin dosage, the purple corn extract administration improved the IFX response in CD but not in UC patients. Our results may pave the way for a new metacentric study of CD patients, recruiting a wider cohort and followed-up over a longer observational time.
Collapse
|
20
|
Exploration of the Potential Mechanisms of Wumei Pill for the Treatment of Ulcerative Colitis by Network Pharmacology. Gastroenterol Res Pract 2022; 2021:4227668. [PMID: 34970312 PMCID: PMC8714398 DOI: 10.1155/2021/4227668] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022] Open
Abstract
Background Wumei pill (WMP) has a long history of colitis treatment in China, but the protective mechanisms have not been elucidated. To uncover the potential mechanisms of WMP against ulcerative colitis (UC), the network pharmacology approach was utilized in this study. Methods Public databases were utilized to identify the potential targets of WMP and genes related to UC. Based on the identified overlapping common targets, drug-ingredient-target gene network, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein-protein interaction (PPI) analysis were conducted. Molecular docking was carried out to verify the selected key active ingredients and core targets. Results 129 active ingredients and 622 target genes were obtained. The drug-ingredient-target gene network revealed 52 active ingredients of WMP acting on 73 targets related to UC. GO analysis revealed that biological processes were mainly associated with oxidative stress, such as, reactive oxygen species metabolic processes, response to oxidative stress, cellular response to oxidative stress, response to reactive oxygen species, and regulation of reactive oxygen species metabolic processes. KEGG analysis revealed that the immune- and inflammation-related pathways, tumor-related signaling pathways, and microbial infection-related signaling pathways were the most significant. PPI network identified 13 core target genes. The molecular docking results indicated the formation of stable bonds between the active ingredients and core target genes. Conclusions The approach of network pharmacology reveals the key ingredients, potential core targets, and biological process of WMP in the treatment of UC. The mechanisms of action of WMP involve anti-inflammation, antioxidation, and modulation of immunity, which provides evidence for the therapeutic role of WMP in UC.
Collapse
|
21
|
Wang W, Xu C, Li X, Wang Z, Yang J, Shen Y, Shi M, Chen L, Zhang L, Guo Y, Wang B, Zhang T, Pu Y. Exploration of the potential mechanism of Banxia Xiexin Decoction for the effects on TNBS-induced ulcerative colitis rats with the assistance of network pharmacology analysis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114197. [PMID: 34004261 DOI: 10.1016/j.jep.2021.114197] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/01/2021] [Accepted: 05/06/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD), an ancient TCM prescription originating from Treatise on Febrile Diseases (Shang Han Lun) of the Han Dynasty, has been widely used in modern clinical practice, especially for gastrointestinal diseases, including ulcerative colitis (UC). However, the modern decoction method of BXD differs from that of the original method. Thus, an exploration of the influence of the different decoction methods on the pharmacological effects is interesting and significant. AIM OF THE STUDY This study aimed to systematically compare the pharmacological effects of extracts of BXD on TNBS induce UC rats that were prepared by different methods, the ancient method and the modern method. The findings may provide important information for the further mechanical exploration of the classical prescription, contributing to the rational application and enhancing the understanding of BXD in modern applications or scientific research. METHODS Fifty-four SD rats were randomly divided into the following nine groups at n = 6/group: control group; model group; salicylazosulfapyridine group; BXD ancient extraction method's low-dose group (BXD-AED-L, 3.6 g BXD-AED/kg), medium-dose group (BXD-AED-M, 7.2 g BXD-AED/kg), and high-dose group (BXD-AED-H, 14.4 g BXD-AED/kg); and BXD modern extraction method's low-dose group (BXD-MED-L, 1 g BXD-MED/kg), medium-dose group (BXD-MED-M, 2 g BXD-MED/kg), and high-dose group (BXD-MED-H, 4 g BXD-MED/kg). All the groups, except the control group, were rectally injected with 70 mg/kg ethanol solution containing TNBS (2,4,6-trinitrobenzenesulfonic acid) to establish the UC models. The pharmacological evaluations including disease activity index, colon weight index, macroscopic and histological evaluation of colon damage, and inflammatory cytokine levels (IL-4, IL-10, IL-1β, TNF-α, and IL-6)were measured. In the network pharmacology analysis, the "herbs-components-targets-disease" network was constructed and visually analyzed with which the targets with a strong correlation with UC were screened out. RESULTS The results showed that both BXD-AED and BXD-MED might alleviate the severity of UC with different degrees according to the majority of indices that were evaluated. At similar doses, the BXD-AED groups performed better compared with the BXD-MED groups. With the assistance of the network pharmacology analysis, some key active components (quercetin, baicalein, wogonin, and baicalin) related to the anti-UC/inflammation were screened out. The contents of the components in BXD-AED were higher than those in BXD-MED. The joint results of the study indicated that BXD, an ancient TCM compound prescription, is an effective drug candidate for the modern treatment of UC.
Collapse
Affiliation(s)
- Weiwei Wang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Congcong Xu
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xinye Li
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zibing Wang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jinchuan Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ye Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengge Shi
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lili Zhang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yilin Guo
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bing Wang
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tong Zhang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yiqiong Pu
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
22
|
Wen S, Zhong Z, He L, Zhao D, Chen X, Mi H, Liu F. Network pharmacology dissection of multiscale mechanisms for jiaoqi powder in treating ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114109. [PMID: 33845143 DOI: 10.1016/j.jep.2021.114109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The incidence of ulcerative colitis (UC) is increasing worldwide, making it a serious public health challenge. Currently, there are no accepted curative treatments for UC. As such, the exploration of new therapeutic strategies for UC treatment is of considerable clinical importance. Jiaoqi powder (JQP) is a classic Chinese medicinal formula commonly used as a complementary and alternative medicine for treating gastrointestinal bleeding. JQP is thus a potential alternative medicine for UC treatment. However, the protective mechanism underlying the action of JQP has not been elucidated, thereby, necessitating further studies to decipher the mechanisms involved in the complex interplay among its components. AIM OF THE STUDY To explore the protective effect of JQP against UC and to further investigate its mechanism in silico and in vivo using a systems pharmacology approach. MATERIALS AND METHODS A systems pharmacology approach was used to predict the active components of JQP. Putative targets and the potential mechanism of JQP on UC were obtained through target fishing, network construction, and enrichment analyses. An animal-based model of dextran sodium sulfate (DSS)-induced colitis in C57BL/6 mice was further used to validate the treatment mechanisms of JQP. The underlying pharmacological mechanisms of JQP in UC were determined using polymerase chain reaction tests, histological staining, immunohistochemistry, enzyme-linked immunoassays, and flow cytometry analysis. RESULTS In this study, 17 effective components and 941 potential targets of JQP were identified. Similarly, 2104 UC-related targets were also identified. Construction of PPI networks led to the identification of 184 putative therapeutic targets of JQP. Sixty-nine core targets among these 184 were further screened based on their DC values. Gene ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that the core targets were primarily enriched in immune response and inflammatory signalling pathways. Subsequent animal-based in vivo experiments revealed that JQP ameliorated symptoms and histological changes in DSS colitis by significantly impairing DSS's ability to induce high expression levels of NF-κB/p65, IL-1β, IL-6, and TNF-α. JQP also reduced the levels of COX-2, CCL2, CXCL2, HIF-1α, MMP3 and MMP9 and regulated the Th17/Treg cell balance in DSS-induced mice. CONCLUSIONS This study demonstrated that JQP could treat UC by improving the mucosal inflammatory response, repairing the intestinal barrier, and modulating the Th17/Treg immune balance. The results of this study provide new insights into UC treatment and further elucidate the theoretical and practical implications of the pharmaceutical development of TCMs.
Collapse
MESH Headings
- Animals
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/pathology
- Cytokines/metabolism
- Dextran Sulfate/toxicity
- Disease Models, Animal
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Inflammation/chemically induced
- Inflammation/drug therapy
- Inflammation/immunology
- Lymph Nodes/immunology
- Male
- Metabolic Networks and Pathways/drug effects
- Mice, Inbred C57BL
- Powders
- Protein Interaction Maps
- Spleen/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/metabolism
- Mice
Collapse
Affiliation(s)
- Shuting Wen
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Zhuotai Zhong
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Long He
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Dike Zhao
- Basic Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xu Chen
- Department of Gastroenterology,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hong Mi
- Department of Gastroenterology,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Fengbin Liu
- Department of Gastroenterology,The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Baiyun Hospital of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
23
|
Duan L, Cheng S, Li L, Liu Y, Wang D, Liu G. Natural Anti-Inflammatory Compounds as Drug Candidates for Inflammatory Bowel Disease. Front Pharmacol 2021; 12:684486. [PMID: 34335253 PMCID: PMC8316996 DOI: 10.3389/fphar.2021.684486] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel disease (IBD) represents chronic recurrent intestinal inflammation resulting from various factors. Crohn’s disease (CD) and ulcerative colitis (UC) have been identified as the two major types of IBD. Currently, most of the drugs for IBD used commonly in the clinic have adverse reactions, and only a few drugs present long-lasting treatment effects. Moreover, issues of drug resistance and disease recurrence are frequent and difficult to resolve. Together, these issues cause difficulties in treating patients with IBD. Therefore, the development of novel therapeutic agents for the prevention and treatment of IBD is of significance. In this context, research on natural compounds exhibiting anti-inflammatory activity could be a novel approach to developing effective therapeutic strategies for IBD. Phytochemicals such as astragalus polysaccharide (APS), quercetin, limonin, ginsenoside Rd, luteolin, kaempferol, and icariin are reported to be effective in IBD treatment. In brief, natural compounds with anti-inflammatory activities are considered important candidate drugs for IBD treatment. The present review discusses the potential of certain natural compounds and their synthetic derivatives in the prevention and treatment of IBD.
Collapse
Affiliation(s)
- Linshan Duan
- School of Pharmaceutical Sciences Xiamen University, Xiamen, China
| | - Shuyu Cheng
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, China
| | - Long Li
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Yanling Liu
- School of Pharmaceutical Sciences Xiamen University, Xiamen, China
| | - Dan Wang
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, China
| | - Guoyan Liu
- School of Pharmaceutical Sciences Xiamen University, Xiamen, China.,Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen, China.,Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
24
|
Molecular Targets and Mechanisms of Scutellariae radix- Coptidis rhizoma Drug Pair for the Treatment of Ulcerative Colitis Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9929093. [PMID: 34149863 PMCID: PMC8195671 DOI: 10.1155/2021/9929093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022]
Abstract
This study aims to analyze the targets of the effective active ingredients of Scutellariae radix-Coptidis rhizoma drug pair (SCDP) in ulcerative colitis (UC) by network pharmacology and molecular docking and to explore the associated therapeutic mechanism. The effective active ingredients and targets of SCDP were determined from the TCMSP database, and the drug ingredient-target network was constructed using the Cytoscape software. The disease targets related to UC were searched in GeneCards, DisGeNET, OMIM, and DrugBank databases. Then, the drug ingredient and disease targets were intersected to construct a protein-protein interaction network through the STRING database. The Metascape database was used for the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the predicted targets of SCDP for UC. The Autodock software was used for molecular docking between the main active ingredient and the core target to evaluate the binding ability. SCDP has 43 effective active ingredients and 134 intersection targets. Core targets included AKT1, TP53, IL-6, VEGFA, CASP3, JUN, TNF, MYC, EGFR, and PTGS2. GO functional enrichment analysis showed that biological process was mainly associated with a cytokine-mediated signaling pathway, response to an inorganic substance, response to a toxic substance, response to lipopolysaccharide, reactive oxygen species metabolic process, positive regulation of cell death, apoptotic signaling pathway, and response to wounding. KEGG enrichment analysis showed main pathway concentrations were related to pathways in cancer, AGE-RAGE signaling pathway in diabetic complications, bladder cancer, IL-17 signaling pathway, apoptosis, p53 signaling pathway, and PI3K-Akt signaling pathway. The drug active ingredient-core target-key pathway network contains 41 nodes and 108 edges, of which quercetin, wogonin, baicalein, acacetin, oroxylin A, and beta-sitosterol are important active ingredients; PTGS2, CASP3, TP53, IL-6, TNF, and AKT1 are important targets; and the pathways involved in UC treatment include pathways in cancer, PI3K-Akt signaling pathway, AGE-RAGE signaling pathway in diabetic, apoptosis, IL-17 signaling pathway and herpes simplex infection. The active ingredient has a good binding capacity to the core target. SCDP key active ingredients are mainly quercetin, wogonin, baicalein, acacetin, oroxylin A, and beta-sitosterol, which function mainly by regulating targets, such as PTGS2, CASP3, TP53, IL-6, TNF, and AKT1, and are associated with multiple signaling pathways as pathways in cancer, PI3K-Akt signaling pathway, apoptosis, IL-17 signaling pathways.
Collapse
|
25
|
Uncovering the mechanism of Ge-Gen-Qin-Lian decoction for treating ulcerative colitis based on network pharmacology and molecular docking verification. Biosci Rep 2021; 41:227516. [PMID: 33409535 PMCID: PMC7876598 DOI: 10.1042/bsr20203565] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Background: Ge-Gen-Qin-Lian Decoction (GGQLD), a traditional Chinese medicine (TCM) formula, has been widely used for ulcerative colitis (UC) in China, but the pharmacological mechanisms remain unclear. This research was designed to clarify the underlying pharmacological mechanism of GGQLD against UC. Method: In this research, a GGQLD-compound-target-UC network was constructed based on public databases to clarify the relationship between active compounds in GGQLD and potential targets. Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were performed to investigate biological functions associated with potential targets. A protein–protein interaction network was constructed to screen and evaluate hub genes and key active ingredients. Molecular docking was used to verify the activities of binding between hub targets and ingredients. Results: Finally, 83 potential therapeutic targets and 118 corresponding active ingredients were obtained by network pharmacology. Quercetin, kaempferol, wogonin, baicalein, and naringenin were identified as potential candidate ingredients. GO and KEGG enrichment analyses revealed that GGQLD had anti-inflammatory, antioxidative, and immunomodulatory effects. The effect of GGQLD on UC might be achieved by regulating the balance of cytokines (e.g., IL-6, TNF, IL-1β, CXCL8, CCL2) in the immune system and inflammation-related pathways, such as the IL-17 pathway and the Th17 cell differentiation pathway. In addition, molecular docking results demonstrated that the main active ingredient, quercetin, exhibited good affinity to hub targets. Conclusion: This research fully reflects the multicomponent and multitarget characteristics of GGQLD in the treatment of UC. Furthermore, the present study provided new insight into the mechanisms of GGQLD against UC.
Collapse
|
26
|
Shen C, Zhao L, Du X, Tian J, Yuan Y, Jia M, He Y, Zeng R, Qiao R, Li C. Smart Responsive Quercetin-Conjugated Glycol Chitosan Prodrug Micelles for Treatment of Inflammatory Bowel Diseases. Mol Pharm 2021; 18:1419-1430. [PMID: 33522827 DOI: 10.1021/acs.molpharmaceut.0c01245] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The incidence and progression of inflammatory bowel disease are closely related to oxidative stress caused by excessive production of reactive oxygen species (ROS). To develop an efficacious and safe nanotherapy against inflammatory bowel diseases (IBD), we designed a novel pH/ROS dual-responsive prodrug micelle GC-B-Que as an inflammatory-targeted drug, which was comprised by active quercetin (Que) covalently linked to biocompatible glycol chitosan (GC) by aryl boronic ester as a responsive linker. The optimized micelles exhibited well-controlled physiochemical properties and stability in a physiological environment. Time-dependent NMR spectra traced the changes in the polymer structure in the presence of H2O2, confirming the release of the drug. The in vitro drug release studies indicated a low release rate (<20 wt %) in physiological conditions, but nearly complete release (>95 wt % after 72 h incubation) in a pH 5.8 medium containing 10 μM H2O2, exhibiting a pH/ROS dual-responsive property and sustained release behavior. Importantly, the negligible drug release in a simulated gastric environment in 1 h allowed us to perform intragastric administration, which has potential to achieve the oral delivery by mature enteric-coating modification in future. Further in vivo activities and biodistribution experiments found that the GC-B-Que micelles tended to accumulate in intestinal inflammation sites and showed better therapeutic efficacy than the free drugs (quercetin and mesalazine) in a colitis mice model. Typical inflammatory cytokines including TNF-α, IL-6, and iNOS were significantly suppressed by GC-B-Que micelle treatment. Our work promoted inflammatory-targeted delivery and intestinal drug accumulation for active single drug quercetin and improved the therapeutic effect of IBD. The current study also provided an alternative strategy for designing a smart responsive nanocarrier for a catechol-based drug to better achieve the target drug delivery.
Collapse
Affiliation(s)
- Cuiyun Shen
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Luqing Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, P. R. China
| | - Xueying Du
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jiaxin Tian
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Yi Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Mengdi Jia
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, P. R. China
| | - Ye He
- Department of Materials Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Rong Zeng
- Department of Materials Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou 510632, P. R. China
| | - Renzhong Qiao
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Chao Li
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
27
|
Caprara G. Mediterranean-Type Dietary Pattern and Physical Activity: The Winning Combination to Counteract the Rising Burden of Non-Communicable Diseases (NCDs). Nutrients 2021; 13:429. [PMID: 33525638 PMCID: PMC7910909 DOI: 10.3390/nu13020429] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Non-communicable diseases (NCDs) (mainly cardiovascular diseases, cancers, chronic respiratory diseases and type 2 diabetes) are the main causes of death worldwide. Their burden is expected to rise in the future, especially in less developed economies and among the poor spread across middle- and high-income countries. Indeed, the treatment and prevention of these pathologies constitute a crucial challenge for public health. The major non-communicable diseases share four modifiable behavioral risk factors: unhealthy diet, physical inactivity, tobacco usage and excess of alcohol consumption. Therefore, the adoption of healthy lifestyles, which include not excessive alcohol intake, no smoking, a healthy diet and regular physical activity, represents a crucial and economical strategy to counteract the global NCDs burden. This review summarizes the latest evidence demonstrating that Mediterranean-type dietary pattern and physical activity are, alone and in combination, key interventions to both prevent and control the rise of NCDs.
Collapse
Affiliation(s)
- Greta Caprara
- Department of Experimental Oncology, IEO, European Institute of Oncology, IRCCS, 20139 Milano, Italy
| |
Collapse
|
28
|
Peng X, Dai C, Zhang M, Das Gupta S. Molecular Mechanisms Underlying Protective Role of Quercetin on Copper Sulfate-Induced Nephrotoxicity in Mice. Front Vet Sci 2021; 7:586033. [PMID: 33490128 PMCID: PMC7821355 DOI: 10.3389/fvets.2020.586033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/19/2020] [Indexed: 01/16/2023] Open
Abstract
Copper overload is an established cause of nephrotoxicity, but the precise molecular mechanism remains unknown. Our study aimed to investigate the molecular mechanism of copper sulfate (CuSO4)-induced nephrotoxicity and the protective effect of the natural compound quercetin using a mouse model. Mice were orally administered CuSO4 only (200 mg/kg per day), or co-administered CuSO4 (200 mg/kg per day) plus quercetin (25, 50, or 100 mg/kg per day), or quercetin only (100 mg/kg per day), or vehicle for 28 days. The blood and kidneys were collected for the examination of serum biomarkers, oxidative stress biomarkers, changes in histopathology and gene and protein expression. Our results show that quercetin supplementation attenuates CuSO4-induced renal dysfunction and tubular necrosis in a dose-dependent manner. Quercetin supplementation at 50 and 100 mg/kg significantly attenuated CuSO4-induced oxidative damage. Quercetin supplementation also inhibited the activities of caspases-9 and-3, and the expression of p53 and Bax mRNAs. Furthermore, quercetin supplementation markedly activated the expression of Nrf2 and HO-1 mRNAs, but inhibited the expression of NF-κB, IL-1β, IL-6, and TNF-α mRNAs. In conclusion, our results revealed that quercetin supplementation could inhibit CuSO4-induced nephrotoxicity in mice via the inhibition of mitochondrial apoptotic and NF-κB pathways and the activation of Nrf2/HO-1 pathway. Our study highlights quercetin as a potential candidate in treating copper overload-induced nephrotoxicity.
Collapse
Affiliation(s)
- Xinyan Peng
- College of Life Sciences, Yantai University, Yantai, China.,College of Food Engineering, Ludong University, Yantai, China
| | - Chongshan Dai
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States.,College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Min Zhang
- College of Life Sciences, Yantai University, Yantai, China
| | - Subhajit Das Gupta
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
29
|
Candellone A, Cerquetella M, Girolami F, Badino P, Odore R. Acute Diarrhea in Dogs: Current Management and Potential Role of Dietary Polyphenols Supplementation. Antioxidants (Basel) 2020; 9:antiox9080725. [PMID: 32784917 PMCID: PMC7465157 DOI: 10.3390/antiox9080725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/28/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Acute diarrhea is one of the most common reasons why pet owners seek veterinary care for their canine companions. In many cases, signs resolve spontaneously or with symptomatic therapy without a specific cause being discovered. However, life-threatening cases can occur. The etiology is complex, including infectious diseases (endoparasites, virus, bacteria, protozoa, fungal agents) by both zoonotic and non-zoonotic pathogens, dietary indiscretion, endocrine diseases, and stress (e.g., travel or environmental changes). In the last years, the role played by oxidative stress in the pathogenesis of acute and chronic enteropathies, independently from the initial noxa, has been highlighted by many researches in both humans and animals. As a result, a series of dietary antioxidant compounds have been studied for their potential use in the treatment of intestinal inflammation. This review summarizes the traditional therapeutic and nutritional options to manage canine acute diarrhea, highlighting the need to explore the role of oxidative stress and potential antioxidant supplementation, especially polyphenols, during acute diarrheic episodes.
Collapse
Affiliation(s)
- Alessia Candellone
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, Italy; (F.G.); (R.O.)
- Correspondence: (A.C.); (P.B.)
| | - Matteo Cerquetella
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Circonvallazione 93/95, 62024 Matelica, Italy;
| | - Flavia Girolami
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, Italy; (F.G.); (R.O.)
| | - Paola Badino
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, Italy; (F.G.); (R.O.)
- Correspondence: (A.C.); (P.B.)
| | - Rosangela Odore
- Department of Veterinary Sciences, University of Turin, Largo Braccini 2, 10095 Grugliasco, Italy; (F.G.); (R.O.)
| |
Collapse
|
30
|
Intestinal Macrophages at the Crossroad between Diet, Inflammation, and Cancer. Int J Mol Sci 2020; 21:ijms21144825. [PMID: 32650452 PMCID: PMC7404402 DOI: 10.3390/ijms21144825] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Intestinal macrophages are key players in the regulation of the oral tolerance, controlling gut homeostasis by discriminating innocuous antigens from harmful pathogens. Diet exerts a significant impact on human health, influencing the composition of gut microbiota and the developing of several non-communicable diseases, including cancer. Nutrients and microbiota are able to modify the profile of intestinal macrophages, shaping their key function in the maintenance of the gut homeostasis. Intestinal disease often occurs as a breakdown of this balance: defects in monocyte-macrophage differentiation, wrong dietary habits, alteration of microbiota composition, and impairment in the resolution of inflammation may contribute to the development of intestinal chronic inflammation and colorectal cancer. Accordingly, dietary interventions and macrophage-targeted therapies are emerging as innovative tools for the treatment of several intestinal pathologies. In this review, we will describe the delicate balance between diet, microbiota and intestinal macrophages in homeostasis and how the perturbation of this equilibrium may lead to the occurrence of inflammatory conditions in the gut. The understanding of the molecular pathways and dietary factors regulating the activity of intestinal macrophages might result in the identification of innovative targets for the treatments of intestinal pathologies.
Collapse
|
31
|
Khare T, Palakurthi SS, Shah BM, Palakurthi S, Khare S. Natural Product-Based Nanomedicine in Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:E3956. [PMID: 32486445 PMCID: PMC7312938 DOI: 10.3390/ijms21113956] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
: Many synthetic drugs and monoclonal antibodies are currently in use to treat Inflammatory Bowel Disease (IBD). However, they all are implicated in causing severe side effects and long-term use results in many complications. Numerous in vitro and in vivo experiments demonstrate that phytochemicals and natural macromolecules from plants and animals reduce IBD-related complications with encouraging results. Additionally, many of them modify enzymatic activity, alleviate oxidative stress, and downregulate pro-inflammatory transcriptional factors and cytokine secretion. Translational significance of natural nanomedicine and strategies to investigate future natural product-based nanomedicine is discussed. Our focus in this review is to summarize the use of phytochemicals and macromolecules encapsulated in nanoparticles for the treatment of IBD and IBD-associated colorectal cancer.
Collapse
Affiliation(s)
- Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Brijesh M. Shah
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.S.P.); (B.M.S.); (S.P.)
| | - Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA;
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
| |
Collapse
|
32
|
Cena H, Chieppa M. Coronavirus Disease (COVID-19-SARS-CoV-2) and Nutrition: Is Infection in Italy Suggesting a Connection? Front Immunol 2020; 11:944. [PMID: 32574257 PMCID: PMC7221157 DOI: 10.3389/fimmu.2020.00944] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Novel coronavirus disease (COVID-19) was declared a global pandemic on March 11, 2020. The outbreak first occurred in Wuhan, Hubei, China, in December 2019 and hit Italy heavily in February 2020. Several countries are adopting complete or partial lockdown to contain the growth of COVID-19 infection. These measures may affect people's mental health and well-being but are necessary to avoid spreading the pandemic. There has been a gradual increase in studies exploring prevention and control measures, and we recommend paying close attention to nutrition, which may contribute to modulating some important consequences of COVID-19 infection, as such pro-inflammatory cytokine storm.
Collapse
Affiliation(s)
- Hellas Cena
- Department of Public Health, Experimental and Forensic Medicine-Dietetics and Clinical Nutrition Laboratory, University of Pavia, Pavia, Italy
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, Pavia, Italy
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Institute of Research, Castellana Grotte, Italy
- Depatrment of Immunology and Cell Biology, European Biomedical Research Institute of Salerno EBRIS, Salerno, Italy
| |
Collapse
|
33
|
Giampieri F, Battino M. Bioactive Phytochemicals and Functional Food Ingredients in Fruits and Vegetables. Int J Mol Sci 2020; 21:ijms21093278. [PMID: 32384609 PMCID: PMC7247320 DOI: 10.3390/ijms21093278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Francesca Giampieri
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo—Vigo Campus, 32004 Ourense, Spain;
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche—Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
- College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - Maurizio Battino
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo—Vigo Campus, 32004 Ourense, Spain;
- Dipartimento di Scienze Cliniche Specialistiche e Odontostomatologiche—Università Politecnica delle Marche, Via Ranieri 65, 60130 Ancona, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Correspondence:
| |
Collapse
|
34
|
De Santis S, Verna G, Serino G, Armentano R, Cavalcanti E, Liso M, Dicarlo M, Coletta S, Mastronardi M, Lippolis A, Tafaro A, Santino A, Pinto A, Campiglia P, Huang AY, Cominelli F, Pizarro TT, Chieppa M. Winnie- APCMin/+ Mice: A Spontaneous Model of Colitis-Associated Colorectal Cancer Combining Genetics and Inflammation. Int J Mol Sci 2020; 21:ijms21082972. [PMID: 32340123 PMCID: PMC7215554 DOI: 10.3390/ijms21082972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: Colorectal cancer (CRC) is among the best examples of the relationship between inflammation and increased cancer risk. (2) Methods: To examine the effects of spontaneous low-grade chronic inflammation on the pathogenesis of CRC, we developed a new murine model of colitis-associated cancer (CAC) by crossing Mucin 2 mutated mice (Winnie) with ApcMin/+ mice. (3) Results: The resulting Winnie-ApcMin/+ model combines an inflammatory background with a genetic predisposition to small intestinal polyposis. Winnie-ApcMin/+ mice show an early occurrence of inflammatory signs and dysplastic lesions in the distal colon with a specific molecular signature. (4) Conclusion: The Winnie-ApcMin/+ model is a perfect model to demonstrate that chronic inflammation represents a crucial risk factor for the onset and progression of tumoral lesions in individuals genetically predisposed to CRC.
Collapse
Affiliation(s)
- Stefania De Santis
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, 70126 Bari, Italy
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
- Correspondence: (S.D.S.); (M.C.); Tel.: +39-080-544-2763 (S.D.S.); +39-080-499-4628 (M.C.)
| | - Giulio Verna
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Grazia Serino
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Raffaele Armentano
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Marina Liso
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Manuela Dicarlo
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Sergio Coletta
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Mauro Mastronardi
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Angela Tafaro
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
| | - Angelo Santino
- Institute of Sciences of Food Production C.N.R., Unit of Lecce, 73100 Lecce, Italy;
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano (SA), Italy; (A.P.); (P.C.)
| | - Alex Y. Huang
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Angie Fowler AYA Cancer Institute, Cleveland, OH 44106, USA;
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (F.C.); (T.T.P.)
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Theresa T. Pizarro
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (F.C.); (T.T.P.)
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, 70013 Castellana Grotte (BA), Italy; (G.V.); (G.S.); (R.A.); (E.C.); (M.L.); (M.D.); (S.C.); (M.M.); (A.L.); (A.T.)
- European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy
- Correspondence: (S.D.S.); (M.C.); Tel.: +39-080-544-2763 (S.D.S.); +39-080-499-4628 (M.C.)
| |
Collapse
|