1
|
Wu K, Wang Q, Zhang Z, Luo W, Peng J, Ma X, Wang L, Xie C, Guo W. Honokiol ameliorates pyroptosis in intestinal ischemia‑reperfusion injury by regulating the SIRT3‑mediated NLRP3 inflammasome. Int J Mol Med 2025; 55:96. [PMID: 40280115 PMCID: PMC12045469 DOI: 10.3892/ijmm.2025.5537] [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: 08/11/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Intestinal ischemia‑reperfusion (IIR) injury is caused by the restoration of blood supply after a period of ischemia. It occurs in numerous clinical pathologies, such as intestinal obstruction, incarcerated hernia and septic shock, with mortality rates of 50‑80%. Honokiol (HKL), isolated from the herb Magnolia officinalis, is a biphenolic natural product with antioxidative, antibacterial, antitumor and anti‑inflammatory properties. Additionally, HKL has protective effects in ischemia‑reperfusion injuries, but its role and specific mechanisms in IIR injury are yet to be elucidated. In the present study, the superior mesenteric artery was ligated in rats to establish an IIR model. Hematoxylin and eosin staining and ELISA revealed that HKL administration ameliorated IIR‑induced injury in rats, which was demonstrated by a reduced destruction to the intestinal mucosa, as well as a reduced serum intestinal fatty acid‑binding protein concentration and Chiu's score in 10 mg/kg HKL treated IIR‑induced rats compared with those without HKL treatment. Additionally, immunohistochemical (IHC) staining and western blotting revealed that the occludin and tight junction protein 1 protein levels were increased in the 10 mg/kg HKL treated IIR‑induced rats compared with those without HKL treatment. Furthermore, an in vitro hypoxia/reoxygenation (H/R) cell model was established using IEC‑6 cells. Cell Counting Kit‑8 and lactate dehydrogenase (LDH) assays indicated that HKL mitigated the H/R‑inhibited cell viability and decreased the LDH levels in cell supernatants. Mechanistically, immunofluorescent (IF) staining and western blotting revealed that HKL inhibited H/R‑triggered pyroptosis. Furthermore, Mito‑Tracker, mitochondrial membrane potential and MitoSOX staining as well as western blotting revealed that reducing mitochondrial reactive oxygen species (ROS) inhibited the H/R‑induced pyroptosis by mitigating mitochondrial dysfunction. In the present H/R cell model, HKL improved the mitochondrial function by increasing the expression of sirtuin 3 (SIRT3), while IF staining and western blotting indicated that silencing SIRT3 notably reduced the beneficial effect of HKL on pyroptosis. In addition, IHC staining and western blotting revealed that HKL treatment mitigated the IIR‑induced pyroptosis in rats. Therefore, HKL treatment may mitigate IIR‑induced mitochondrial dysfunction and reduce mitochondrial ROS production by increasing the expression of SIRT3 protein, potentially resulting in an inhibition of pyroptosis during IIR.
Collapse
Affiliation(s)
- Ke Wu
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qiuling Wang
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhengyu Zhang
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wei Luo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jing Peng
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xin Ma
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Wang
- Research Center of Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chunguang Xie
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Wubin Guo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
2
|
Liu Z, Zhao Z, Xiao Z, Li M, Wang X, Huang Y, Li Y. Extracellular vesicles derived from bone marrow mesenchymal stem cells regulate SREBF2/HMGB1 axis by transporting miR-378a-3p to inhibit ferroptosis in intestinal ischemia-reperfusion injury. Cell Death Discov 2025; 11:223. [PMID: 40335466 PMCID: PMC12058992 DOI: 10.1038/s41420-025-02509-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
Intestinal ischemia-reperfusion (II/R) injury represents a life-threatening and complex pathophysiological process that remains challenging to treat clinically, and emerging evidence suggests that ferroptosis plays an essential role in its pathogenesis. This study aimed to investigate whether extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) can mitigate II/R-induced ferroptosis in a murine model. Using a bioinformatics database, we initially identified genes with abnormal expression patterns in II/R injury. Then, we confirmed the association between II/R injury, ferroptosis, and the HMGB1/SREBF2 axis through in vivo and in vitro experiments. To determine the role of HMGB1 in hypoxia/reoxygenation (H/R)-induced ferroptosis in Caco-2 cells, we transfected cells with either sh-HMGB1 or control sh-NC constructs and developed an H/R model in vitro. Subsequently, we examined factors regulating HMGB1-mediated ferroptosis in Caco-2 cells and assessed the effect of BMSC-EVs on this process. To further explore the mechanism underlying the protective effects of BMSC-EVs in II/R injury, we screened for miRNAs with reduced expression during II/R and verified their involvement. Among these, miR-378a-3p was identified as a candidate for regulating ferroptosis. To confirm its functional role, we treated II/R mice with BMSC-EVs overexpressing miR-378a-3p and assessed the outcomes. Our findings revealed that HMGB1, which is a key regulatory factor of ferroptosis, was significantly upregulated during II/R injury, and its knockdown alleviated H/R-induced ferroptosis in Caco-2 cells. We also found that SREBF2 directly regulates HMGB1 expression to promote H/R-induced ferroptosis in vitro. Importantly, BMSC-EVs alleviated II/R injury by suppressing ferroptosis in Caco-2 cells, and mechanistically, miR-378a-3p, a miRNA derived from BMSC-EVs, inhibited II/R-induced ferroptosis by modulating the SREBF2/HMGB1 axis. In conclusion, BMSC-EVs may exert protective effects against II/R injury by delivering miR-378a-3p, which regulates the SREBF2/HMGB1 axis to suppress ferroptosis, providing important insights into the pathological mechanisms underlying II/R injury and potential therapeutic strategies for its management.
Collapse
Affiliation(s)
- Zan Liu
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, College of Pharmacy, Changsha Medical University, Changsha, PR China
| | - Zitong Zhao
- Center of Reproductive Medicine, Changsha Hospital for Maternal and Child Health Care of Hunan Normal University, Changsha, PR China
| | - Zhenghui Xiao
- Emergency center of Hunan Children's Hospital, Changsha, Hunan, PR China
| | - Ming Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
| | - Xiyang Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yan Huang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, PR China.
- Hunan Provincial Key Laboratory of Neurorestoration, Changsha, Hunan, PR China.
| | - Yong Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China.
| |
Collapse
|
3
|
Xu B, Zhuang Y, Zhang Y, Liu S, Fan R, Jiang W. Apigenin Alleviates Intestinal Ischemia/Reperfusion Injury via Upregulating Nrf2-Mediated Tight Junction Integrity. Mol Nutr Food Res 2025; 69:e70043. [PMID: 40150847 DOI: 10.1002/mnfr.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Epithelial barrier dysfunction, critically involved in intestinal ischemia/reperfusion (I/R) injury, is significantly regulated by Nrf2-mediated oxidative stress. Apigenin, a flavonoid commonly found in fruits and vegetables with diverse biological properties, has an unclear impact on intestinal I/R injury. We hypothesize that apigenin improves intestinal barrier dysfunction by activating Nrf2 signaling. Thirty rats were randomly divided into five groups to establish an I/R model using superior mesenteric artery occlusion. Hypoxia and re-oxygenation (H/R) model was developed utilizing Caco-2 and IEC-6 cells, which were exposed to hypoxic conditions followed by re-oxygenation. Apigenin protected against intestinal mucosal damage by suppressing inflammatory cytokines release (TNF-α, IL-1β, IL-6, MPO, p < 0.01), ameliorating oxidative stress (MDA, SOD, GSH, GSH-Px, p < 0.01), and improving barrier dysfunction (DAO and TEER, p < 0.01) both in vivo and in vitro, without causing significant changes in the corresponding normal controls (p > 0.05). Apigenin up-regulated the protein expression of Nrf2, HO-1, and tight junction (TJ) proteins (p < 0.01). Furthermore, the knockdown of Nrf2 significantly abrogated apigenin-enhanced the TJ expression. Apigenin pretreatment alleviates intestinal I/R-induced barrier damage through Nrf2 activation and TJ upregulation, offering new strategies for preventing or treating I/R-associated intestinal diseases.
Collapse
Affiliation(s)
- Bin Xu
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Biomedical Engineering & Technology, Qilu Medical University, Zibo, Shandong, China
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yan Zhuang
- Key Laboratory of Biomedical Engineering & Technology, Qilu Medical University, Zibo, Shandong, China
| | - Ying Zhang
- Department of Environmental Physiology Faculty of Medicine, Shimane University, Shimane, Japan
| | - Suoning Liu
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Rongjun Fan
- Key Laboratory of Biomedical Engineering & Technology, Qilu Medical University, Zibo, Shandong, China
| | - Weiru Jiang
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Liu Y, Bao H. Ergosterone ameliorates RRR-induced spleen deficiency by gut microbiota-gut metabolites and P38MAPK signaling pathway. Front Microbiol 2025; 16:1501068. [PMID: 40109979 PMCID: PMC11920137 DOI: 10.3389/fmicb.2025.1501068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Spleen deficiency is an important immune and digestive system change. Ergosterone (ER) is bioactive steroid; however, to date, no relevant studies have explored its potential efficacy in treating spleen deficiency. The aim of the present study was to investigate the therapeutic effects and mechanism of action of ER on spleen deficiency syndrome induced by Rhei Radix et Rhizoma (RRR). RRR was used to induce the development of a spleen deficiency rat model to observe changes in body weight and pathological changes in organ tissues. Additionally, the levels of relevant immune factors and gastrointestinal hormones were measured, as well as the expression of intestinal tight junction proteins and the P38MAPK signaling pathway. Changes in intestinal microbiota and metabolites were measured, and the effect of ER on the RRR-induced spleen deficiency rat model was evaluated. ER notably alleviated the symptoms of RRR-induced spleen deficiency induced in rats and offered protection against organ damage. Ergosterone can increase the expression of immunoglobulins, inhibits the increase in inflammatory factors, improve gastrointestinal hormone disorders, protect the intestinal mucosa, and repair intestinal barrier damage. The ER-treated group exhibited substantial upregulation of claudin and occludin mRNA and protein expression levels in the colonic tissue. Additionally, ER inhibited the P38MAPKsignaling pathway, thereby improving RRR induced spleen deficiency syndrome in rats. ER also influences the metabolic pathways of protein digestion and absorption, biosynthesis of unsaturated fatty acids, and arachidonic acid metabolism. In addition, ER can regulate and enhance the composition of intestinal flora in rats with spleen deficiency, increase the diversity of dominant flora, and inhibit the proliferation of harmful bacteria. ER can treat spleen deficiency syndrome by enhancing immune function, improving gastrointestinal function, repairing the intestinal barrier, and regulating intestinal flora and intestinal metabolites.
Collapse
Affiliation(s)
- Ying Liu
- Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun, Jilin, China
- College of Traditional Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| | - Haiying Bao
- Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, Changchun, Jilin, China
- College of Traditional Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin, China
| |
Collapse
|
5
|
Sulejmanović M, Panić M, Redovniković IR, Milić N, Drljača J, Damjanović A, Vidović S. Sustainable isolation of ginger (Zingiber officinale) herbal dust bioactive compounds with favorable toxicological profile employing natural deep eutectic solvents (NADES). Food Chem 2025; 464:141545. [PMID: 39395331 DOI: 10.1016/j.foodchem.2024.141545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
The usage of ginger (Zingiber officinale) has increased in recent years due to its positive effect on human health affiliated with its richness in gingerols and shogaols. This study optimized the Ultrasound-assisted extraction (UAE) for better phenolic compounds isolation from ginger herbal dust (GHD), a filter tea industry by-product. The extraction was performed using raw and defatted GHD-previously processed by Supercritical fluid extraction - CO2. An additional advantage was using COSMOtherm software for 71 natural deep eutectic solvents (NADES) screening, to select the optimal one for GHD 6-gingerol recovery. As an optimal NADES, Malic acid:Glucose (MA:Glc) in the 1:1 ratio was determined. The optimal MA: Glc-based extract with a 6-gingerol content of 1.90±0.05 mg/g, an antioxidant activity of 321.28±5.09 μmol TE/g, and a favorable toxicological profile was obtained in 2 min of UAE under the sonication amplitude of 20 %, approving the benefits and the sustainability of the present study.
Collapse
Affiliation(s)
- Mirjana Sulejmanović
- Department of Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Boulevard Cara Lazara 1, 21000 Novi Sad, Serbia; Laboratory for Cell Culture Technology and Biotransformations, Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierotti Street 6, 10000 Zagreb, Croatia.
| | - Manuela Panić
- Laboratory for Cell Culture Technology and Biotransformations, Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierotti Street 6, 10000 Zagreb, Croatia.
| | - Ivana Radojčić Redovniković
- Laboratory for Cell Culture Technology and Biotransformations, Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierotti Street 6, 10000 Zagreb, Croatia.
| | - Nataša Milić
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia.
| | - Jovana Drljača
- Department of Pharmacy, Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000, Novi Sad, Serbia.
| | - Anja Damjanović
- Laboratory for Cell Culture Technology and Biotransformations, Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierotti Street 6, 10000 Zagreb, Croatia.
| | - Senka Vidović
- Department of Pharmaceutical Engineering, Faculty of Technology Novi Sad, University of Novi Sad, Boulevard Cara Lazara 1, 21000 Novi Sad, Serbia.
| |
Collapse
|
6
|
Yang M, Xu W, Yue C, Li R, Huang X, Yan Y, Yan Q, Liu S, Liu Y, Li Q. Adipose-derived stem cells promote the recovery of intestinal barrier function by inhibiting the p38 MAPK signaling pathway. Eur J Histochem 2025; 69:4158. [PMID: 39836101 PMCID: PMC11788713 DOI: 10.4081/ejh.2025.4158] [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: 11/05/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Intestinal barrier damage causes an imbalance in the intestinal flora and microbial environment, promoting a variety of gastrointestinal diseases. This study aimed to explore the mechanism by which adipose-derived stem cells (ADSCs) repair intestinal barrier damage. The human colon adenocarcinoma cell line Caco-2 and rats were treated with lipopolysaccharide (LPS) to establish in vitro and in vivo models, respectively, of intestinal barrier damage. The expression of inflammatory cytokines (TNF-α, HMGB1, IL-1β and IL-6), antioxidant enzymes (iNOS, SOD and CAT), and oxidative products (MDA and 8-iso-PGF2α) was detected using ELISA kits and related reagent kits. Apoptosis-related proteins (Bcl-2, Bax, Caspase-3 and Caspase-9), tight junction proteins (ZO-1, Occludin, E-cadherin, and Claudin-1) and p38 MAPK pathway-associated protein were detected by Western blotting. In addition, cell viability and apoptosis was determined by a CCK-8 kit and flow cytometry, respectively. Cell permeability was assayed by the transepithelial electrical resistance value and FITC-dextran concentration. The homing effect of ADSCs was detected by fluorescence labeling, and intestinal barrier tissue was observed by HE staining. After ADSC treatment, the level of phosphorylated p38 MAPK protein decreased, the expression of inflammatory factors, oxidative stress and cell apoptosis decreased, the expression of tight junction proteins increased, and cell permeability decreased in Caco-2 cells stimulated with LPS. In rats, ADSCs are directionally recruited to damaged intestinal tissue. ADSCs significantly decreased the levels of D-lactate, diamine oxidase (DAO) and FITC-dextran induced by LPS. ADSCs promoted tight junction proteins and inhibited oxidative stress in intestinal tissue. These effects were reversed after the use of a p38 MAPK activator. ADSCs can be directionally recruited to intestinal tissue, upregulate tight junction proteins, and reduce apoptosis and oxidative stress by inhibiting the p38MAPK signaling pathway. This study provides novel insights into the treatment of intestinal injury.
Collapse
Affiliation(s)
- Mei Yang
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Wangbin Xu
- Department of Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chaofu Yue
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Rong Li
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Xian Huang
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Yongjun Yan
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Qinyong Yan
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Shisheng Liu
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Yuan Liu
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| | - Qiaolin Li
- Department of Critical Care Medicine, The Qujing No.1 People's Hospital, Qujing
| |
Collapse
|
7
|
Li J, Huang J, Ai G, Zheng X, Chen B, Gong S, Lu X, Su Z, Chen J, Xie Q, Li Y, Yan F. The Protective Effects of Modified Dachaihu Decoction against LPS-induced Acute Lung Injury via Modulating PI3K/Akt Signalling Pathway. Comb Chem High Throughput Screen 2025; 28:755-767. [PMID: 40326256 DOI: 10.2174/0113862073282311240226113714] [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: 11/03/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 05/07/2025]
Abstract
BACKGROUND Modified Dachaihu decoction (MDD) is a herbal prescription that has shown promising therapeutic benefits in ameliorating pulmonary diseases in clinical practice. However, the detailed mechanisms remain unclear. OBJECTIVE This study aimed to elucidate the lung-protective effects of MDD against acute lung injury (ALI) and the involvement of underlying mechanisms. METHODS High-performance liquid chromatography (HPLC) was performed to identify the main active ingredients of MDD. Network pharmacological method was adapted to explore the potential mechanisms. Mice were orally administered MDD (11.25, 22.5, and 45 g/kg) once daily for 7 days. H&E staining was performed to evaluate histological changes in the lungs. Levels of inflammatory cytokines and oxidative stress markers were measured to determine the extent of lung injury. Total protein content in bronchoalveolar lavage fluid (BALF) and lung wet/dry weight ratio were measured to assess the severity of pulmonary edema. TUNEL staining and immunohistochemistry analysis were performed to detect apoptosis. RT-qPCR and western blotting were performed to validate the mechanisms involved. RESULTS About 10 main active ingredients of MDD were identified. Notably, treatment with MDD resulted in a remarkable reduction in total protein content in BALF and lung W/D weight ratio, as well as substantial mitigation of the inflammatory response and oxidative stress. Mechanistically, the PI3K/Akt signalling pathway was activated. Moreover, MDD pretreatment downregulated p53 and caspase-9 mRNA expression and decreased the Bax/Bcl-2 ratio to ameliorate lung apoptosis. CONCLUSIONS MDD exhibited pronounced therapeutic effects via attenuating inflammatory response, oxidative stress, and apoptosis. These therapeutic effects could be attributed to the synergistic effect of the main active ingredients and are believed to be associated with the activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Jincan Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Jiechun Huang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
| | - Gaoxiang Ai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Xiaohong Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Baoyi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Shiting Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Xiaowei Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Qingfeng Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Fang Yan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510006, PR China
| |
Collapse
|
8
|
Qian J, Li Z, Wang J, Lin Y, Yu Y. 6-gingerol and its derivatives inhibit Helicobacter pylori-induced gastric mucosal inflammation and improve gastrin and somatostatin secretion. Front Microbiol 2024; 15:1451563. [PMID: 39234535 PMCID: PMC11371576 DOI: 10.3389/fmicb.2024.1451563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/17/2024] [Indexed: 09/06/2024] Open
Abstract
The resistance of Helicobacter pylori (H. pylori) has increased in recent years, prompting a trend in the research and development of new drugs. In our study, three derivatives (JF-1, JF-2, and JF-3) were synthesized using 6-gingerol as the main component, while JF-4, containing both 6-gingerol and 6-shogaol as the main components, was extracted from dried ginger. The minimum inhibitory concentrations (MICs), determined using the ratio dilution method, were 80 μg/mL for JF-1, 40 μg/mL for JF-2, 30 μg/mL for JF-3, 40 μg/mL for JF-4, 60 μg/mL for 6-gingerol standard (SS), and 0.03 μg/mL for amoxicillin (AMX). After treating H. pylori-infected mice, the inflammation of the gastric mucosa was suppressed. The eradication rate of H. pylori was 16.7% of JF-3 low-dose treatment (LDT), 25.0% of JF-3 high-dose treatment (HDT), 16.7% of JF-4 LDT, 16.7% of JF-4 HDT, 30% of SS LDT, 50% of SS HDT, and 36.4% of the positive control group (PCG). The levels of gastrin, somatostatin (SST), IFN-γ, IL-4, and IL-8 were significantly recovered in the JF-3 and JF-4 administration groups, but the effect was stronger in the high-dose group. These results demonstrate that 6-gingerol and its derivatives have significant anti-Helicobacter pylori effects and are promising potential treatments for H. pylori infection.
Collapse
Affiliation(s)
- Jiali Qian
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- Department of Gastroenterology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhennan Li
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Jinhui Wang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuxian Lin
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China
| | - Yingcong Yu
- The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, China
- School of Medicine, Shanghai University, Shanghai, China
- The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Xu R, Bi Y, He X, Zhang Y, Zhao X. Kidney-tonifying blood-activating decoction delays ventricular remodeling in rats with chronic heart failure by regulating gut microbiota and metabolites and p38 mitogen-activated protein kinase/p65 nuclear factor kappa-B/aquaporin-4 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118110. [PMID: 38580189 DOI: 10.1016/j.jep.2024.118110] [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: 12/09/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial infarction has likely contributed to the increased prevalence of heart failure(HF).As a result of ventricular remodeling and reduced cardiac function, colonic blood flow decreases, causing mucosal ischemia and hypoxia of the villous structure of the intestinal wall.This damage in gut barrier function increases bowel wall permeability, leading to fluid metabolism disorder,gut microbial dysbiosis, increased gut bacteria translocation into the circulatory system and increased circulating endotoxins, thus promoting a typical inflammatory state.Traditional Chinese Medicine plays a key role in the prevention and treatment of HF.Kidney-tonifying Blood-activating(KTBA) decoction has been proved for clinical treatment of chronic HF.However,the mechanism of KTBA decoction on chronic HF is still unclear. AIMS OF THE STUDY The effect of KTBA decoction on gut microbiota and metabolites and p38MAPK/p65NF-κB/AQP4 signaling in rat colon was studied to investigate the mechanism that KTBA decoction delays ventricular remodeling and regulates water metabolism disorder in rats with HF after myocardial infarction based on the theory of "Kidney Storing Essence and Conducting Water". MATERIAL AND METHODS In vivo,a rat model of HF after myocardial infarction was prepared by ligating the left anterior descending coronary artery combined with exhaustive swimming and starvation.The successful modeling rats were randomly divided into five groups:model group, tolvaptan group(gavaged 1.35mg/(kg•D) tolvaptan),KTBA decoction group(gavaged 15.75g/(kg•D) of KTBA decoction),KTBA decoction combined with SB203580(p38MAPK inhibitor) group(gavaged 15.75g/(kg•D) of KTBA decoction and intraperitoneally injected 1.5mg/(kg•D) of SB203580),and KTBA decoction combined with PDTC(p65NF-kB inhibitor) group(gavaged 15.75g/(kg•D) of KTBA decoction and intraperitoneally injected 120mg/(kg•D) of PDTC).The sham-operation group and model group were gavaged equal volume of normal saline.After 4 weeks of intervention with KTBA decoction,the effect of KTBA decoction on the cardiac structure and function of chronic HF model rats was observed by ultrasonic cardiogram.General state and cardiac index in rats were evaluated.Enzyme linked immunosorbent assay(ELISA) was used to measure N-terminal pro-brain natriuretic peptide (NT-proBNP) concentration in rat serum.Hematoxylin and eosin(H&E) staining,and transmission electron microscope(TEM) were used to observe the morphology and ultrastructure of myocardial and colonic tissue,and myocardial fibrosis was measured by Masson's staining.Cardiac E-cadherin level was detected by Western blot.The mRNA expression and protein expression levels of p38MAPK,I-κBα, p65NF-κB,AQP4,Occludin and ZO-1 in colonic tissue were detected by reverse transcription-quantitative real-time polymerase chain reaction(RT-qPCR) and immunohistochemistry. Protein expression of p38MAPK, p-p38MAPK,I-κBα,p-I-κBα,p65NF-κB, p-p65NF-κB,AQP4,Occludin and ZO-1 in rat colon was detected using Western blot.Colonic microbiota and serum metabolites were respectively analyzed by amplicon sequencing and liquid chromatography-mass spectrometry.In vitro, CCD-841CoN cell was placed in the ischemic solution under hypoxic conditions (94%N2,5%CO2,and 1%O2) in a 37 °C incubator to establish an ischemia and hypoxia model.The CCD-841CoN cells were divided into 7 groups, namely blank group and model group with normal rat serum plus control siRNA, tolvaptan group with rat serum containing tolvaptan plus control siRNA, KTBA group with rat serum containing KTBA plus control siRNA, KTBA plus p38MAPK siRNA group, KTBA plus p65NF-κB siRNA group,and KTBA plus AQP4siRNA group.After 24h and 48h of intervention with KTBA decoction,RT-qPCR,immunofluorescence and Western blot was used to detect the mRNA expression and protein expression levels of p38MAPK,I-κBα,p65NF-κB,AQP4, Occludin and ZO-1 in CCD-841CoN cells. RESULTS Compared with the model, KTBA decoction improved the general state, decraesed the serum NT-proBNP level,HW/BW ratio, LVIDd and LVIDs, increased E-cadherin level,EF and FS,reduced number of collagen fibers deposited in the myocardial interstitium,and recovered irregular arrangement of myofibril and swollen or vacuolated mitochondria with broken crista in myocardium.Moreover, KTBA decoction inhibited the expression of p38MAPK,I-κBα,and p65NF-κB and upregulated AQP4, Occludin and ZO-1 in colon tissues and CCD-841CoN cells.Additionally,p38siRNA or SB203580, p65siRNA or PDTC, and AQP4siRNA partially weakened the protective effects of KTBA in vitro and vivo.Notably,The LEfSe analysis results showed that there were six gut biomaker bacteria in model group, including Allobaculum, Bacillales,Turicibacter, Turicibacterales,Turicibacteraceae,and Bacilli. Besides, three gut biomaker bacteria containing Deltaproteobacteria, Desulfovibrionaceae,and Desulfovibrionales were enriched by KTBA treatment in chronic HF model.There were five differential metabolites, including L-Leucine,Pelargonic acid, Capsidiol,beta-Carotene,and L- Erythrulose, which can be regulated back in the same changed metabolic routes by the intervention of KTBA.L-Leucine had the positive correlation with Bacillales, Turicibacterales,Turicibacteraceae,and Turicibacter.L-Leucine significantly impacts Protein digestion and absorption, Mineral absorption,and Central carbon metabolism in cancer regulated by KTBA, which is involved in the expression of MAPK and tight junction in intestinal epithelial cells. CONCLUSIONS KTBA decoction manipulates the expression of several key proteins in the p38MAPK/p65NF-κB/AQP4 signaling pathway, modulates gut microbiota and metabolites toward a more favorable profile, improves gut barrier function, delays cardiomyocyte hypertrophy and fibrosis,and improves cardiac function.
Collapse
Affiliation(s)
- Rui Xu
- Liaoning University of Traditional Chinese Medicine,Shenyang,Liaoning 110847,China
| | - Yanping Bi
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine,Jilin,Jilin 132000,China
| | - Xiaoteng He
- Liaoning University of Traditional Chinese Medicine,Shenyang,Liaoning 110847,China
| | - Yan Zhang
- The Affiliated Hospital, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110032, China.
| | - Xin Zhao
- The Second Hospital, Dalian Medical University, Dalian, Liaoning 116023, China.
| |
Collapse
|
10
|
Liu C, Zhao Y, Zhao WJ. Positive Effect of 6-Gingerol on Functional Plasticity of Microglia in a rat Model of LPS-induced Depression. J Neuroimmune Pharmacol 2024; 19:20. [PMID: 38758335 DOI: 10.1007/s11481-024-10123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
Neuroinflammation has emerged as a crucial factor in the development of depression. Despite the well-known anti-inflammatory properties of 6-gingerol, its potential impact on depression remains poorly understood. This study aimed to investigate the antidepressant effects of 6-gingerol by suppressing microglial activation. In vivo experiments were conducted to evaluate the effect of 6-gingerol on lipopolysaccharide (LPS)-induced behavioral changes and neuroinflammation in rat models. In vitro studies were performed to examine the neuroprotective properties of 6-gingerol against LPS-induced microglial activation. Furthermore, a co-culture system of microglia and neurons was established to assess the influence of 6-gingerol on the expression of synaptic-related proteins, namely synaptophysin (SYP) and postsynaptic density protein 95 (PSD95), which are influenced by microglial activation. In the in vivo experiments, administration of 6-gingerol effectively alleviated LPS-induced depressive behavior in rats. Moreover, it markedly suppressed the activation of rat prefrontal cortex (PFC) microglia induced by LPS and the activation of the NF-κB/NLRP3 inflammatory pathway, while also reducing the levels of inflammatory cytokines IL-1β and IL-18. In the in vitro experiments, 6-gingerol mitigated nuclear translocation of NF-κB p65, NLRP3 activation, and maturation of IL-1β and IL-18, all of which were induced by LPS. Furthermore, in the co-culture system of microglia and neurons, 6-gingerol effectively restored the decreased expression of SYP and PSD95. The findings of this study demonstrate the neuroprotective effects of 6-gingerol in the context of LPS-induced depression-like behavior. These effects are attributed to the inhibition of microglial hyperactivation through the suppression of the NF-κB/NLRP3 inflammatory pathway.
Collapse
Affiliation(s)
- Chong Liu
- Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Dadao, Binhu District, Wuxi, Jiangsu, 214122, P.R. China
| | - Yan Zhao
- College of Environmental Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - Wei-Jiang Zhao
- Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Dadao, Binhu District, Wuxi, Jiangsu, 214122, P.R. China.
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, P.R. China.
| |
Collapse
|
11
|
Rong Y, Xu M, Hu T, Zhang S, Fu J, Liu H. Effects of butyrate on intestinal ischemia-reperfusion injury via the HMGB1-TLR4-MyD88 signaling pathway. Aging (Albany NY) 2024; 16:7961-7978. [PMID: 38709282 PMCID: PMC11131991 DOI: 10.18632/aging.205797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND This study combined bioinformatics and experimental verification in a mouse model of intestinal ischemia-reperfusion injury (IRI) to explore the protection mechanism exerted by butyrate against IRI. METHODS GeneCards, Bioinformatics Analysis Tool for Molecular Mechanisms of Traditional Chinese Medicine and GSE190581 were used to explore the relationship between butyrate and IRI and aging. Protein-protein interaction networks involving butyrate and IRI were constructed via the STRING database, with hub gene analysis performed through Cytoscape. Functional enrichment analysis was conducted on intersection genes. A mouse model of IRI was established, followed by direct arterial injection of butyrate. The experiment comprised five groups: normal, sham, model, vehicle, low-dose butyrate, and high-dose butyrate. Intestinal tissue observation was done via transmission electron microscopy (TEM), histological examination via hematoxylin and eosin (H&E) staining, tight junction proteins detection via immunohistochemistry, and Western blot analysis of hub genes. Drug-target interactions were evaluated through molecular docking. RESULTS Butyrate protected against IRI by targeting 458 genes, including HMGB1 and TLR4. Toll-like receptor pathway was implicated. Butyrate improved intestinal IRI by reducing mucosal damage, increasing tight junction proteins, and lowering levels of HMGB1, TLR4, and MyD88. Molecular docking showed strong binding energies between butyrate and HMGB1 (-3.7 kcal/mol) and TLR4 (-3.8 kcal/mol). CONCLUSIONS According to bioinformatics predictions, butyrate mitigates IRI via multiple-target and multiple-channel mechanisms. The extent of IRI can be reduced by butyrate through the inhibition of the HMGB1-TLR4-MyD88 signaling pathway, which is related to senescence.
Collapse
Affiliation(s)
- Yuanyuan Rong
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Meili Xu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Tao Hu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Shasha Zhang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Jianfeng Fu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Huaqin Liu
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| |
Collapse
|
12
|
Yu Q, Li J, Cui M, Mei C, He Q, Du X. 6-Gingerol attenuates hepatic ischemia/reperfusion injury through regulating MKP5-mediated P38/JNK pathway. Sci Rep 2024; 14:7747. [PMID: 38565569 PMCID: PMC10987508 DOI: 10.1038/s41598-024-58392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
6-Gingerol, the main bioactive compound of ginger, has antioxidant, anti-inflammatory, anti-cancer and neuroprotective effects. However, it is unclear whether 6-Gingerol has protective effects against hepatic ischemia/reperfusion (I/R) injury. In this study, the mouse liver I/R injury model and the mouse AML12 cell hypoxia/reoxygenation (H/R) model were established by pretreatment with 6-Gingerol at different concentrations to explore the potential effects of 6-Gingerol. Serum transaminase levels, liver necrotic area, cell viability, inflammatory response, and cell apoptosis were used to assess the effect of 6-Gingerol on hepatic I/R or cell H/R injury. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to detect the mRNA and protein expression. The results show that 6-Gingerol decreased serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels, liver necrosis, inflammatory cytokines IL-1β, IL-6, MCP-1, TNF-α expression, Ly6g+ inflammatory cell infiltration, protein phosphorylation of NF-κB signaling pathway, Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) positive cells, cell apoptosis rate, the protein expression of pro-apoptotic protein BAX and C-Caspase3, increased cell viability, and expression of anti-apoptotic protein BCL-2. Moreover, 6-Gingerol could increase the mRNA and protein expression of mitogen activated protein kinase phosphatase 5 (MKP5) and inhibit the activation of P38/JNK signaling pathway. In MKP5 knockout (KO) mice, the protective effect of 6-gingerol and the inhibition of P38/JNK pathway were significantly weakened. Therefore, our results suggest that 6-Gingerol exerts anti-inflammatory and anti-apoptotic effects to attenuate hepatic I/R injury by regulating the MKP5-mediated P38/JNK signaling pathway.
Collapse
Affiliation(s)
- Qiwen Yu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiye Li
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mengwei Cui
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chaopeng Mei
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qianqian He
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoxiao Du
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Erqi, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
13
|
Li Z, Huang Z, Jia G, Zhao H, Liu G, Chen X. L-theanine attenuates H 2O 2-induced inflammation and apoptosis in IPEC-J2 cells via inhibiting p38 MAPK signaling pathway. Food Chem Toxicol 2024; 186:114561. [PMID: 38438008 DOI: 10.1016/j.fct.2024.114561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
This study investigated the protective effects of L-theanine on hydrogen peroxide (H2O2)-induced intestinal barrier dysfunction in IPEC-J2 cells. Results showed that L-theanine reduced H2O2-induced IPEC-J2 cells inflammation and apoptosis, and decreased protein phosphorylation levels of p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor kappa-B (NF-κB). The p38 MAPK inhibitor (SB203580) decreased oxidative stress, the protein expression of phosphorylation of p38 MAPK and NF-κB, the H2O2-induced increase in mRNA expression of pro-apoptotic and pro-inflammatory related genes expression and secretion, and tight junction protein related genes expression, which was similar to the effect of L-theanine. In conclusion, L-theanine inhibited H2O2-induced oxidative damage and inflammatory reaction, eliminated apoptosis, and protected intestinal epithelial barrier damage by inhibiting the activation of p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Zhongqing Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, 611130, PR China.
| |
Collapse
|
14
|
Liu Y, Gong Y, Li M, Li J. Quercetin protects against hyperglycemia-induced retinopathy in Sprague Dawley rats by regulating the gut-retina axis and nuclear factor erythroid-2-related factor 2 pathway. Nutr Res 2024; 122:55-67. [PMID: 38185061 DOI: 10.1016/j.nutres.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024]
Abstract
Hyperglycemia-related retinopathy is a disease with a high blindness rate. Recent reports indicate that many flavonol compounds have the potential to prevent the occurrence of disease in the retina by regulating the gut-retina axis. Here, we hypothesized that quercetin could alleviate the symptoms of retinopathy. To clarify the mechanism, Sprague Dawley rats were fed a high-fat diet containing quercetin for 12 weeks and injected with streptozotocin in the ninth week. Additionally, neomycin and ampicillin were used to establish a pseudo-sterile rat model. Afterward, changes in the retina were investigated by using electroretinogram and optical coherence tomography. Blood and tissue samples were collected and biochemical components were analyzed. The extent of intestinal injury was determined via hematoxylin-eosin staining. Microbial community structure was analyzed by using 16S ribosomal RNA sequencing. Finally, the expression of genes was analyzed using real-time polymerase chain reaction. The results showed that quercetin reduced the decline in electroretinography amplitude and outer nuclear layer thickness, increased the activities of antioxidant enzymes, decreased the contents of proinflammatory factors and blood glucose, enhanced the concentration of insulin, and inhibited intestinal dysbiosis and improved gut morphology. Importantly, the underexpression of nuclear factor erythroid-2 related factor 2 in the retina was reversed by quercetin. However, trend changes were no longer significant in most of the indicators after antibiotic treatment. In summary, quercetin has therapeutic effects on retinopathy by regulating the gut-retina axis and nuclear factor erythroid-2 related factor 2 pathway, and the presence of gut microbiota helps quercetin exert its effects on the retina.
Collapse
Affiliation(s)
- Yaojie Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yibo Gong
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin Eye Institute, Tianjin 300384, China
| | - Mengting Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Jianke Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
15
|
Wang Y, Li B, Liu G, Han Q, Diao Y, Liu J. Corilagin attenuates intestinal ischemia/reperfusion injury in mice by inhibiting ferritinophagy-mediated ferroptosis through disrupting NCOA4-ferritin interaction. Life Sci 2023; 334:122176. [PMID: 37858718 DOI: 10.1016/j.lfs.2023.122176] [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: 08/24/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
AIMS Intestinal ischemia reperfusion (II/R) is a common clinical emergency. Ferroptosis is reported to play a role in II/R injury. Our previous studies revealed that corilagin significantly attenuates intestinal ischemia/reperfusion injuries. However, the underlying molecular mechanism is unclear and requires further study. MATERIALS AND METHODS DAO, GSSG/T-GSH, MDA, and Fe2+ were measured by assay kits, 4-HNE was assessed by IHC, and 15-LOX was measured by ELISA. Mitochondrial damage was observed by TEM and cellular oxidation levels were detected by C11-BODIPY 581/591 and DHE probes. LC3, p62, Beclin1, ACSL4, GPX4, NCOA4, and ferritin expression were examined by WB in vivo and in vitro. IF, co-IF, q-PCR, and constructed NCOA4-knock-down IEC-6 cells were used to evaluate the role of NCOA4 in the effect of corilagin against II/R injury. Temporal and nucleoplasmic variations with or without corilagin were observed by WB. Co-IP and molecular docking were used to investigate the NCOA4-ferritin interaction. KEY FINDINGS Corilagin attenuated II/R-induced ferroptosis both in vitro and in vivo. Further study revealed that the anti-ferroptosis bioactivity of corilagin might be due to the modulation of iron homeostasis via inhibition of ferritinophagy in an NCOA4-dependent manner. SIGNIFICANCE Corilagin might be a potential therapeutic agent for II/R-induced tissue injury.
Collapse
Affiliation(s)
- Yunxiang Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China
| | - Guanting Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Qipeng Han
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China.
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China; Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China.
| |
Collapse
|
16
|
Zhang P, Wang Y, Wang J, Li G, Li S, Ma J, Peng X, Yin J, Liu Y, Zhu Y. Transcriptomic and physiological analyses reveal changes in secondary metabolite and endogenous hormone in ginger (Zingiber officinale Rosc.) in response to postharvest chilling stress. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 201:107799. [PMID: 37271022 DOI: 10.1016/j.plaphy.2023.107799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/08/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023]
Abstract
Storing postharvest ginger at low temperatures can extend its shelf life, but can also lead to chilling injury, loss of flavor, and excessive water loss. To investigate the effects of chilling stress on ginger quality, morphological, physiological, and transcriptomic changes were examined after storage at 26 °C, 10 °C, and 2 °C for 24 h. Compared to 26 °C and 10 °C, storage at 2 °C significantly increased the concentrations of lignin, soluble sugar, flavonoids, and phenolics, as well as the accumulation of H2O2, O2-, and thiobarbituric acid reactive substances (TBARS). Additionally, chilling stress inhibited the levels of indoleacetic acid, while enhancing gibberellin, abscisic acid, and jasmonic acid, which may have increased postharvest ginger's adaptation to chilling. Storage at 10 °C decreased lignin concentration and oxidative damage, and induced less fluctuant changes in enzymes and hormones than storage at 2 °C. RNA-seq revealed that the number of differentially expressed genes (DEGs) increased with decreasing temperature. Functional enrichment analysis of the 523 DEGs that exhibited similar expression patterns between all treatments indicated that they were primarily enriched in phytohormone signaling, biosynthesis of secondary metabolites, and cold-associated MAPK signaling pathways. Key enzymes related to 6-gingerol and curcumin biosynthesis were downregulated at 2 °C, suggesting that cold storage may negatively impact ginger quality. Additionally, 2 °C activated the MKK4/5-MPK3/6-related protein kinase pathway, indicating that chilling may increase the risk of ginger pathogenesis.
Collapse
Affiliation(s)
- Pan Zhang
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Yanhong Wang
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Jie Wang
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Gang Li
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Siyun Li
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Jiawei Ma
- Jingzhou Jiazhiyuan Biotechnology Co. Ltd., Jingzhou, 434025, Hubei, China
| | - Xiangyan Peng
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Junliang Yin
- College of Agriculture, Yangtze University, Jingzhou, 434025, Hubei, China
| | - Yiqing Liu
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China.
| | - Yongxing Zhu
- Spice Crops Research Institute, College of Horticulture and Gardening, Yangtze University, Jingzhou, 434025, Hubei, China.
| |
Collapse
|
17
|
Gu L, Wang F, Wang Y, Sun D, Sun Y, Tian T, Meng Q, Yin L, Xu L, Lu X, Peng J, Lin Y, Sun P. Naringin protects against inflammation and apoptosis induced by intestinal ischemia-reperfusion injury through deactivation of cGAS-STING signaling pathway. Phytother Res 2023; 37:3495-3507. [PMID: 37125528 DOI: 10.1002/ptr.7824] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/02/2023]
Abstract
Effective amelioration of ischemia/reperfusion (I/R)-induced intestinal injury and revealing its mechanisms remain the challenges in both preclinic and clinic. Potential mechanisms of naringin in ameliorating I/R-induced intestinal injury remain unknown. Based on pre-experiments, I/R-injured rat intestine in vivo and hypoxia-reoxygenation (H/R)-injured IEC-6 cells in vitro were used to verify that naringin-alleviated I/R-induced intestinal injury was mediated via deactivating cGAS-STING signaling pathway. Naringin improved intestinal damage using hematoxylin and eosin staining and decreased alanine aminotransferase and aspartate aminotransferase contents in plasma. Naringin decreased inflammation characterized by reducing IL-6, IL-1β, TNF-α, and IFN-β contents in both plasma and IEC-6 cells. Naringin mitigated oxidative stress via recovering superoxide dismutase, glutathione, and malondialdehyde levels in the I/R-injured intestine. Naringin reduced the expression of apoptotic proteins, including Bax, caspase-3, and Bcl-2, and reduced terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling-positive cells both in vivo and in vitro, and decreased Hoechst 33342 signals in vitro. cGAS, STING, p-TBK1, p-IRF3, and NF-κB expressions were up-regulated both in vivo and in vitro respectively and the up-regulated indexes were reversed by naringin. Transfection of cGAS-siRNA and cGAS-cDNA significantly down-regulated and up-regulated cGAS-STING signaling-related protein expressions, respectively, and partially weakened naringin-induced amelioration on these indexes, suggesting that deactivation of cGAS-STING signaling is the crucial target for naringin-induced amelioration on I/R-injured intestine.
Collapse
Affiliation(s)
- Lidan Gu
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Fei Wang
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yilin Wang
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Deen Sun
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yiming Sun
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Tingting Tian
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| | - Lianhong Yin
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| | - Lina Xu
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| | - Xiaolong Lu
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| | - Jinyong Peng
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| | - Yuan Lin
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Pengyuan Sun
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, China
| |
Collapse
|
18
|
Fang Q, Yu L, Tian F, Zhang H, Chen W, Zhai Q. Effects of dietary irritants on intestinal homeostasis and the intervention strategies. Food Chem 2023; 409:135280. [PMID: 36587512 DOI: 10.1016/j.foodchem.2022.135280] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Abundant diet components are unexplored as vital factors in intestinal homeostasis. Dietary irritants stimulate the nervous system and provoke somatosensory responses, further inducing diarrhea, gut microbiota disorder, intestinal barrier damage or even severe gastrointestinal disease. We depicted the effects of food with piquancy, high fat, low pH, high-refined carbohydrates, and indigestible texture. The mechanism of dietary irritants on intestinal homeostasis were comprehensively summarized. Somatosensory responses to dietary irritants are palpable and have specific chemical and neural mechanisms. In contrast, even low-dose exposure to dietary irritants can involve multiple intestinal barriers. Their mechanisms in intestinal homeostasis are often overlapping and dose-dependent. Therefore, treating symptoms caused by dietary irritants requires personalized nutritional advice. The reprocessing of stimulant foods, additional supplementation with probiotics or prebiotics, and enhancement of the intestinal barrier are effective intervention strategies. This review provides promising preliminary guidelines for the treatment of symptoms and gastrointestinal injury caused by dietary irritants.
Collapse
Affiliation(s)
- Qingying Fang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute, Wuxi Branch, PR China; Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, PR China; Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, PR China.
| |
Collapse
|
19
|
Yeo EJ, Shin MJ, Youn GS, Park JH, Yeo HJ, Kwon HJ, Lee LR, Kim NY, Kwon SY, Kim SM, Lee J, Lee KW, Lee CH, Cho YJ, Kwon OS, Kim DW, Jung HY, Eum WS, Choi SY. Tat-RAN attenuates brain ischemic injury in hippocampal HT-22 cells and ischemia animal model. Neurochem Int 2023; 167:105538. [PMID: 37207854 DOI: 10.1016/j.neuint.2023.105538] [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: 02/16/2023] [Revised: 04/30/2023] [Accepted: 05/07/2023] [Indexed: 05/21/2023]
Abstract
Oxidative stress plays a key role in the pathogenesis of neuronal injury, including ischemia. Ras-related nuclear protein (RAN), a member of the Ras superfamily, involves in a variety of biological roles, such as cell division, proliferation, and signal transduction. Although RAN reveals antioxidant effect, its precise neuroprotective mechanisms are still unclear. Therefore, we investigated the effects of RAN on HT-22 cell which were exposed to H2O2-induced oxidative stress and ischemia animal model by using the cell permeable Tat-RAN fusion protein. We showed that Tat-RAN transduced into HT-22 cells, and markedly inhibited cell death, DNA fragmentation, and reactive oxygen species (ROS) generation under oxidative stress. This fusion protein also controlled cellular signaling pathways, including mitogen-activated protein kinases (MAPKs), NF-κB, and apoptosis (Caspase-3, p53, Bax and Bcl-2). In the cerebral forebrain ischemia animal model, Tat-RAN significantly inhibited both neuronal cell death, and astrocyte and microglia activation. These results indicate that RAN significantly protects against hippocampal neuronal cell death, suggesting Tat-RAN will help to develop the therapies for neuronal brain diseases including ischemic injury.
Collapse
Affiliation(s)
- Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Gi Soo Youn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Jung Hwan Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Hyun Jung Kwon
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Lee Re Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Na Yeon Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Su Yeon Kwon
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Su Min Kim
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Jaehak Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Chan Hee Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University Medical Center, Chuncheon, 24253, South Korea
| | - Oh-Shin Kwon
- School of Life Sciences, College of Natural Sciences Kyungpook National University, Taegu, 41566, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - Hyo Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
20
|
Foshati S, Poursadeghfard M, Heidari Z, Amani R. The effect of ginger ( Zingiber officinale) supplementation on clinical, biochemical, and anthropometric parameters in patients with multiple sclerosis: a double-blind randomized controlled trial. Food Funct 2023; 14:3701-3711. [PMID: 36974730 DOI: 10.1039/d3fo00167a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Introduction: different lines of evidence have shown that ginger administration may be beneficial for patients with multiple sclerosis (MS). Therefore, we aimed to investigate the effect of ginger supplementation on disability, physical and psychological quality of life (QoL), body mass index (BMI), neurofilament light chain (NfL), interlukin-17 (IL-17), matrix metalloproteinase-9 (MMP-9), and neutrophil to lymphocyte ratio (NLR) in patients with relapsing-remitting MS. Methods: this was a 12 week double-blind parallel randomized placebo-controlled trial with a 3 week run-in period. The treatment (n = 26) and control (n = 26) groups received 500 mg ginger and placebo (corn) supplements 3 times daily, respectively. Disability was evaluated using the Expanded Disability Status Scale (EDSS). QoL was rated using the Multiple Sclerosis Impact Scale (MSIS-29). BMI was calculated by dividing weight by height squared. Serum levels of NfL, IL-17, and MMP-9 were measured using the enzyme-linked immunosorbent assay. NLR was determined using a Sysmex XP-300™ automated hematology analyzer. All outcomes were assessed before and after the intervention and analyzed using the intention-to-treat principle. Results: in comparison with placebo, ginger supplementation caused a significant reduction in the EDSS (-0.54 ± 0.58 vs. 0.08 ± 0.23, P < 0.001), the MSIS-29 physical scale (-8.15 ± 15.75 vs. 4.23 ± 8.46, P = 0.001), the MSIS-29 psychological scale (-15.71 ± 19.59 vs. 6.68 ± 10.41, P < 0.001), NfL (-0.14 ± 0.97 vs. 0.38 ± 1.06 ng mL-1, P = 0.049), IL-17 (-3.34 ± 4.06 vs. 1.77 ± 6.51 ng L-1, P = 0.003), and NLR (-0.09 ± 0.53 vs. 0.53 ± 1.90, P = 0.038). Nevertheless, the differences in BMI and MMP-9 were not significant between the groups. Conclusion: ginger supplementation may be an effective adjuvant therapy for patients with relapsing-remitting MS.
Collapse
Affiliation(s)
- Sahar Foshati
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Poursadeghfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Heidari
- Department of Biostatistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Amani
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
21
|
Qian W, Li W, Chen X, Cui L, Liu X, Yao J, Wang X, Liu Y, Li C, Wang Y, Wang W. Exploring the mechanism of Xingpi Capsule in diarrhea predominant-irritable bowel syndrome treatment based on multiomics technology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154653. [PMID: 36641976 DOI: 10.1016/j.phymed.2023.154653] [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: 08/27/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Xingpi Capsule (XP), a commercially available over-the-counter herbal medicine in China, plays a prominent role in treating diarrhea-predominant irritable bowel syndrome (IBS-D). Nevertheless, the potential mechanisms remain unclear. PURPOSE This study aimed to investigate XP efficacy in IBS-D and elucidate the underlying molecular mechanisms. METHODS A rat IBS-D model was established by senna decoction gavage combined with restraint stress and swimming exhaustion. The changes in rat body weight and stool were recorded daily. Colon pathological changes and the number of colonic goblet cells of rats were observed by hematoxylin-eosin (HE) staining and Alcian blue plus periodic acid-Schiff (AB-PAS) staining, respectively. The expression of Occludin, a tight-junction-associated protein, was examined via immunohistochemistry. Images of colonic microvilli were obtained by TEM. Western blotting (WB) was used to analyze the protein expression of the ASK1/P38 MAPK pathway. The composition of the rat intestinal microbiota was detected by 16S rRNA sequencing. Changes in colonic metabolites were evaluated by liquid chromatography-mass spectrometry (LC-MS). Changes in colon RNA expression were assessed by RNA sequencing (RNA-Seq). The nontoxic range of hypoxanthine (HPX) was screened by Cell Counting Kit-8 (CCK8), the cell model of human colonic epithelial cells (NCM460) induced by lipopolysaccharide (LPS) was established, and the effective concentration of HPX was screened by CCK8. After transfection of pcDNA3.1-MAP3K5, Hoechst 33,342 staining, flow cytometry to detect cell apoptosis, and immunofluorescence to detect the fluorescence changes of ASK1 and ZO-1. WB detection of ASK1/P38 MAPK pathway protein expression changes. RESULTS XP increased the body weight of IBS-D patients and reduced the loose stool rate, loose stool index, and Bristo score. In addition, XP mitigated colon lesions, increased the number of goblet cells and the expression of Occludin, and prevented severe distortion and effacement of the microvillous structure. Specifically, 16S rRNA gene sequence analysis showed that XP decreased the abundance of Desulfurium and Prevotella 9 at the phylum and genus levels while increasing the abundance of Bacteroides at the genus level. RNA-Seq combined with WB validation showed that XP exerted antidiarrheal effects by inhibiting the ASK1/P38 MAPK signaling pathway. Additionally, XP also increased the relative expression level of the metabolite HPX, as revealed by untargeted metabolomics analysis. Impressively, the correlation analysis between 16S rRNA sequencing and LC-MS suggested that HPX and Prevotella 9 are negatively correlated, which indicated that XP might increase the content of HPX by reducing the abundance of Prevotella 9. Meanwhile, a negative correlation between HPX and ASK1 was indicated through RNA-Seq and LC-MS, which suggested that the inhibition of ASK1 (Map3k5) may be ascribed to the increase in HPX after XP treatment. In vitro experiments have proven that HPX can alleviate LPS-induced NCM460 damage, specifically manifested as enhancing cell viability, reducing cell apoptosis, increasing ZO-1 expression, reducing the fluorescence intensity of MAP3K5 in the model group, and inhibiting the expression of ASK1/P38 MAPK pathway proteins. The protective effect of HPX was reversed after transfection with pcDNA 3.1-MAP3K5, which fully demonstrated that the protective mechanism of HPX was achieved by inhibiting MAP3K5 and its downstream pathways. CONCLUSION XP displayed multifaceted protection against IBS-D in rats by regulating the intestinal microbiota, increasing the relative expression level of HPX, a metabolite of the microbiota, and inhibiting the ASK1/P38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Weina Qian
- School of Basic Medical Sciences, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Weili Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoyang Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Lingwen Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiangning Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Junkai Yao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoping Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yizhou Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Wei Wang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Beijing Key Laboratory of Syndrome and Formula, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China.
| |
Collapse
|
22
|
Li B, Li W, Zheng M, Wang Y, Diao Y, Mou X, Liu J. Corilagin alleviates intestinal ischemia/reperfusion injury by relieving oxidative stress and apoptosis via AMPK/Sirt1-autophagy pathway. Exp Biol Med (Maywood) 2023; 248:317-326. [PMID: 36680375 PMCID: PMC10159520 DOI: 10.1177/15353702221147560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Intestinal ischemia/reperfusion (II/R) injury is a common pathological process with high clinical morbidity and mortality. Autophagy plays an important role in the pathological development of II/R. Corilagin (CA) is a natural ellagitannin with various pharmacological effects such as autophagy regulation, antioxidant, and antiapoptosis. However, whether CA alleviates II/R injury is still unclear. In this study, we had found that CA significantly attenuated II/R induced intestinal tissue pathological damage, oxidative stress, and cell apoptosis in rats. Further studies showed that CA significantly promoted AMPK phosphorylation and sirt1 expression, and thus activated autophagy by upregulating protein expression of autophagy-related proteins Beclin1 and LC3II and promoting SQSTM1/P62 degradation both in vivo and in vitro. Inhibition of AMPK phosphorylation by its inhibitor compound C(CC) significantly abolished CA-mediated autophagy activation and the relievable effects on oxidative stress and apoptosis in vitro, suggesting the excellent protective activity of CA against II/R injury via AMPK/Sirt1-autophagy pathway. These findings confirmed the potent effects of CA against II/R injury, and provided novel insights into the mechanisms of the compound as a potential candidate for the treatment of II/R.
Collapse
Affiliation(s)
- Bin Li
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China
| | - Wenlian Li
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Meiling Zheng
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yunxiang Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China
| | - Xiaojuan Mou
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian 116044, China
| |
Collapse
|
23
|
Gumbarewicz E, Jarząb A, Stepulak A, Kukula-Koch W. Zingiber officinale Rosc. in the Treatment of Metabolic Syndrome Disorders-A Review of In Vivo Studies. Int J Mol Sci 2022; 23:15545. [PMID: 36555184 PMCID: PMC9779757 DOI: 10.3390/ijms232415545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a protective reaction of the innate immune system as a response to imbalances caused by a specific stimulus, a disease or a pathogen. A prolonged inflammatory condition may lead to the development of metabolic syndrome, which affects more than one-fourth of the world's population. This condition leads to the development of multi-organ disorders based on disrupted blood lipid and sugar levels, hypertension and oxidative stress. The review aims to present Zingiber officinale Rosc. as a plant that exhibits a variety of healing properties and restores the organism's equilibrium. Ginger (GI) rhizomes have been commonly used in traditional medicine to treat arthritis, stomach ache, nonalcoholic fatty liver disease, rheumatism, nervous system syndromes, asthma, diabetes and nausea caused by pregnancy or chemotherapy. This review gathers together data from in vivo experiments related to the application of ginger for the treatment of inflammatory conditions, obesity, diabetes and other related disorders as a consequence of metabolic syndrome, including the confirmed molecular mechanisms of action.
Collapse
Affiliation(s)
- Ewelina Gumbarewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Agata Jarząb
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland
| |
Collapse
|
24
|
Ganjikunta VS, Maddula RR, Bhasha S, Sahukari R, Kondeti Ramudu S, Chenji V, Kesireddy SR, Zheng Z, Korivi M. Cardioprotective Effects of 6-Gingerol against Alcohol-Induced ROS-Mediated Tissue Injury and Apoptosis in Rats. Molecules 2022; 27:8606. [PMID: 36500700 PMCID: PMC9738005 DOI: 10.3390/molecules27238606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
The present study investigated the cardioprotective properties of 6-gingerol against alcohol-induced ROS-mediated cardiac tissue damage in rats. Experiments were conducted on 4 groups of rats, orally treated with control, 6-gingerol (10 mg/kg body weight), alcohol (6 g/kg body weight) and combination of 6-gingerol plus alcohol for two-month. In the results, we found 6-ginger treatment to alcohol-fed rats substantially suppressed ROS production in cardiac tissue. Alcohol-induced elevated 8-OHDG and protein carbonyls which represent oxidative modification of DNA and proteins were completely reversed by 6-gingerol. This was further endorsed by restored superoxide dismutase and catalase activities with 6-gingerol against alcohol-induced loss. The elevated cardiac biomarkers (CK-MB, cTn-T, cTn-I) and dyslipidemia in alcohol-intoxicated rats was significantly reversed by 6-gingerol. Furthermore, alcohol-induced apoptosis characterized by overexpression of cytochrome C, caspase-8 and caspase-9 was diminished with 6-gingerol treatment. Transmission electron microscope images conferred the cardioprotective properties of 6-gingerol as we have seen less structural derangements in mitochondria and reappearance of myofilaments. Our findings conclude that 6-ginger effectively protect alcohol-induced ROS-mediated cardiac tissue damage, which may be due to its potent antioxidant efficacy. Therefore, 6-gingerol could be a potential therapeutic molecule that can be used in the treatment of alcohol-induced myocardial injury.
Collapse
Affiliation(s)
| | - Ramana Reddy Maddula
- Division of Molecular Biology and Ethanopharmacology, Sri Venkateswara University, Tirupati 517 502, India
| | - Shanmugam Bhasha
- Division of Molecular Biology and Ethanopharmacology, Sri Venkateswara University, Tirupati 517 502, India
| | - Ravi Sahukari
- Division of Molecular Biology and Ethanopharmacology, Sri Venkateswara University, Tirupati 517 502, India
| | | | - Venkatrayulu Chenji
- Department of Marine Biology, Vikarama Simhapuri University, Nellore 524320, India
| | - Sathyavelu Reddy Kesireddy
- Division of Molecular Biology and Ethanopharmacology, Sri Venkateswara University, Tirupati 517 502, India
| | - Zhe Zheng
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Mallikarjuna Korivi
- Exercise and Metabolism Research Center, College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, China
| |
Collapse
|
25
|
Phenol-Rich Botanicals Modulate Oxidative Stress and Epithelial Integrity in Intestinal Epithelial Cells. Animals (Basel) 2022; 12:ani12172188. [PMID: 36077907 PMCID: PMC9454507 DOI: 10.3390/ani12172188] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 12/16/2022] Open
Abstract
Botanicals are mainly known for their role as antimicrobials and anti-inflammatories. Thus, the dual purpose of the study was to verify the antioxidant potential of the tested botanicals and to evaluate their possible modulation of intestinal barrier integrity. As the effects of various phenol-rich extracts were screened, the human Caco-2 cell line was determined to be most suitable for use as the in vitro model for the intestinal epithelium. The tested botanicals, all approved as feed additives, are ginger essential oil, tea tree oil, grape seed extract, green tea extract, olive extract, chestnut extract, pomegranate extract, thyme essential oil, and capsicum oleoresin. The cells were treated with incremental doses of each botanical, followed by measurements of transepithelial electrical resistance (TEER), gene expression of tight junctions (TJs), and reactive oxygen species (ROS). The results showed how different phenol-rich botanicals could modulate barrier functions and oxidative stress in different ways. Interestingly, all the botanicals tested exerted an antioxidant potential by dropping the cytoplasmatic ROS, while the beneficial effect was exerted at different concentrations for each botanical. Our data support the role of plant extracts and essential oils in controlling gut barrier function and in reducing the negative effects of oxidative stress in intestinal epithelial cells, thereby supporting gut barrier functionality.
Collapse
|
26
|
Wan Y, Dong P, Zhu X, Lei Y, Shen J, Liu W, Liu K, Zhang X. Bibliometric and visual analysis of intestinal ischemia reperfusion from 2004 to 2022. Front Med (Lausanne) 2022; 9:963104. [PMID: 36052333 PMCID: PMC9426633 DOI: 10.3389/fmed.2022.963104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
Background Intestinal ischemia/reperfusion (I/R) injury is a common tissue-organ damage occurring in surgical practice. This study aims to comprehensively review the collaboration and impact of countries, institutions, authors, subject areas, journals, keywords, and critical literature on intestinal I/R injury from a bibliometric perspective, and to assess the evolution of clustering of knowledge structures and identify hot trends and emerging topics. Methods Articles and reviews related to intestinal I/R were retrieved through subject search from Web of Science Core Collection. Bibliometric analyses were conducted on Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio). Results A total of 1069 articles and reviews were included from 2004 to 2022. The number of articles on intestinal I/R injury gradually plateaued, but the number of citations increased. These publications were mainly from 985 institutions in 46 countries, led by China and the United States. Liu Kx published the most papers, while Chiu Cj had the largest number of co-citations. Analysis of the journals with the most outputs showed that most journals focused on surgical sciences, cell biology, and immunology. Macroscopic sketch and microscopic characterization of the entire knowledge domain were achieved through co-citation analysis. The roles of cell death, exosomes, intestinal flora, and anesthetics in intestinal I/R injury are the current and developing research focuses. The keywords "dexmedetomidine", "proliferation", and "ferroptosis" may also become new trends and focus of future research. Conclusion This study comprehensively reviews the research on intestinal I/R injury using bibliometric and visualization methods, and will help scholars better understand the dynamic evolution of intestinal I/R injury and provide directions for future research.
Collapse
Affiliation(s)
- Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Xiaobing Zhu
- Department of Anesthesiology, Hospital of Traditional Chinese Medicine of Zhongshan City, Zhongshan, China
| | - Yuqiong Lei
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Junyi Shen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Weifeng Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Kexuan Liu
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| | - Xiyang Zhang
- Department of Anesthesiology, Nan Fang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Wen Y, Wang W, Ni Y, Gui Y, Hu Z, Wang Y, Han M, Zhang D, Shu S, Zhou S. Dose-effect relationship of ginger interposed moxibustion for allergic rhinitis: study protocol for a randomized, placebo-controlled and parallel clinical trial. Eur J Integr Med 2022. [DOI: 10.1016/j.eujim.2022.102166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
28
|
Wang W, Gu W, He C, Zhang T, Shen Y, Pu Y. Bioactive components of Banxia Xiexin Decoction for the treatment of gastrointestinal diseases based on flavor-oriented analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115085. [PMID: 35150814 DOI: 10.1016/j.jep.2022.115085] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD) was first recorded in a Chinese medical classic, Treatise on Febrile Diseases and Miscellaneous Diseases, which was written in the Eastern Han dynasty of China. This ancient prescription consists of seven kinds of Chinese herbal medicine, namely, Pinellia ternata, Rhizoma Coptidis, Radix scutellariae, Rhizoma Zingiberis, Ginseng, Jujube, and Radix Glycyrrhizaepreparata. In clinic practice, its original application in China mainly has focused on the treatment of chronic gastritis for several hundred years. BXD is also effective in treating other gastrointestinal diseases (GIDs) in modern medical application. Despite available literature support and clinical experience, the treatment mechanisms or their relationships with the bioactive compounds in BXD responsible for its pharmacological actions, still need further explorations in more diversified channels. According to the analysis based on the five-flavor theory of TCM, BXD is traditionally viewed as the most representative prescription for pungent-dispersion, bitter-purgation and sweet-tonification. Consequently, based on the flavor-oriented analysis, the compositive herbs in BXD can be divided into three flavor groups, namely, the pungent, bitter, and sweet groups, each of which has specific active ingredients that are possibly relevant to GID treatment. AIM OF THE REVIEW This paper summarized recent literatures on BXD and its bioactive components used in GID treatment, and provided the pharmacological or chemical basis for the further exploration of the ancient prescription and the relative components. METHOD ology: Relevant literature was collected from various electronic databases such as Pubmed, Web of Science, and China National Knowledge Infrastructure (CNKI). Citations were based on peer-reviewed articles published in English or Chinese during the last decade. RESULTS Multiple components were found in the pungent, bitter, and sweet groups in BXD. The corresponding bioactive components include gingerol, shogaol, stigmasterol, and β-sitosterol in the pungent group; berberine, palmatine, coptisine, baicalein, and baicalin in the bitter group; and ginsenosides, polysaccharides, liquiritin, and glycyrrhetinic acid in the sweet group. These components have been found directly or indirectly responsible for the remarkable effects of BXD on GID. CONCLUSION This review provided some valuable reference to further clarify BXD treatment for GID and their possible material basis, based on the perspective of the flavor-oriented analysis.
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
| | - Weiliang Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao He
- 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
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yao Shen
- Shanghai Center of Biomedicine Development, 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
|
29
|
Shin SH, Oh SM, Yoon Park JH, Lee KW, Yang H. OptNCMiner: a deep learning approach for the discovery of natural compounds modulating disease-specific multi-targets. BMC Bioinformatics 2022; 23:218. [PMID: 35672685 PMCID: PMC9175487 DOI: 10.1186/s12859-022-04752-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022] Open
Abstract
Background Due to their diverse bioactivity, natural product (NP)s have been developed as commercial products in the pharmaceutical, food and cosmetic sectors as natural compound (NC)s and in the form of extracts. Following administration, NCs typically interact with multiple target proteins to elicit their effects. Various machine learning models have been developed to predict multi-target modulating NCs with desired physiological effects. However, due to deficiencies with existing chemical-protein interaction datasets, which are mostly single-labeled and limited, the existing models struggle to predict new chemical-protein interactions. New techniques are needed to overcome these limitations. Results We propose a novel NC discovery model called OptNCMiner that offers various advantages. The model is trained via end-to-end learning with a feature extraction step implemented, and it predicts multi-target modulating NCs through multi-label learning. In addition, it offers a few-shot learning approach to predict NC-protein interactions using a small training dataset. OptNCMiner achieved better prediction performance in terms of recall than conventional classification models. It was tested for the prediction of NC-protein interactions using small datasets and for a use case scenario to identify multi-target modulating NCs for type 2 diabetes mellitus complications. Conclusions OptNCMiner identifies NCs that modulate multiple target proteins, which facilitates the discovery and the understanding of biological activity of novel NCs with desirable health benefits.
Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04752-5.
Collapse
Affiliation(s)
- Seo Hyun Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Man Oh
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung Han Yoon Park
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 08826, Republic of Korea. .,Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea. .,Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Hee Yang
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
30
|
Feng YD, Ye W, Tian W, Meng JR, Zhang M, Sun Y, Zhang HN, Wang SJ, Wu KH, Liu CX, Liu SY, Cao W, Li XQ. Old targets, new strategy: Apigenin-7-O-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic Biol Med 2022; 184:74-88. [PMID: 35398494 DOI: 10.1016/j.freeradbiomed.2022.03.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022]
Abstract
With the increasing morbidity and mortality, intestinal ischemia/reperfusion injury (IIRI) has attracted more and more attention, but there is no efficient therapeutics at present. Apigenin-7-O-β-D-(-6″-p-coumaroyl)-glucopyranoside (APG) is a new flavonoid glycoside isolated from Clematis tangutica that has strong antioxidant abilities in previous studies. However, the pharmacodynamic function and mechanism of APG on IIRI remain unknown. This study aimed to investigate the effects of APG on IIRI both in vivo and in vitro and identify the potential molecular mechanism. We found that APG could significantly improve intestinal edema and increase Chiu's score. MST analysis suggested that APG could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis with a dose-dependent manner. Moreover, we used siRNA silencing technology to confirm that knocking down both HO-1 and MAO-B had a positive effect on intestine. In addition, we found the HO-1 and MAO-B inhibitors also could reduce endothelial cell loss and protect vascular endothelial after reperfusion. We demonstrate that APG plays a protective role on decreasing activation of HO-1 and MAO-B, attenuating IIRI-induced ROS generation and Fe2+ accumulation, maintaining mitochondria function thus inhibiting ferroptosis.
Collapse
Affiliation(s)
- Ying-Da Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Hui-Nan Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shou-Jia Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Ke-Han Wu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chen-Xu Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shao-Yuan Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
31
|
Lyu H, Ni H, Huang J, Yu G, Zhang Z, Zhang Q. VX-765 prevents intestinal ischemia-reperfusion injury by inhibiting NLRP3 inflammasome. Tissue Cell 2022; 75:101718. [PMID: 35131633 DOI: 10.1016/j.tice.2021.101718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intestinal ischemia-reperfusion injury (IIRI) is a common clinical event that can cause serious consequences. The study aimed to investigated the effect of VX-765 in IIRI and its mechanism. METHODS The hypoxia-reoxygenation (H/R) cell model and IIRI mouse model were generated to examine the in vitro and in vivo effects of VX-765 on IIRI. IIRI was evaluated by histological assessment. ELISA was performed to determine the levels of IL-6, TNF-α, IL-1β, caspase-1, and GSDMD in intestinal tissues as well as the levels of MDA, SOD, CAT, caspase-1, and GSDMD in Caco-2 cells. Relative protein levels of NLRP3, ASC, IL-18, IL-1β, cleaved Caspase1, and GSDMD-N were analyzed by Western blotting. CCK-8 Assay was conducted to determine the optimal concentration of VX-765 for the in vitro studies. Flow cytometry, fluorescence microscopy and real-time PCR (RT-PCR) were used to assess ROS levels and the mRNA levels of IL-18 and IL-1β, respectively. Immunofluorescence staining was performed to examine the subcellular localization of P65 and NLRP3. RESULTS VX-765 reduced IIRI-induced oxidative stress and inflammatory response both in vivo and in vitro, while it decreased the levels of TNF-α, IL-6, IL-1β as well as the modified Park/Chiu scores. The optimal concentration of VX-765 for the in vitro studies was 10 μM. Moreover, VX-765 inhibited the nuclear translocation of P65, reduced oxidative stress and down-regulated the activation of NLRP3 inflammasome. CONCLUSION VX-765 prevents IIRI presumably by inhibiting the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Heping Lyu
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Haizhen Ni
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Jingyong Huang
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Guanfeng Yu
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Zhongjing Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China
| | - Qiyu Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, PR China.
| |
Collapse
|
32
|
6-Gingerol exerts a protective effect against hypoxic injury through the p38/Nrf2/HO-1 and p38/NF-κB pathway in H9c2 cells. J Nutr Biochem 2022; 104:108975. [PMID: 35245652 DOI: 10.1016/j.jnutbio.2022.108975] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/02/2021] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Ginger, one of the most widely consumed condiment for various foods and beverages, has many pharmacological effects. 6-gingerol, a naturally occurring phenol, is one of the major pungent constituents of ginger. The purpose of this study was to characterize the effect of 6-gingerol on the p38/Nrf2/HO-1 and p38/NF-κB signaling pathway, as a possible means of combating hypoxia-related oxidative stress. H9c2 cells were chemically induced with CoCl2 to mimic hypoxia-associated cellular damage. Cardiomyocyte injury was assessed by lactate dehydrogenase and creatine kinase. Reactive oxygen species production was assessed by 2',7'-dichlorodihydrofluorescein diacetate. The antioxidative property of 6-gingerol was measured by estimating the activities of superoxide dismutase, catalase, glutathione and glutathione disulfide. Apoptosis was detected by flow cytometry after Annexin V-FITC-propidium iodide double staining. Western blotting was used to evaluate levels of p-p38, p38, cytoplasm p65, nuclear p65, total p65, nuclear Nrf2, total Nrf2, Keap1, HIF-1α, and HO-1. 6-gingerol was able to counter hypoxia-induced cardiomyocyte injury as evidenced by inhibiting the levels of oxidative stress indexes and increasing the percentage of apoptosis. Furthermore, 6-gingerol was able to down-regulate p-p38/p38, nuclear p65, total p65 and Keap1 expression induced by CoCl2 stimulation and increased cytoplasm p65, nuclear Nrf2, total Nrf2, HO-1, and HIF-1α expression. However, treatment with specific Nrf2 inhibitor blunted the activation of Nrf2 signaling and removed the protective effects of 6-gingerol. These experiments provide evidence that 6-gingerol exerts cytoprotective effects, which may be associated with the regulation of oxidative stress and apoptosis, potentially through activating the Nrf2 pathway and inhibiting the p38/NF-κB pathways.
Collapse
|
33
|
Sun C, Han B, Zhai Y, Zhao H, Li X, Qian J, Hao X, Liu Q, Shen J, Kai G. Dihydrotanshinone I inhibits ovarian tumor growth by activating oxidative stress through Keap1-mediated Nrf2 ubiquitination degradation. Free Radic Biol Med 2022; 180:220-235. [PMID: 35074488 DOI: 10.1016/j.freeradbiomed.2022.01.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
Dihydrotanshinone I (DHT), a bioactive compound in Salvia miltiorrhiza, was reported to exhibit cytotoxicity against various malignancies. However, the underlying mechanism on ovarian cancer remains unclear. Here, DHT inhibited cell viability of ovarian cancer HO8910PM, SKOV3, A2780 and ES2 cells. It showed moderate inhibitory effect on ovarian epithelial IOSE80 cells and lower toxicity than chemotherapy drugs. DHT induced apoptosis and G2 cell cycle arrest accompanied by reduced expression of Bcl-2, Caspase-3, and increased Bax. Meanwhile, DHT increased ROS accumulation, decreased mitochondrial membrane potential and activated oxidative stress in HO8910PM and ES2 cells. Mechanistically, DHT inhibited Nrf2 and p62 expression, Nrf2 target genes and enzymes, and Nrf2 nuclear translocation, while increased the expression of Nrf2 inhibitor Keap1. NAC, a ROS scavenger, rescued DHT-induced proliferation inhibition, ROS generation and Nrf2 inhibition. DHT alleviated tBHQ-induced Nrf2 expression and increased its mRNA level. However, the proteasome inhibitor MG132 blocked DHT-induced Nrf2 inhibition, suggesting a post-translational regulation manner. DHT enhanced Nrf2 binding with Keap1, leading to potentiated Nrf2 ubiquitination degradation. Furthermore, Nrf2 and p62 overexpression blocked DHT-induced Nrf2 and p62 inhibition. Consistent with the in vitro results, DHT significantly delayed tumor growth in HO8910PM and ES2 xenograft nude mice, decreased tumor marker HE4 and CA125 levels, reversed the abnormally expressed proteins including Ki67, Nrf2, p62, Keap1, Bcl-2, CyclinB1, Cdc-2, and antioxidant enzymes SOD, CAT in vivo. Serum from DHT-treated mice also inhibited cell growth in vitro. Taken together, DHT exhibits anti-ovarian tumor effect by activating oxidative stress through ubiquitination-mediated Nrf2 degradation. Our findings implicate a potential application of DHT for ovarian cancer therapy.
Collapse
Affiliation(s)
- Chengtao Sun
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bing Han
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yufei Zhai
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huan Zhao
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xuan Li
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jun Qian
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaolong Hao
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qun Liu
- Institute of Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden Mem. Sun Yat-Sen), Nanjing, 210014, China
| | - Jiayan Shen
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Guoyin Kai
- Laboratory for Core Technology of TCM Quality Improvement and Transformation, College of Pharmaceutical Science, The Third Affiliated Hospital, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
34
|
Chen C, Yu LT, Cheng BR, Xu JL, Cai Y, Jin JL, Feng RL, Xie L, Qu XY, Li D, Liu J, Li Y, Cui XY, Lu JJ, Zhou K, Lin Q, Wan J. Promising Therapeutic Candidate for Myocardial Ischemia/Reperfusion Injury: What Are the Possible Mechanisms and Roles of Phytochemicals? Front Cardiovasc Med 2022; 8:792592. [PMID: 35252368 PMCID: PMC8893235 DOI: 10.3389/fcvm.2021.792592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is one of the most effective reperfusion strategies for acute myocardial infarction (AMI) despite myocardial ischemia/reperfusion (I/R) injury, causing one of the causes of most cardiomyocyte injuries and deaths. The pathological processes of myocardial I/R injury include apoptosis, autophagy, and irreversible cell death caused by calcium overload, oxidative stress, and inflammation. Eventually, myocardial I/R injury causes a spike of further cardiomyocyte injury that contributes to final infarct size (IS) and bound with hospitalization of heart failure as well as all-cause mortality within the following 12 months. Therefore, the addition of adjuvant intervention to improve myocardial salvage and cardiac function calls for further investigation. Phytochemicals are non-nutritive bioactive secondary compounds abundantly found in Chinese herbal medicine. Great effort has been put into phytochemicals because they are often in line with the expectations to improve myocardial I/R injury without compromising the clinical efficacy or to even produce synergy. We summarized the previous efforts, briefly outlined the mechanism of myocardial I/R injury, and focused on exploring the cardioprotective effects and potential mechanisms of all phytochemical types that have been investigated under myocardial I/R injury. Phytochemicals deserve to be utilized as promising therapeutic candidates for further development and research on combating myocardial I/R injury. Nevertheless, more studies are needed to provide a better understanding of the mechanism of myocardial I/R injury treatment using phytochemicals and possible side effects associated with this approach.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Tong Yu
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bai-Ru Cheng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiang-Lin Xu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yun Cai
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Lin Jin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ru-Li Feng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Long Xie
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Yan Qu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Yun Cui
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Jin Lu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Kun Zhou
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qian Lin
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
- Jie Wan
| |
Collapse
|
35
|
Guo XX, Zhang YD, Wang TC, Wang XL, Xu YY, Wang Y, Qiu J. Ginger and 6-gingerol prevent lipopolysaccharide-induced intestinal barrier damage and liver injury in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:1066-1075. [PMID: 34309869 DOI: 10.1002/jsfa.11442] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Inflammation-related diseases present a significant public health problem. Ginger is a flavoring spice and medicinal herb with anti-inflammatory activity. This study investigated the preventive effects of ginger extract (GE) and its main bioactive component, 6-gingerol (6G), on lipopolysaccharide (LPS)-induced intestinal barrier dysfunction and liver injury in mice. RESULTS GE and 6G were orally administered to mice for seven consecutive days before LPS administration. After 24 h, the mice were sacrificed. GE and 6G were found to significantly reverse LPS-induced inflammation in the mouse ileum by modifying the NF-κB pathway. They also alleviated apoptosis in the ileum by downregulating Bax and cytochrome c gene expression and by inhibiting the caspase-3 pathway. Through the aforementioned mechanisms, GE and 6G restored the intestinal barrier by increasing ZO-1 and claudin-1 protein expressions. Gut-derived LPS induced inflammation and apoptosis in the liver; these effects were markedly reversed through GE and 6G treatment. 6G was the most abundant component in GE, as evidenced through liquid chromatography-mass spectrometry, and accounted for >50% of total gingerols and shogaols in GE. CONCLUSION The current results support the use of GE and 6G as dietary supplements to protect against gut-derived endotoxemia-associated inflammatory response and disorders. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiao-Xuan Guo
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yun-Dan Zhang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Tian-Cai Wang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xin-Lu Wang
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yan-Yang Xu
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yong Wang
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Jing Qiu
- Institute of Quality Standard and Testing Technology for Agro-products, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
36
|
Shen J, Zhan Y, He Q, Deng Q, Li K, Wen S, Huang W. Remifentanil Promotes PDIA3 Expression by Activating p38MAPK to Inhibit Intestinal Ischemia/Reperfusion-Induced Oxidative and Endoplasmic Reticulum Stress. Front Cell Dev Biol 2022; 10:818513. [PMID: 35155431 PMCID: PMC8826554 DOI: 10.3389/fcell.2022.818513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Remifentanil protects against intestinal ischemia/reperfusion (I/R) injury; however, its exact mechanism remains to be elucidated. The objective of this study was to investigate the underlying molecular mechanism of remifentanil in intestinal I/R injury in mice.Methods: We evaluated the intestine-protective effect of remifentanil in adult male mice with 45 min superior mesenteric artery occlusion followed by 4 h reperfusion by determining the following: intestinal Chiu’s scores, diamine oxidase, and intestinal fatty acid binding protein in serum; the apoptotic index, lipid peroxidation product malondialdehyde (MDA), and superoxide dismutase (SOD) activity in the intestinal mucosa; and the intestinal mRNA and protein expressions of Bip, CHOP, caspase-12, and cleaved caspase-3, reflecting endoplasmic reticulum (ER) stress. Furthermore, conditional knockout mice, in which the protein disulfide isomerase A3 (PDIA3) gene was deleted from the intestinal epithelium, and SB203580 (a selective p38MAPK inhibitor) were used to determine the role of PDIA3 and p38MAPK in I/R progression and intestinal protection by remifentanil.Results: Our data showed that intestinal I/R induced obvious oxidative stress and endoplasmic reticulum stress–related cell apoptosis, as evidenced by an increase in the intestinal mucosal malondialdehyde, a decrease in the intestinal mucosal SOD, and an increase in the apoptotic index and the mRNA and protein expression of Bip, CHOP, caspase-12, and cleaved caspase-3. Remifentanil significantly improved these changes. Moreover, the deletion of intestinal epithelium PDIA3 blocked the protective effects of remifentanil. SB203580 also abolished the intestinal protection of remifentanil and downregulated the mRNA and protein expression of PDIA3.Conclusion: Remifentanil appears to act via p38MAPK to protect the small intestine from intestinal I/R injury by its PDIA3-mediated antioxidant and anti-ER stress properties.
Collapse
Affiliation(s)
| | | | | | | | | | - Shihong Wen
- *Correspondence: Shihong Wen, ; Wenqi Huang,
| | - Wenqi Huang
- *Correspondence: Shihong Wen, ; Wenqi Huang,
| |
Collapse
|
37
|
HUA LY, NING Z, YUEBAO N. Determination of SOD in black ginger extract and its effect on the liver of rats with type 2 diabetes. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.115021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Li Yue HUA
- Inner Mongolia University of Science & Technology, China
| | - Zhou NING
- Zhou NING, Baotou Tumor Hospital, China
| | - Ning YUEBAO
- Inner Mongolia University of Science & Technology, China
| |
Collapse
|
38
|
[Dexmedetomidine preconditioning alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats by inhibiting NLRP3 inflammasome activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1857-1863. [PMID: 35012919 PMCID: PMC8752431 DOI: 10.12122/j.issn.1673-4254.2021.12.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To investigate the protective effect of dexmedetomidine (Dex) against acute lung injury induced by intestinal ischemia-reperfusion (II/R) in rats and its effect on NLRP3 inflammasome activity. METHODS Thirty-two normal male SD rats were randomly divided into 4 groups (n=8): the sham operation group, where the superior mesenteric artery (SMA) was exposed only; II/R group, where the SMA was occluded for 1 h followed by reperfusion for 2 h; Dex+II/R group, where the rats were subjected to II/R and received intraperitoneal injection of Dex before reperfusion; and Dex group, where the rats received Dex pretreatment and sham operation. The rats in sham operation group and II/R group received intraperitoneal injection of normal saline. The wet/dry weight ratio (W/D) and myeloperoxidase (MPO) activity in the lung tissues were measured, and HE staining was used to evaluate lung pathologies and determine lung injury score of the rats. The levels of inflammatory cytokines (TNF-α, IL-18, and IL-1β) in the lung tissue were detected using ELISA, and the expressions of NLRP3, ASC, caspase-1 and p-AMPK proteins were determined with Western blotting. RESULTS Compared with the sham-operated rats, the rats with II/R injury showed obvious lung pathologies and significantly increased W/D value, MPO activity and expression of TNF-α, IL-18 and IL-1β in the lung tissue (P < 0.05) with also significantly increased expressions of NLRP3, ASC, and caspase-1 proteins (P < 0.05) but obviously lowered expression of p-AMPK protein (P < 0.05) in the lung tissues. Compared with those in II/R group, the rats in Dex+II/R group showed milder lung pathologies, significantly reduced W/D value, MPO activity and expressions of TNF-α, IL-18 and IL-1β in the lung tissue (P < 0.05), and significant lower expressions of NLRP3, ASC, and caspase-1 (P < 0.05) but higher expression of p-AMPK protein (P < 0.05). CONCLUSION Dex treatment reduces II/R-induced inflammatory response by inhibiting the activation of NLRP3 inflammasomes, thereby improving acute lung injury caused by II/R in rats.
Collapse
|
39
|
Preoperative fasting confers protection against intestinal ischaemia/reperfusion injury by modulating gut microbiota and their metabolites in a mouse model. Br J Anaesth 2021; 128:501-512. [PMID: 34930601 DOI: 10.1016/j.bja.2021.11.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Intestinal ischaemia/reperfusion (I/R) injury is a grave surgical event with high morbidity and mortality. Preoperative fasting might confer protection against intestinal I/R injury by altering the composition of gut microbiota and their respective metabolites. METHODS An intestinal I/R mouse model was established and subjected to preoperative fasting for 24 h or fed ad libitum. Intestinal I/R injury was assessed using histological examination and survival analysis. Faecal samples were collected for 16S rDNA sequencing and metabolomic analysis. Faecal transplantation of fasted and non-fasted mice and humans was conducted to evaluate the effects of gut microbiota on intestinal I/R. Murine small intestinal cells wecre subjected to oxygen and glucose deprivation/reoxygenation as an in vitro I/R model. RESULTS Preoperative fasting protected against intestinal I/R injury and improved survival in mice (P<0.001). In addition, 16S rDNA sequencing revealed that preoperative fasting increased the diversity and restructured the composition of the gut microbiota after intestinal I/R. Mice that received microbiota from fasted mice and humans showed less intestinal damage than those that received microbiota from fed subjects. Metabolomic analysis showed that the profiles of gut microbial metabolites differed between fasted and fed groups. Specifically, the concentration of petroselinic acid was significantly higher in the fasted group (P=0.009). Treatment of intestinal I/R mice with petroselinic acid alleviated intestinal injury in vivo and decreased cell apoptosis by mediating AMP-activated protein kinase-mammalian target of rapamycin-P70S6K signaling in vitro. CONCLUSIONS Preoperative fasting protected against intestinal I/R injury by modulating gut microbiota and petroselinic acid, suggesting a novel therapeutic strategy.
Collapse
|
40
|
Luo J, Chen J, Yang C, Tan J, Zhao J, Jiang N, Zhao Y. 6-Gingerol protects against cerebral ischemia/reperfusion injury by inhibiting NLRP3 inflammasome and apoptosis via TRPV1 / FAF1 complex dissociation-mediated autophagy. Int Immunopharmacol 2021; 100:108146. [PMID: 34537481 DOI: 10.1016/j.intimp.2021.108146] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/22/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our previous studies demonstrated that autophagy alleviates cerebral I/R injury by inhibiting NLRP3 inflammasome-mediated inflammation. 6-Gingerol, a phenolic compound extracted from ginger, was reported to possess potent antiapoptotic and anti-inflammatory activities and is associated with autophagy. However, the effects of 6-Gingerol in cerebral I/R injury have not been elucidated, and whether they involve autophagy-induced NLRP3 inflammasome inhibition remains unclear. METHODS Adult male Sprague-Dawley (SD) rats were subjected to middle cerebral artery occlusion (MCAO) for 1 h, followed by reperfusion for 24 h. 6-Gingerol and 3-methyladenine (3-MA) were injected intraperitoneally, and si-TRPV1 was injected via the lateral ventricle. Cerebral infarct volume, brain edema, neurological deficits, HE and Nissl were used to evaluate the morphological and functional changes of brain tissue, respectively. TRPV1, FAF1, autophagy related (LC3II/I, P62, Beclin1), inflammation related (NLRP3, cleaved-caspase-1, caspase-1, cleaved-IL-1β, IL-1β, cleaved-IL-18, IL-18) and apoptosis related (Bcl-2, Bax, cleaved-caspase-3) proteins were assessed by Western blot, immunofluorescence staining and coimmunoprecipitation, respectively. Enzyme linked immunosorbent assay (ELISA) was used to evaluate the changes in the expression levels of interleukin-1 (IL-1β) and interleukin-18(IL-18), respectively. The degree of neuronal apoptosis was evaluated by TUNEL staining. Neuronal ultrastructure was examined by transmission electron microscopy. RESULT 6-Gingerol treatment significantly reduced cerebral infarct volume, improved brain edema and neurological scores, and reversed brain histomorphological damage after I/R injury. In addition, 6-Gingerol significantly reduced NLRP3 inflammasome-derived inflammation and neuronal apoptosis and upregulated autophagy. The autophagy inhibitor 3-MA rescued the effects of 6-Gingerol on the NLRP3 inflammasome and apoptosis. Moreover, the findings illustrated that 6-Gingerol inhibited autophagy-induced NLRP3 inflammasome activation and apoptosis through the dissociation of TRPV1 from FAF1. CONCLUSION In brief, 6-Gingerol exerts antiapoptotic and anti-inflammatory effects via TRPV1/FAF1 complex dissociation-mediated autophagy during cerebral I/R injury. Therefore, 6-Gingerol may be an effective drug for the treatment of I/R injury.
Collapse
Affiliation(s)
- Jing Luo
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- Department of First Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Changhong Yang
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China
| | - Junyi Tan
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
41
|
Ma SQ, Guo Z, Liu FY, Hasan SG, Yang D, Tang N, An P, Wang MY, Wu HM, Yang Z, Fan D, Tang QZ. 6-Gingerol protects against cardiac remodeling by inhibiting the p38 mitogen-activated protein kinase pathway. Acta Pharmacol Sin 2021; 42:1575-1586. [PMID: 33462378 PMCID: PMC8463710 DOI: 10.1038/s41401-020-00587-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 11/20/2020] [Indexed: 02/02/2023]
Abstract
6-Gingerol, a pungent ingredient of ginger, has been reported to possess anti-inflammatory and antioxidant activities, but the effect of 6-gingerol on pressure overload-induced cardiac remodeling remains inconclusive. In this study, we investigated the effect of 6-gingerol on cardiac remodeling in in vivo and in vitro models, and to clarify the underlying mechanisms. C57BL/6 mice were subjected to transverse aortic constriction (TAC), and treated with 6-gingerol (20 mg/kg, ig) three times a week (1 week in advance and continued until the end of the experiment). Four weeks after TAC surgery, the mice were subjected to echocardiography, and then sacrificed to harvest the hearts for analysis. For in vitro study, neonatal rat cardiomyocytes and cardiac fibroblasts were used to validate the protective effects of 6-gingerol in response to phenylephrine (PE) and transforming growth factor-β (TGF-β) challenge. We showed that 6-gingerol administration protected against pressure overload-induced cardiac hypertrophy, fibrosis, inflammation, and dysfunction in TAC mice. In the in vitro study, we showed that treatment with 6-gingerol (20 μM) blocked PE-induced-cardiomyocyte hypertrophy and TGF-β-induced cardiac fibroblast activation. Furthermore, 6-gingerol treatment significantly decreased mitogen-activated protein kinase p38 (p38) phosphorylation in response to pressure overload in vivo and extracellular stimuli in vitro, which was upregulated in the absence of 6-gingerol treatment. Moreover, transfection with mitogen-activated protein kinase kinase 6 expressing adenoviruses (Ad-MKK6), which specifically activated p38, abolished the protective effects of 6-gingerol in both in vitro and in vivo models. In conclusion, 6-gingerol improves cardiac function and alleviates cardiac remodeling induced by pressure overload in a p38-dependent manner. The present study demonstrates that 6-gingerol is a promising agent for the intervention of pathological cardiac remodeling.
Collapse
Affiliation(s)
- Shu-Qing Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Shahzad-Gul Hasan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
- Department of Medicine, Bahawal Victoria Hospital, Bahawalpur, 63100, Pakistan
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Hai-Ming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, China.
| |
Collapse
|
42
|
Jafarzadeh A, Jafarzadeh S, Nemati M. Therapeutic potential of ginger against COVID-19: Is there enough evidence? JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [PMCID: PMC8492833 DOI: 10.1016/j.jtcms.2021.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In addition to the respiratory system, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strikes other systems, including the digestive, circulatory, urogenital, and even the central nervous system, as its receptor angiotensin-converting enzyme 2 (ACE2) is expressed in various organs, such as lungs, intestine, heart, esophagus, kidneys, bladder, testis, liver, and brain. Different mechanisms, in particular, massive virus replication, extensive apoptosis and necrosis of the lung-related epithelial and endothelial cells, vascular leakage, hyper-inflammatory responses, overproduction of pro-inflammatory mediators, cytokine storm, oxidative stress, downregulation of ACE2, and impairment of the renin-angiotensin system contribute to the COVID-19 pathogenesis. Currently, COVID-19 is a global pandemic with no specific anti-viral treatment. The favorable capabilities of the ginger were indicated in patients suffering from osteoarthritis, neurodegenerative disorders, rheumatoid arthritis, type 2 diabetes, respiratory distress, liver diseases and primary dysmenorrheal. Ginger or its compounds exhibited strong anti-inflammatory and anti-oxidative influences in numerous animal models. This review provides evidence regarding the potential effects of ginger against SARS-CoV-2 infection and highlights its antiviral, anti-inflammatory, antioxidative, and immunomodulatory impacts in an attempt to consider this plant as an alternative therapeutic agent for COVID-19 treatment.
Collapse
|
43
|
Wang Y, Wen J, Almoiliqy M, Wang Y, Liu Z, Yang X, Lu X, Meng Q, Peng J, Lin Y, Sun P. Sesamin Protects against and Ameliorates Rat Intestinal Ischemia/Reperfusion Injury with Involvement of Activating Nrf2/HO-1/NQO1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5147069. [PMID: 34630849 PMCID: PMC8494576 DOI: 10.1155/2021/5147069] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023]
Abstract
Intestinal ischemia-reperfusion (I/R) may induce cell/tissue injuries, leading to multiple organ failure. Based on our preexperiments, we proposed that sesamin could protect against and ameliorate intestinal I/R injuries and related disorders with involvement of activating Nrf2 signaling pathway. This proposal was evaluated using SD intestinal I/R injury rats in vivo and hypoxia/reoxygenation- (H/R-) injured rat small intestinal crypt epithelial cell line (IEC-6 cells) in vitro. Sesamin significantly alleviated I/R-induced intestinal histopathological injuries and significantly reduced serum biochemical indicators ALT and AST, alleviating I/R-induced intestinal injury in rats. Sesamin also significantly reversed I/R-increased TNF-α, IL-6, IL-1β, and MPO activity in serum and MDA in tissues and I/R-decreased GSH in tissues and SOD in both tissues and IEC-6 cells, indicating its anti-inflammatory and antioxidative stress effects. Further, sesamin significantly decreased TUNEL-positive cells, downregulated the increased Bax and caspase-3 protein expression, upregulated the decreased protein expression of Bcl-2 in I/R-injured intestinal tissues, and significantly reversed H/R-reduced IEC-6 cell viability as well as reduced the number of apoptotic cells among H/R-injured IEC-6 cell, showing antiapoptotic effects. Activation of Nrf2 is known to ameliorate tissue/cell injuries. Consistent with sesamin-induced ameliorations of both intestinal I/R injuries and H/R injuries, transfection of Nrf2 cDNA significantly upregulated the expression of Nrf2, HO-1, and NQO1, respectively. On the contrary, either Nrf2 inhibitor (ML385) or Nrf2 siRNA transfection significantly decreased the expression of these proteins. Our results suggest that activation of the Nrf2/HO-1/NQO1 signaling pathway is involved in sesamin-induced anti-inflammatory, antioxidative, and antiapoptotic effects in protection against and amelioration of intestinal I/R injuries.
Collapse
Affiliation(s)
- Yilin Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001 Liaoning, China
| | - Jin Wen
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Marwan Almoiliqy
- Key Lab of Aromatic Plant Resources Exploitation and Utilization in Sichuan Higher Education, Yibin University, Yibin, 644000 Sichuan, China
| | - Yaojia Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Zhihao Liu
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Xiaobo Yang
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Xiaolong Lu
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Qiang Meng
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Yuan Lin
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
| | - Pengyuan Sun
- College of Pharmacy, Dalian Medical University, Dalian, 116044 Liaoning, China
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian, 116044 Liaoning, China
| |
Collapse
|
44
|
JIANG Q, XU N, KONG L, WANG M, LEI H. Promoting effects of 6-Gingerol on probiotic adhesion to colonic epithelial cells. FOOD SCIENCE AND TECHNOLOGY 2021. [DOI: 10.1590/fst.17420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Qian JIANG
- Nanjing University of Finance and Economics, China
| | - Na XU
- Nanjing University of Finance and Economics, China
| | - Lingyan KONG
- Nanjing University of Finance and Economics, China
| | - Mengxi WANG
- Nanjing University of Finance and Economics, China
| | - Hong LEI
- Nanjing University of Finance and Economics, China
| |
Collapse
|
45
|
Lin H, Zhang X, Wang D, Liu J, Yuan L, Liu J, Wang C, Sun J, Chen J, Li H, Jing S. Anwulignan Ameliorates the Intestinal Ischemia/Reperfusion. J Pharmacol Exp Ther 2021; 378:222-234. [PMID: 34131018 DOI: 10.1124/jpet.121.000587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/04/2021] [Indexed: 11/22/2022] Open
Abstract
Anwulignan is one of the monomer compounds in the lignans from Schisandra sphenanthera In this study, we observed the effect of anwulignan on intestinal ischemia/reperfusion (II/R) injury in male Sprague-Dawley rats and explored the underlying mechanisms. The results showed that pretreatment with oral anwulignan could significantly increase the mesenteric blood microcirculatory flow velocity; relieve the congestion and pathologic injury of jejunum; enhance the autonomic tension of jejunum smooth muscle and its reactivity to acetylcholine; increase the activities of superoxide dismutase, catalase, glutathione S-transferase, and choline acetyltransferase; increase the contents of acetylcholine and glutathione in the serum or jejunal tissue; decrease the activities of myeloperoxidase, protein kinase C, and nicotinamide adenine dinucleotide phosphate oxidase; reduce the contents of malondialdehyde, 8-hydroxy-2-deoxyguanosine, nicotinamide adenine, reactive oxygen species, tumor necrosis factor-α, interleukin (IL)-6, and IL-1β; increase the expression levels of muscarinic receptor 3, PI3K, phosphorylation protein kinase B, p-GSK3β Ser9, Nrf2, p-Nrf2, heme oxygenase (decycling) 1, and b-cell lymphoma 2 in the jejunal tissue; and decrease the expression levels of p-GSK3β Tyr216, kelch-like ECH-associated protein 1, Bax, and cleaved caspase-3, suggesting that anwulignan can ameliorate II/R-induced jejunal tissue injury in rats and that the mechanism may be related to its activating the PI3K/protein kinase B pathway and then regulating the Nrf2/Anti-oxidative Response Element signaling pathway and the expression of apoptosis-related proteins to play antioxidant and antiapoptotic roles. SIGNIFICANCE STATEMENT: Anwulignan can significantly reduce jejunal tissue injury and the production of inflammatory factors in rats with intestinal ischemia-reperfusion injury, improve the antioxidant capacity, and reduce the apoptosis of jejunal tissue, and it has the effect of significantly improving intestinal ischemia-reperfusion injury in rats, suggesting that anwulignan may be used as a potential drug for the prevention and treatment of intestinal ischemia-reperfusion injury or a resource for the development of health food.
Collapse
Affiliation(s)
- Huijiao Lin
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Xinyun Zhang
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Dan Wang
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Jiawei Liu
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Liwei Yuan
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Jiale Liu
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Jinghui Sun
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Jianguang Chen
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - He Li
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| | - Shu Jing
- Department of Pharmacology, College of Pharmacy (Hu.L., X.Z., Jiaw.L., L.Y., C.W., J.S., J.C., He.L.); College of Basic Medicine (D.W.), Beihua University, Jilin City, China; Jilin City Central Hospital, Jilin City, China (Jial.L.); and Affiliated Hospital of Beihua University, Jilin City, China (S.J.)
| |
Collapse
|
46
|
Chen X, Hu C, Fan X, Wang Y, Li Q, Su YQ, Zhang DM, Yang Q, Passerini AG, Sun C. mTOR Inhibition Promotes Pneumonitis Through Inducing Endothelial Contraction and Hyperpermeability. Am J Respir Cell Mol Biol 2021; 65:646-657. [PMID: 34251297 DOI: 10.1165/rcmb.2020-0390oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Compromised endothelial (EC) barrier function is a hallmark of inflammatory diseases. Mammalian target of rapamycin (mTOR) inhibitors, widely applied as clinical therapies, cause pneumonitis through mechanisms not yet fully understood. This study aimed to elucidate the EC mechanisms underlying the pathogenesis of pneumonitis caused by mTOR inhibition (mTORi). Mice with EC-specific deletion of mTOR complex components (Mtor, Rptor or Rictor) were administered LPS to induce pulmonary injury. Cultured EC were treated with pharmacological inhibitors, small interfering RNA or overexpression-plasmids. EC barrier function was evaluated in vivo with Evan's blue assay and in vitro by measurement of transendothelial electrical resistance and albumin flux. mTORi increased basal and TNFα-induced EC permeability, which was caused by myosin light chain (MLC) phosphorylation-dependent cell contraction. Inactivation of mTOR kinase activity by mTORi triggered PKCδ/p38/NF-κB signaling that significantly upregulated TNFα-induced MLC kinase (MLCK) expression, while Raptor promoted the phosphorylation of PKCα/MYPT1 independent of its interaction with mTOR, leading to suppression of MLC phosphatase (MLCP) activity. EC-specific deficiency in mTOR, Raptor or Rictor aggravated lung inflammation in LPS-treated mice. These findings reveal that mTORi induces PKC-dependent endothelial MLC phosphorylation, contraction and hyperpermeability that promote pneumonitis.
Collapse
Affiliation(s)
- Xiaolin Chen
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,2Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Chengxiu Hu
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Xing Fan
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Yiying Wang
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Qiannan Li
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - You-Qiang Su
- Nanjing Medical University, 12461, State Key Laboratory of Reproductive Medicine, Nanjing, China
| | - Dai-Min Zhang
- Nanjing First Hospital, Nanjing Medical University, Department of Cardiology, Nanjing, China
| | - QianLu Yang
- Nanjing Medical University Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, 540955, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing, China.,Key laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, China
| | - Anthony G Passerini
- University of California Davis, 8789, Department of Biomedical Engineering, Davis, California, United States
| | - ChongXiu Sun
- Nanjing Medical University, 12461, Nanjing, China;
| |
Collapse
|
47
|
Wallner C, Drysch M, Becerikli M, Schmidt SV, Hahn S, Wagner JM, Reinkemeier F, Dadras M, Sogorski A, von Glinski M, Lehnhardt M, Behr B. Deficiency of myostatin protects skeletal muscle cells from ischemia reperfusion injury. Sci Rep 2021; 11:12572. [PMID: 34131275 PMCID: PMC8206371 DOI: 10.1038/s41598-021-92159-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/26/2021] [Indexed: 02/05/2023] Open
Abstract
Ischemia reperfusion (IR) injury plays a pivotal role in many diseases and leads to collateral damage during surgical interventions. While most studies focus on alleviating its severity in the context of brain, liver, kidney, and cardiac tissue, research as regards to skeletal muscle has not been conducted to the same extent. In the past, myostatin (MSTN), primarily known for supressing muscle growth, has been implicated in inflammatory circuits, and research provided promising results for cardiac IR injury mitigation by inhibiting MSTN cell surface receptor ACVR2B. This generated the question if interrupting MSTN signaling could temper IR injury in skeletal muscle. Examining human specimens from free myocutaneous flap transfer demonstrated increased MSTN signaling and tissue damage in terms of apoptotic activity, cell death, tissue edema, and lipid peroxidation. In subsequent in vivo MstnLn/Ln IR injury models, we identified potential mechanisms linking MSTN deficiency to protective effects, among others, inhibition of p38 MAPK signaling and SERCA2a modulation. Furthermore, transcriptional profiling revealed a putative involvement of NK cells. Collectively, this work establishes a protective role of MSTN deficiency in skeletal muscle IR injury.
Collapse
Affiliation(s)
- Christoph Wallner
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Marius Drysch
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Mustafa Becerikli
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Sonja Verena Schmidt
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Stephan Hahn
- grid.5570.70000 0004 0490 981XDepartment of Molecular Gastrointestinal Oncology, Ruhr University Bochum, Universitätsstraße 150, 44780 Bochum, Germany
| | - Johannes Maximilian Wagner
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Felix Reinkemeier
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Mehran Dadras
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Alexander Sogorski
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Maxi von Glinski
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Marcus Lehnhardt
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| | - Björn Behr
- grid.412471.50000 0004 0551 2937Department of Plastic Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bürkle-de-la-Camp Platz 1, 44789 Bochum, Germany
| |
Collapse
|
48
|
Mo Y, Sun H, Zhang L, Geng W, Wang L, Zou C, Wu Y, Ji C, Liu X, Lu Z. Microbiome-Metabolomics Analysis Reveals the Protection Mechanism of α-Ketoacid on Adenine-Induced Chronic Kidney Disease in Rats. Front Pharmacol 2021; 12:657827. [PMID: 34045965 PMCID: PMC8144710 DOI: 10.3389/fphar.2021.657827] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/26/2021] [Indexed: 01/14/2023] Open
Abstract
Objectives: As nitrogen-free precursors of corresponding essential amino, α-ketoacid have been widely prescribed to end-stage renal disease patients together with a low protein diet However, the impact of α-ketoacid on intestinal microbiota in chronic kidney disease (CKD) individuals is unknown. The study aims at investigating the variation in the intestinal microbiota and metabolic profile in response to α-ketoacid treatment in an adenine-induced CKD rat model. Design: Rats in the treatment groups were given solution of compound α-ketoacid tablets. At the end of the study, blood, feces, colon tissues and kidney tissues were collected and processed for biochemical analyses, histological and western blot analyses, 16S rRNA sequence and untargeted metabolomic analyses. Results: α-Ketoacid treatment reduced serum creatinine, blood urea nitrogen and 24 h urine protein, and alleviated tubular atrophy, glomerulosclerosis and interstitial fibrosis in adenine-induced CKD rats. Moreover, α-ketoacid significantly improved intestinal barrier and increased the abundance of Methanobrevibacter, Akkermansia, Blautia and Anaerositipes while reduced the abundance of Anaerovorax and Coprococcus_3 at the genus level. In addition, our results also demonstrated that α-ketoacid significantly reduced the concentrations of indoxyl sulfate, betaine, choline and cholesterol. Spearman's correlation analysis revealed that the abundance of Coprococcus_3 was positively correlated with serum level of betaine, trimethylamine N-oxide, indoxyl sulfate, cholic acid and deoxycholic acid. Conclusion: α-Ketoacid has a reno-protective effect against adenine-induced CKD, which may be mediated regulation of serum metabolic profiles via affecting intestinal microbial community.
Collapse
Affiliation(s)
- Yenan Mo
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huang Sun
- Department of Emergency, TCM-Integrated Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zhang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjia Geng
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lixin Wang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuan Zou
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuchi Wu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunlan Ji
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhaoyu Lu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Nephrology Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
49
|
Yeo EJ, Eum WS, Yeo HJ, Choi YJ, Sohn EJ, Kwon HJ, Kim DW, Kim DS, Cho SW, Park J, Han KH, Lee KW, Park JK, Shin MJ, Choi SY. Protective Role of Transduced Tat-Thioredoxin1 (Trx1) against Oxidative Stress-Induced Neuronal Cell Death via ASK1-MAPK Signal Pathway. Biomol Ther (Seoul) 2021; 29:321-330. [PMID: 33436533 PMCID: PMC8094070 DOI: 10.4062/biomolther.2020.154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/23/2020] [Accepted: 12/10/2020] [Indexed: 01/07/2023] Open
Abstract
Oxidative stress plays a crucial role in the development of neuronal disorders including brain ischemic injury. Thioredoxin 1 (Trx1), a 12 kDa oxidoreductase, has anti-oxidant and anti-apoptotic functions in various cells. It has been highly implicated in brain ischemic injury. However, the protective mechanism of Trx1 against hippocampal neuronal cell death is not identified yet. Using a cell permeable Tat-Trx1 protein, protective mechanism of Trx1 against hydrogen peroxide-induced cell death was examined using HT-22 cells and an ischemic animal model. Transduced Tat-Trx1 markedly inhibited intracellular ROS levels, DNA fragmentation, and cell death in H2O2-treatment HT-22 cells. Tat-Trx1 also significantly inhibited phosphorylation of ASK1 and MAPKs in signaling pathways of HT-22 cells. In addition, Tat-Trx1 regulated expression levels of Akt, NF-κB, and apoptosis related proteins. In an ischemia animal model, Tat-Trx1 markedly protected hippocampal neuronal cell death and reduced astrocytes and microglia activation. These findings indicate that transduced Tat-Trx1 might be a potential therapeutic agent for treating ischemic injury.
Collapse
Affiliation(s)
- Eun Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Won Sik Eum
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyeon Ji Yeo
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Yeon Joo Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Eun Jeong Sohn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
| | - Duk-Soo Kim
- Department of Anatomy and BK21 Plus Center, College of Medicine, Soonchunhyang University, Cheonan 31538, Republic of Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyu Hyung Han
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Keun Wook Lee
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Min Jea Shin
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
50
|
Li C, Li J, Jiang F, Tzvetkov NT, Horbanczuk JO, Li Y, Atanasov AG, Wang D. Vasculoprotective effects of ginger ( Zingiber officinale Roscoe) and underlying molecular mechanisms. Food Funct 2021; 12:1897-1913. [PMID: 33592084 DOI: 10.1039/d0fo02210a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ginger (Zingiber officinale Roscoe) is a common and widely used spice. It is rich in various chemical constituents, including phenolic compounds, terpenes, polysaccharides, lipids, organic acids, and raw fibers. Herein, we reviewed its effects on the vascular system. Studies utilizing cell cultures or animal models showed that ginger constituents alleviate oxidative stress and inflammation, increase nitric oxide synthesis, suppress vascular smooth muscle cell proliferation, promote cholesterol efflux from macrophages, inhibit angiogenesis, block voltage-dependent Ca2+ channels, and induce autophagy. In clinical trials, ginger was shown to have a favorable effect on serum lipids, inflammatory cytokines, blood pressure, and platelet aggregation. Taken together, these studies point to the potential benefits of ginger and its constituents in the treatment of hypertension, coronary artery disease, peripheral arterial diseases, and other vascular diseases.
Collapse
Affiliation(s)
- Chao Li
- Experimental center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Jie Li
- Experimental center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Feng Jiang
- Department of Cardiology, Affiliated Hospital of Shandong University of traditional Chinese medicine, Jinan, 250000, China
| | - Nikolay T Tzvetkov
- Institute of Molecular Biology "Roumen Tsanev", Department of Biochemical Pharmacology and Drug Design, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | - Jaroslaw O Horbanczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, 05-552 Jastrzębiec, Poland
| | - Yunlun Li
- Experimental center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China. and Department of Cardiology, Affiliated Hospital of Shandong University of traditional Chinese medicine, Jinan, 250000, China
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, 05-552 Jastrzębiec, Poland and Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria and Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchevstr., 1113 Sofia, Bulgaria and Department of Pharmacognosy, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Dongdong Wang
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Main Street West 1280, L8S4L8 Hamilton, Ontario, Canada.
| |
Collapse
|