1
|
Yadav S, Pandey A, Mali SN. From lab to nature: Recent advancements in the journey of gastroprotective agents from medicinal chemistry to phytotherapy. Eur J Med Chem 2024; 272:116436. [PMID: 38704935 DOI: 10.1016/j.ejmech.2024.116436] [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: 02/01/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
Peptic ulcer, affecting 10 % of the global population, results from imbalances in gastric juice pH and diminished mucosal defences. Key underlying factors are non-steroidal anti-inflammatory drugs (NSAIDs) and Helicobacter pylori infection, undermining mucosal resistance. Traditional treatments like proton pump inhibitors (PPIs) and histamine-2 (H2) receptor antagonists exhibit drawbacks such as adverse effects, relapses, and drug interactions. This review extensively explores the ethnomedicinal, synthetic and pharmacological facets of various potential peptic ulcer treatments. Rigorous methodologies involving electronic databases, and chemical structure verification via 'PubChem' and 'SciFinder' enhance the review's credibility. The provided information, spanning medicinal insights to intricate pharmacological mechanisms, establishes a robust groundwork for future research and the development of plant-derived or synthetic molecules for peptic ulcers, offering a promising alternative to conventional therapies.
Collapse
Affiliation(s)
- Susmita Yadav
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Anima Pandey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Suraj N Mali
- School of Pharmacy, D.Y. Patil University (Deemed to be University), Sector 7, Nerul, Navi Mumbai, 400706, India.
| |
Collapse
|
2
|
de Sousa DP, de Assis Oliveira F, Arcanjo DDR, da Fonsêca DV, Duarte ABS, de Oliveira Barbosa C, Ong TP, Brocksom TJ. Essential Oils: Chemistry and Pharmacological Activities-Part II. Biomedicines 2024; 12:1185. [PMID: 38927394 PMCID: PMC11200837 DOI: 10.3390/biomedicines12061185] [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: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
The importance of essential oils and their components in the industrial sector is attributed to their chemical characteristics and their application in the development of products in the areas of cosmetology, food, and pharmaceuticals. However, the pharmacological properties of this class of natural products have been extensively investigated and indicate their applicability for obtaining new drugs. Therefore, this review discusses the use of these oils as starting materials to synthesize more complex molecules and products with greater commercial value and clinic potential. Furthermore, the antiulcer, cardiovascular, and antidiabetic mechanisms of action are discussed. The main mechanistic aspects of the chemopreventive properties of oils against cancer are also presented. The data highlight essential oils and their derivatives as a strategic chemical group in the search for effective therapeutic agents against various diseases.
Collapse
Affiliation(s)
| | | | - Daniel Dias Rufino Arcanjo
- LAFMOL—Laboratory of Functional and Molecular Studies in Physiopharmacology, Department of Biophysics and Physiology, Federal University of Piaui, Teresina 64049-550, Brazil; (D.D.R.A.); (C.d.O.B.)
| | - Diogo Vilar da Fonsêca
- Collegiate of Medicine, Federal University of São Francisco Valley, Bahia 48607-190, Brazil;
| | - Allana Brunna S. Duarte
- Laboratory of Pharmaceutical Chemistry, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| | - Celma de Oliveira Barbosa
- LAFMOL—Laboratory of Functional and Molecular Studies in Physiopharmacology, Department of Biophysics and Physiology, Federal University of Piaui, Teresina 64049-550, Brazil; (D.D.R.A.); (C.d.O.B.)
| | - Thomas Prates Ong
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo 05508-000, Brazil;
- Food Research Center (FoRC), University of São Paulo, São Paulo 05508-000, Brazil
| | - Timothy John Brocksom
- Department of Chemistry, Federal University of São Carlos, São Carlos 13565-905, Brazil;
| |
Collapse
|
3
|
Gelen V, Gedikli S, Gelen SU, Şengül E, Makav M. Probiotic bacteria protect against indomethacin-induced gastric ulcers through modulation of oxidative stress, inflammation, and apoptosis. Mol Biol Rep 2024; 51:684. [PMID: 38796650 DOI: 10.1007/s11033-024-09627-x] [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: 02/26/2024] [Accepted: 05/08/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Indomethacin is an anti-inflammatory drug that causes ulcers on the gastric mucosa due to its use. Probiotic bacteria are live microorganisms, and it has been stated by various studies that these bacteria have antioxidant and anti-inflammatory effects. In this study, we investigated the possible protective effect of various types of probiotic bacteria (Lactobacillus rhamnosus, Lactobacillus fermentum, and Lactobacillus brevis) against acute gastric mucosal damage caused by indomethacin. METHODS Control group - Physiological saline was administered daily for 10 days. Indo group-Physiological saline was administered daily for 10 days. Ranitidine + Indo group 5 mg/kg ranitidine dose was administered daily for 5 days. On day 11, a single dose of 100 mg/kg of indomethacin was given to the same group. Probiotic + Indo group 1 ml/kg of oral probiotic bacteria was administered daily for 10 days. On day 11, a single 100 mg/kg dose of indomethacin was given. After the application, the rats were anesthetized with ketamine xylazine, killed under appropriate conditions, the abdominal cavity was opened and the stomach tissues were removed. The obtained gastric tissues were used in the biochemical and histopathological analyses discussed below. All data were statistically evaluated by one-way ANOVA using SPSS 20.00, followed by Duncan Post hoc test. The data were expressed as mean ± SD. P < 0.05 was considered statistically significant. RESULTS As a result, the administration of indomethacin caused gastric damage, stimulating oxidative stress, inflammation, and apoptosis. We found that the use of probiotic bacteria reduces oxidative stress (TOC), increases the activity of antioxidant enzymes (TAC), suppresses inflammation (IL-6 and Tnf-α), and inhibits apoptosis (Bax and Bcl-2) (P < 0.05). CONCLUSION Probiotic treatment can mitigate gastric damage and apoptosis caused by indomethacin-induced gastric damage in rats. Probiotic also enhances the restoration of biochemical oxidative enzymes as it has anti-inflammatory, antioxidant, and antiapoptotic properties.
Collapse
Affiliation(s)
- Volkan Gelen
- Department of Physiology, Veterinary Faculty, Kafkas University, Kars, Turkey.
| | - Semin Gedikli
- Department of Histology, Veterinary Faculty, Ataturk University, Erzurum, Turkey
| | - Sevda Urçar Gelen
- Department of Food Hygiene and Technology, Faculty of Veterinary, Ataturk University, Erzurum, Turkey
| | - Emin Şengül
- Department of Physiology, Veterinary Faculty, Ataturk University, Erzurum, Turkey
| | - Mustafa Makav
- Department of Physiology, Veterinary Faculty, Kafkas University, Kars, Turkey
| |
Collapse
|
4
|
Gong H, Zhao N, Zhu C, Luo L, Liu S. Treatment of gastric ulcer, traditional Chinese medicine may be a better choice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117793. [PMID: 38278376 DOI: 10.1016/j.jep.2024.117793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/30/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastric ulcer (GU) is the injury of the gastric mucosa caused by the stimulation of various pathogenic factors penetrating the deep mucosal muscle layer. An increasing number of studies have shown that traditional Chinese medicine (TCM) is highly effective in treating GU due to its multitarget, multilevel, and multi-pathway effects. AIM OF THE STUDY To review the latest research progress in the treatment of GU by TCM, including clinical and experimental studies, focusing on the target and mechanism of action of drugs and providing a theoretical basis for the treatment of GU by natural herbs. MATERIALS AND METHODS Electronic databases (PubMed, Elsevier, Springer, Web of Science, and CNKI) were searched using the keywords "gastric ulcer", "gastric mucosal lesion", "TCM" and or paired with "peptic ulcer" and "natural drugs" for studies published in the last fifteen years until 2023. RESULTS TCM, including single components of natural products, Chinese patent medicines (CPM), and TCM decoction, is expected to treat GU by regulating various mechanisms, such as redox balance, inflammatory factors, angiogenesis, gastric mucosal protective factors, intestinal flora, apoptosis, and autophagy. CONCLUSIONS We discussed and summarized the mechanism of TCM in the treatment of GU, which provided a sufficient basis for TCM treatment of GU.
Collapse
Affiliation(s)
- Haiying Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China; Department of Gastroenterology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Conglei Zhu
- Department of Pharmacy, Fuyang People's Hospital, Fuyang, Anhui, China
| | - Lin Luo
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Sen Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Xu J, Yang XW. LC-MS-Based Metabolomics Reveals the Mechanism of Protection of Berberine against Indomethacin-Induced Gastric Injury in Rats. Molecules 2024; 29:1055. [PMID: 38474567 DOI: 10.3390/molecules29051055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Berberine is a natural isoquinoline alkaloid with low toxicity, which exists in a wide variety of medicinal plants. Berberine has been demonstrated to exhibit potent prevention of indomethacin-induced gastric injury (GI) but the related mechanism remains unclear. In the present study, liquid chromatography-mass spectrometry (LC-MS)-based metabolomics was applied for the first time to investigate the alteration of serum metabolites in the protection of berberine against indomethacin-induced gastric injury in rats. Subsequently, bioinformatics was utilized to analyze the potential metabolic pathway of the anti-GI effect of berberine. The pharmacodynamic data indicated that berberine could ameliorate gastric pathological damage, inhibit the level of proinflammatory factors in serum, and increase the level of antioxidant factors in serum. The LC-MS-based metabolomics analysis conducted in this study demonstrated the presence of 57 differential metabolites in the serum of rats with induced GI caused by indomethacin, which was associated with 29 metabolic pathways. Moreover, the study revealed that berberine showed a significant impact on the differential metabolites, with 45 differential metabolites being reported between the model group and the group treated with berberine. The differential metabolites were associated with 24 metabolic pathways, and berberine administration regulated 14 of the 57 differential metabolites, affecting 14 of the 29 metabolic pathways. The primary metabolic pathways affected were glutathione metabolism and arachidonic acid metabolism. Based on the results, it can be concluded that berberine has a gastroprotective effect on the GI. This study is particularly significant since it is the first to elucidate the mechanism of berberine's action on GI. The results suggest that berberine's action may be related to energy metabolism, oxidative stress, and inflammation regulation. These findings may pave the way for the development of new therapeutic interventions for the prevention and management of NSAID-induced GI disorders.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiu-Wei Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
6
|
Wu Y, Guo Y, Huang T, Huang D, Liu L, Shen C, Jiang C, Wang Z, Chen H, Liang P, Hu Y, Zheng Z, Liang T, Zhai D, Zhu H, Liu Q. Licorice flavonoid alleviates gastric ulcers by producing changes in gut microbiota and promoting mucus cell regeneration. Biomed Pharmacother 2023; 169:115868. [PMID: 37952360 DOI: 10.1016/j.biopha.2023.115868] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
Licorice flavonoid (LF) is the main component of Glycyrrhizae Radix et Rhizoma, a "medicine food homology" herbal medicine, which has anti-digestive ulcer activity, but the mechanism in anti-gastric ulcer (GU) remains to be elucidated. In this study, we manifested that LF increased the viability of human gastric mucosal epithelial (GES-1) cells, attenuated ethanol (EtOH)-induced manifestations, reduced histological injury, suppressed inflammation, and restored gastric mucosal barrier in GU rats. After LF therapy, the EtOH-induced gut dysbiosis was partly modulated, and short-chain fatty acids (SCFAs) like butyric acid, propionic acid, and valeric acid were found in higher concentrations. We discovered that the majority of genera that increased in the GU group had a negative correlation with SCFAs in the intestinal tract. In addition, LF-upregulated SCFAs boosted mucus secretion in the gastric epithelium and the expression of mucoprotein (MUC) 5AC and MUC6, particularly the MUC5AC in the gastric foveola. Moreover, LF triggered the EGFR/ERK signal pathway which promoted gastric mucus cell regeneration. Therefore, the findings indicated that LF could inhibit inflammation, promote mucosal barrier repair and angiogenesis, regulate gut microbiota and SCFA metabolism; more importantly, promote epithelial proliferation via activation of the EGFR/ERK pathway, exerting a protective and regenerative effect on the gastric mucosa.
Collapse
Affiliation(s)
- Yufan Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yinglin Guo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Tairun Huang
- Faculty of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
| | - Dehao Huang
- Huizhou Jiuhui Pharmaceutical Co., Ltd., Huizhou 516000, China
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chunyan Shen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Cuiping Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zhuxian Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hongkai Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Peiyi Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yi Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zeying Zheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Tao Liang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Dan Zhai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hongxia Zhu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
7
|
Yuan Y, Wang X, Wang Y, Liu Y, Zhao L, Zhao L, Cai S. The Gastroprotective Effect of Walnut Peptides: Mechanisms and Impact on Ethanol-Induced Acute Gastric Mucosal Injury in Mice. Nutrients 2023; 15:4866. [PMID: 38068724 PMCID: PMC10708498 DOI: 10.3390/nu15234866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
The objective of this research was to explore the protective impact of walnut peptides (WP) against ethanol-induced acute gastric mucosal injury in mice and to investigate the underlying defense mechanisms. Sixty male BALB-c mice were divided into five groups, and they were orally administered distilled water, walnut peptides (200 and 400 mg/kg bw), and omeprazole (20 mg/kg bw) for 24 days. Acute gastric mucosal injury was then induced with 75% ethanol in all groups of mice except the blank control group. Walnut peptides had significant protective and restorative effects on tissue indices of ethanol-induced gastric mucosal damage, with potential gastric anti-ulcer effects. Walnut peptides significantly inhibited the excessive accumulation of alanine aminotransferase (ALT), aspartate transferase (AST), and malondialdehyde (MDA), while promoting the expression of reduced glutathione (GSH), total antioxidant capacity (T-AOC), glutathione disulfide (GSSG), and mouse epidermal growth factor (EGF). Furthermore, the Western blot analysis results revealed that walnut peptides significantly upregulated the expression of HO-1 and NQO1 proteins in the Nrf2 signaling pathway. The defensive impact of walnut peptides on the gastric mucosa may be achieved by mitigating the excessive generation of lipid peroxides and by boosting cellular antioxidant activity.
Collapse
Affiliation(s)
- Yutong Yuan
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (X.W.); (Y.L.); (L.Z.)
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Xinyi Wang
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (X.W.); (Y.L.); (L.Z.)
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Yumeng Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Yaqi Liu
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (X.W.); (Y.L.); (L.Z.)
| | - Liang Zhao
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (X.W.); (Y.L.); (L.Z.)
| | - Lei Zhao
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (Y.Y.); (X.W.); (Y.L.); (L.Z.)
| | - Shengbao Cai
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
8
|
Ma M, Zhao M. Investigation into the Enhancement of Gas-Liquid Mass Transfer of Absorption of H 2S by MDEA with Carbon Quantum Dots. ACS OMEGA 2023; 8:34678-34686. [PMID: 37779968 PMCID: PMC10535251 DOI: 10.1021/acsomega.3c03597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023]
Abstract
Although the addition of fine particles can enhance mass transfer, the stability of suspension is still a challenge. Responding to this, this study introduced carbon quantum dots (CQDs) with good hydrophilicity into a desulfurizer. N-doped carbon quantum dots (N-CQDs) were prepared by the hydrothermal method and characterized by TEM, FT-IR, and XPS. The stability and rheological properties of MDEA-based CQD solutions with different concentrations were studied. CQD solutions with low concentrations showed good stability, and the viscosity of CQD solutions was positively correlated with concentration and inversely correlated with temperature. The desulfurization experiment showed that the desulfurization effect and mass transfer enhancement of MDEA-based CQD solutions were coinfluenced by the viscosity and concentration of the solution; 0.01 vol % CQD solution had the best desulfurization effect, and the mass transfer coefficient was 0.66 mol/(m3h kPa), which increased by 26.61% compared to the base solution.
Collapse
Affiliation(s)
- Mengyu Ma
- School of Chemistry and Pharmaceutical
Engineering, Huanghuai University, Zhumadian 463000, China
| | - Mengxi Zhao
- School of Chemistry and Pharmaceutical
Engineering, Huanghuai University, Zhumadian 463000, China
| |
Collapse
|
9
|
Li P, Xiao X, Gong J, Zhang X, Cai K, Liang R, Wang D, Chen Y, Chen H, Xie Z, Liao Q. Pogostemon cablin (Blanco) Benth granule revealed a positive effect on improving intestinal barrier function and fecal microbiota in mice with irinotecan-induced intestinal mucositis. Arch Microbiol 2023; 205:179. [PMID: 37029820 DOI: 10.1007/s00203-023-03526-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023]
Abstract
Pogostemon cablin (Blanco) Benth (PCB), a medicinal and edible homologous Chinese herb, has a protective effect on the structure and function of intestine. In this study, we aimed to investigate the effect of PCB granule (PCBG) on the improvement of irinotecan-induced intestinal mucositis and the regulation of intestinal microorganisms in mice. Our results demonstrated that PCBG supplementation significantly improved diarrhea symptoms caused by irinotecan, as evidenced by inhibiting weight loss, reversing intestinal atrophy, protecting against splenomegaly and balancing oxidative stress. Furthermore, compared with the model group, PCBG restored the intestinal morphology and improved intestinal barrier dysfunction by promoting the expression of tight junction proteins and mucin. Moreover, high-throughput sequencing analysis revealed that PCBG improved the flora disorder caused by irinotecan and regulated microbial community structure, such as decreasing the relative abundance of Bacteroides as well as increasing the relative abundance of Lactobacillus. Meanwhile, the disordered microbial functions in intestinal mucositis mice were recovered more closely to the controls by PCBG. Finally, we found that a robust correlation between the specific microbiota and intestinal mucositis-related index. In summary, these findings revealed the beneficial effects of PCBG on the intestinal barrier and gut microbiota of irinotecan-induced intestinal mucositis, which may be one of the potential strategies to reduce the clinical side effects of irinotecan.
Collapse
Affiliation(s)
- Pei Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiaoyi Xiao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jing Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xingyuan Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Kaiwei Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Rongyao Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Dawei Wang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yanlong Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hongying Chen
- Guangzhou Baiyunshan Mingxing Pharmaceutical Co., Ltd, Guangzhou, 510250, China
| | - Zhiyong Xie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518106, China
| | - Qiongfeng Liao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
10
|
Peydayesh M, Kistler S, Zhou J, Lutz-Bueno V, Victorelli FD, Meneguin AB, Spósito L, Bauab TM, Chorilli M, Mezzenga R. Amyloid-polysaccharide interfacial coacervates as therapeutic materials. Nat Commun 2023; 14:1848. [PMID: 37012278 PMCID: PMC10070338 DOI: 10.1038/s41467-023-37629-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Coacervation via liquid-liquid phase separation provides an excellent opportunity to address the challenges of designing nanostructured biomaterials with multiple functionalities. Protein-polysaccharide coacervates, in particular, offer an appealing strategy to target biomaterial scaffolds, but these systems suffer from the low mechanical and chemical stabilities of protein-based condensates. Here we overcome these limitations by transforming native proteins into amyloid fibrils and demonstrate that the coacervation of cationic protein amyloids and anionic linear polysaccharides results in the interfacial self-assembly of biomaterials with precise control of their structure and properties. The coacervates present a highly ordered asymmetric architecture with amyloid fibrils on one side and the polysaccharide on the other. We demonstrate the excellent performance of these coacervates for gastric ulcer protection by validating via an in vivo assay their therapeutic effect as engineered microparticles. These results point at amyloid-polysaccharides coacervates as an original and effective biomaterial for multiple uses in internal medicine.
Collapse
Affiliation(s)
- Mohammad Peydayesh
- ETH Zurich, Department of Health Sciences and Technology, 8092, Zurich, Switzerland
| | - Sabrina Kistler
- ETH Zurich, Department of Materials, 8093, Zurich, Switzerland
| | - Jiangtao Zhou
- ETH Zurich, Department of Health Sciences and Technology, 8092, Zurich, Switzerland
| | - Viviane Lutz-Bueno
- ETH Zurich, Department of Health Sciences and Technology, 8092, Zurich, Switzerland
- Paul Scherrer Institute PSI, 5232, Villigen, Switzerland
| | | | - Andréia Bagliotti Meneguin
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University, 14800-903, Araraquara, Sao Paulo, Brazil
| | - Larissa Spósito
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University, 14800-903, Araraquara, Sao Paulo, Brazil
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University, 14800-903, Araraquara, Sao Paulo, Brazil
| | - Tais Maria Bauab
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University, 14800-903, Araraquara, Sao Paulo, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University, 14800-903, Araraquara, Sao Paulo, Brazil
| | - Raffaele Mezzenga
- ETH Zurich, Department of Health Sciences and Technology, 8092, Zurich, Switzerland.
- ETH Zurich, Department of Materials, 8093, Zurich, Switzerland.
| |
Collapse
|
11
|
Liu R, Zhu N, Hao Y, Liu X, Kang J, Mao R, Yu X, Li Y. The Protective Effect of Walnut Oligopeptides against Indomethacin-Induced Gastric Ulcer in Rats. Nutrients 2023; 15:nu15071675. [PMID: 37049515 PMCID: PMC10096596 DOI: 10.3390/nu15071675] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
The aim of this study was to investigate the potential protective effects of walnut oligopeptides (WOPs) on indomethacin-induced gastric ulcers in rats. The rats were divided into the following groups: normal group, model group, omeprazole group (0.02 g/kg), and WOPs groups (0.22, 0.44, and 0.88 g/kg, respectively). After receiving gavage once per day for 30 consecutive days, the rats were injected intraperitoneally with indomethacin 48 mg/kg to induce gastric ulcers. Then, the serum inflammatory cytokines and gastric prostaglandin E2 (PGE2), oxidative stress-related indicators, and the RNA expression of COX-1 and COX-2 were measured. The results revealed that WOPs confer significant gastroprotection on gastric ulcers caused by indomethacin, regulating inflammatory cytokines, oxidative stress, and prostaglandins synthesis, and enhancing the expression of COX-1 and COX-2 in gastric tissue, thus exerting its protective effect on gastric mucosa. The gastroprotective mechanism may be related to the involvement of the arachidonic acid metabolism and upregulation of tryptophan, phenylalanine, tyrosine, and alpha-Linolenic acid metabolism synthesis in vivo.
Collapse
Affiliation(s)
- Rui Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Na Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Yuntao Hao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Xinran Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Jiawei Kang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Ruixue Mao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Xiaochen Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
| |
Collapse
|
12
|
Protective Effects of Piperine on Ethanol-Induced Gastric Mucosa Injury by Oxidative Stress Inhibition. Nutrients 2022; 14:nu14224744. [PMID: 36432431 PMCID: PMC9695505 DOI: 10.3390/nu14224744] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022] Open
Abstract
Piper nigrum Linnaeus is often used as a treatment for chills, stomach diseases, and other ailments. Piperine has many biological functions; however, its mechanism for preventing gastric mucosal damage is still unclear. The objective of this study was to investigate the preventive effects of piperine on ethanol-induced gastric mucosal injury by using GES-1 cells and rats. SOD, CAT, GSH-Px and MDA were effectively regulated in GES-1 cells pre-treated with piperine. Piperine significantly increased SOD, CAT and GSH-Px activities, but decreased the ulcer area, MDA, ROS and MPO levels in the gastric tissues of rats. RT-PCR analysis showed that piperine downregulated the mRNA expression levels of keap1, JNK, ERK and p38, and upregulated the mRNA transcription levels of Nrf2 and HO-1. Western blotting results indicated that piperine could activate the protein expression levels of Nrf2 and HO-1 and inhibit the protein expression levels of keap1, p-JNK, p-ERK and p-p38. In conclusion, piperine suppressed ethanol-induced gastric ulcers in vitro and in vivo via oxidation inhibition and improving gastric-protecting activity by regulating the Nrf2/HO-1 and MAPK signalling pathways.
Collapse
|
13
|
Huang Q, Xin X, Sun Q, An Z, Gou X, Feng Q. Plant-derived bioactive compounds regulate the NLRP3 inflammasome to treat NAFLD. Front Pharmacol 2022; 13:896899. [PMID: 36016562 PMCID: PMC9396216 DOI: 10.3389/fphar.2022.896899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver disorder characterized by abnormal accumulation of hepatic fat and inflammatory response with complex pathogenesis. Over activation of the pyrin domain-containing protein 3 (NLRP3) inflammasome triggers the secretion of interleukin (IL)-1β and IL-18, induces pyroptosis, and promotes the release of a large number of pro-inflammatory proteins. All of which contribute to the development of NAFLD. There is a great deal of evidence indicating that plant-derived active ingredients are effective and safe for NAFLD management. This review aims to summarize the research progress of 31 active plant-derived components (terpenoids, flavonoids, alkaloids, and phenols) that alleviate lipid deposition, inflammation, and pyroptosis by acting on the NLRP3 inflammasome studied in both in vitro and in vivo NAFLD models. These studies confirmed that the NLRP3 inflammasome and its related genes play a key role in NAFLD amelioration, providing a starting point for further study on the correlation of plant-derived compounds treatment with the NLRP3 inflammasome and NAFLD.
Collapse
Affiliation(s)
- Qian Huang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Xin
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - QinMei Sun
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziming An
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaojun Gou
- Central Laboratory, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai, China
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
- *Correspondence: Qin Feng,
| |
Collapse
|
14
|
Tu H, Wang W, Feng Y, Zhang L, Zhou H, Cheng C, Ji L, Cai Q, Feng Y. β-Patchoulene represses hypoxia-induced proliferation and epithelial-mesenchymal transition of liver cancer cells. Bioengineered 2022; 13:11907-11922. [PMID: 35546067 PMCID: PMC9275994 DOI: 10.1080/21655979.2022.2065945] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor originating from liver epithelial cells with a high clinical mortality rate. β-Patchoulene (β-PAE) is a compound extracted from patchouli, which has analgesic, anti-inflammatory and antioxidant effects. This research aims to probe the impacts of β-PAE on hypoxia-induced HCC cell proliferation and epithelial-mesenchymal transition (EMT). Firstly, hypoxic injury models were constructed in HCC Huh-7 and MHCC97 cells, and the hypoxic injury cell models were then treated with different concentrations of β-PAE. The cell viability, proliferation, migration, invasion and apoptosis were checked by the cell counting kit-8 (CCK-8) assay, colony formation assay, Transwell assay, flow cytometry and terminal deoxyribonucleotide transferase (TdT)-mediated dUTP nick end labeling (TUNEL) assay. The expression of Survivin protein, EMT markers and the NF-κB/HIF-1α pathway was gauged by Western blot (WB) or cellular immunofluorescence or reverse transcription-polymerase chain reaction (RT-PCR). The in-vivo experiment was conducted to confirm the anti-tumor role of β-PAE. As a result, β-PAE abated hypoxia-induced HCC cell growth, proliferation, migration, invasion and EMT and facilitated apoptosis in vitro and in vivo dose-dependently. Further mechanism studies displayed that β-PAE inactivated the NF-κB/HIF-1α pathway, and HIF-1α activation significantly reversed the β-PAE-mediated tumor inhibition. β-PAE repressed the proliferation and EMT of hypoxia-induced HCC cells by choking the NF-κB/HIF-1α pathway, suggesting that β-PAE was a potential drug for HCC treatment.
Collapse
Affiliation(s)
- Huahua Tu
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Yanqing Feng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Linfei Zhang
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Huadong Zhou
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Caitao Cheng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Lei Ji
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Qinghe Cai
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Yong Feng
- Department of Hepatobiliary Surgery, Renmin Hospital, Hubei University of Medicine, Shiyan, P.R. China
| |
Collapse
|
15
|
Xu F, Cai W, Ma T, Zeng H, Kuang X, Chen W, Liu B. Traditional Uses, Phytochemistry, Pharmacology, Quality Control, Industrial Application, Pharmacokinetics and Network Pharmacology of Pogostemon cablin: A Comprehensive Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:691-721. [PMID: 35282804 DOI: 10.1142/s0192415x22500288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pogostemonis Herba (PH) is the dried aerial parts of Pogostemon cablin (Blanco) Benth, which is mainly distributed and used in Asian countries. PH is an aromatic damp-resolving drug in traditional Chinese medicine (TCM), which is usually used for the treatment of vomiting, chest tension, tiredness, abdominal pain, diarrhea, and headache. In this review, the summary of chemical constituents in the aerial parts, biological activities, history of uses, quality control methods, industrial applications, pharmacokinetics and network pharmacology are reported. By collating the chemical constituents of various parts of PH, a total of 174 components were identified, including 66 terpenes, 6 pyrones, 40 flavonoids, 21 phenylpropanoids, 9 steroids, 4 polysaccharides and 28 others. Pharmacological research has found that PH possesses multi-pharmacological activities, including regulating the gastrointestinal tract, inhibition of pathogenic microorganisms, and anti-inflammation, which provide more scientific interpretation for the clinical usage of PH. In addition, the shortcomings of the current research on PH and the recommendation of future studies on PH are analyzed. We hope this review can provide some insight for further research and applications of PH in future.
Collapse
Affiliation(s)
- Fangfang Xu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Wanna Cai
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Ting Ma
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Huimei Zeng
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaolan Kuang
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Weiying Chen
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| | - Bo Liu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Clinical Medicial College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Guangzhou Key Laboratory of Chirality Research on Active Components of Traditional Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
16
|
Li M, Lv R, Xu X, Ge Q, Lin S. Tricholoma matsutake-Derived Peptides Show Gastroprotective Effects against Ethanol-Induced Acute Gastric Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14985-14994. [PMID: 34866395 DOI: 10.1021/acs.jafc.1c07050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Acute gastric injury caused by ethanol is a frequent disorder of the gastrointestinal tract. In this study, we investigated the potential gastroprotective effects of Tricholoma matsutake-derived peptides against ethanol-triggered acute gastric injury and the associated mechanisms. Peptides SDLKHFPF and SDIKHFPF significantly attenuated the ethanol-induced decrease in GES-1 cell survival (82.39 ± 1.93 and 80.10 ± 1.08% vs 56.58 ± 1.86%), inhibited GES-1 cell apoptosis, and alleviated the ethanol-induced gastric mucosal injury (64.76 ± 3.98 and 49.29 ± 3.25%), ulcer index (3.33 ± 0.47 and 4.67 ± 0.47 vs 6.67 ± 0.47), and histopathological changes in mice. Peptide treatment inhibited the phosphorylation and nuclear translocation of nuclear factor kappa B (NF-κB), the secretion of tumor necrosis factor-α, interleukin-6, and endothelin-1. In addition, T. matsutake peptide pretreatment increased growth factor secretion, upregulated B-cell lymphoma-2, downregulated Bcl-2-associated X (Bax), and cleaved cysteinyl aspartate specific proteinase 3, thereby promoting gastric cell survival. These findings strongly suggest that T. matsutake peptides attenuate ethanol-induced inflammatory responses and apoptosis by suppressing NF-κB signaling activation, thereby enhancing gastric epithelial barrier functions.
Collapse
Affiliation(s)
- Mengqi Li
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Renzhi Lv
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Xiaomeng Xu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Qi Ge
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
17
|
Wu J, Gan Y, Luo H, Xu N, Chen L, Li M, Guan F, Su Z, Lin Z, Xie J, Liu Y. β-Patchoulene Ameliorates Water Transport and the Mucus Barrier in 5-Fluorouracil-Induced Intestinal Mucositis Rats via the cAMP/PKA/CREB Signaling Pathway. Front Pharmacol 2021; 12:689491. [PMID: 34512326 PMCID: PMC8424048 DOI: 10.3389/fphar.2021.689491] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Intestinal mucositis (IM) is the main side effect observed in patients who receive cancer chemotherapy. The characteristics of ulceration, vomiting, and severe diarrhea cause patients to delay or abandon further treatment, thereby aggravating their progress. Hence, IM cannot be overlooked. β-patchoulene (β-PAE) is an active ingredient isolated from Pogostemon cablin (Blanco) Benth (Labiatae) and has shown a marked protective effect against gastrointestinal diseases in previous studies. However, whether β-PAE plays a positive role in IM is still unknown. Herein, we explore the effects and the underlying mechanism of β-PAE against 5-fluorouracil (5-FU)-induced IM in IEC-6 cells and rats. β-PAE significantly recovered cell viability, upregulated the IM-induced rat body weight and food intake and improved the pathological diarrhea symptoms. Aquaporin is critical for regulating water fluid homeostasis, and its abnormal expression was associated with pathological diarrhea in IM. β-PAE displayed an outstanding effect in inhibiting aquaporin 3 (AQP3) via the cAMP/protein kinase A (PKA)/cAMP-response element-binding protein (CREB) signaling pathway. Besides, inflammation-induced mucus barrier injury deteriorated water transport and aggravated diarrhea in IM-induced rats. β-PAE’s effect on suppressing inflammation and recovering the mucus barrier strengthened its regulation of water transport and thus alleviated diarrhea in IM-induced rats. In sum, β-PAE improved IM in rats mainly by improving water transport and the mucus barrier, and these effects were correlated with its function on inhibiting the cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Jiazhen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuxuan Gan
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huijuan Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Nan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Mengyao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengkun Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Basic Research, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Zhixiu Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhui Xie
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Basic Research, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, China
| |
Collapse
|
18
|
Luo H, Xu N, Wu J, Gan Y, Chen L, Guan F, Li M, Li Y, Chen J, Su Z, Liu Y. β-patchoulene protects against non-alcoholic steatohepatitis via interrupting the vicious circle among oxidative stress, histanoxia and lipid accumulation in rats. Int Immunopharmacol 2021; 98:107915. [PMID: 34198236 DOI: 10.1016/j.intimp.2021.107915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/06/2023]
Abstract
Non-alcoholic steatohepatitis (NASH), an extreme progressive subtype of metabolic associated fatty liver disease, is well characterized by hepatic steatosis, injury and inflammation. It causes irreversible hepatic damage and there are no approved interventions for it. β-PAE, a representatively pharmacological active substance isolated from Pogostemon cablin, has been indicated to alleviate hepatic steatosis and injury through modulating lipid metabolism in rats with simple steatosis. However, its protection against NASH remains unclear. Here, this study explored the potential effect of β-PAE against high-fat diet-induced NASH in rats. The results displayed that β-PAE significantly reduced the gains of body weight and epididymal adipose tissue, liver index and attenuated liver histological damages in NASH rats. It also markedly alleviated hepatic inflammation by inhibiting NLRP3 inflammasome activation. In NASH, the active NLRP3 inflammasome is caused by hepatic lipid abnormal accumulation-induced oxidative stress. Excessive oxidative stress results in hepatic histanoxia, which exacerbates lipid metabolism disorders by elevating CD36 to suppress AMPK signalling pathways. Moreover, the lipid accumulation led by lipid metabolism dysfunction intensifies oxidative stress. A vicious circle is formed among oxidative stress, histanoxia and lipid accumulation, eventually, but β-PAE effectively interrupted it. Interestingly, soluble CD36 (sCD36) was tightly associated not only with hepatic steatosis and injury but also with inflammation. Collectively, β-PAE exerted a positive effect against NASH by interrupting the vicious circle among oxidative stress, histanoxia and lipid accumulation, and sCD36 may be a promising non-invasive tool for NASH diagnosis.
Collapse
Affiliation(s)
- Huijuan Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Nan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiazhen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuxuan Gan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Fengkun Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Mengyao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523808, China.
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523808, China.
| |
Collapse
|
19
|
Tong S, Wang H, A LS, Bai TN, Gong JH, Jin WJ, Dai LL, Ba GN, Cho SB, Fu MH. Protective effect and mechanisms of action of Mongolian medicine Sulongga-4 on pyloric ligation-induced gastroduodenal ulcer in rats. World J Gastroenterol 2021; 27:1770-1784. [PMID: 33967556 PMCID: PMC8072194 DOI: 10.3748/wjg.v27.i16.1770] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/29/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sulongga-4 (SL-4) is a herbal formula used in traditional Mongolian medical clinics for the treatment of peptic ulcers and gastroenteritis, even though its pharmacological mechanism has not been well characterized.
AIM To evaluate the protective effect and identify the mechanisms of action of SL-4 on gastroduodenal ulcer induced by pyloric ligation (PL) in rats.
METHODS PL was performed to induce gastric and duodenal ulcers in rats, which were then treated with oral SL-4 (1.3, 2.6, or 3.9 g/kg per day) for 15 d. PL-induced gastroduodenal ulceration. Therapeutic effects were characterized by pathological and histological evaluations and inflammatory indicators were analyzed by enzyme-linked immunosorbent assay. Microarray analyses were conducted to identify gene expression profiles of gastroduodenal tissue in PL rats with or without SL-4 treatment. The candidate target genes were selected and verified by quantitative reverse transcription polymerase chain reaction (qRT-PCR).
RESULTS SL-4 decreased histopathological features in the PL-induced ulcerated rats. SL-4 significantly (P < 0.05) decreased expression of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, endotoxin, platelet-activating factor, and increased prostaglandin E2 and epidermal growth factor in ulcer tissue. Microarray analysis was used to identify a panel of candidate target genes for SL-4 acting on PL-induced ulceration. Genes included some complement and coagulation cascade and retinol metabolism pathways that are closely associated with inflammatory responses and gastric mucosal protective mechanisms. qRT-PCR showed that altered expression of the selected genes, such as CYP2b2, UGT2b1, A2m, and MASP1 was consistent with the microarray results.
CONCLUSION SL-4 exerts protective effects against PL-induced gastroduodenal ulcers via reducing inflammatory cytokines and elevating expression of gastric acid inhibitory factors. Downregulation of CYP2b2 and UGT2b1 genes in retinol metabolism and upregulation of A2m and MASP1 genes in the complement and coagulation cascades pathways are possibly involved in SL-4-mediated protection against gastroduodenal ulcer.
Collapse
Affiliation(s)
- Shan Tong
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
- Mongolian Medicine Surgery Department, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Huan Wang
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Li-Sha A
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
- Traditional Mongolian Medicine Research Institute, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Ta-Na Bai
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Ju-Hua Gong
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Wen-Jie Jin
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
- Traditional Mongolian Medicine Research Institute, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Li-Li Dai
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
- Traditional Mongolian Medicine Research Institute, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Gen-Na Ba
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Sung-Bo Cho
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Ming-Hai Fu
- School of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
- Traditional Mongolian Medicine Research Institute, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| |
Collapse
|
20
|
Xu N, Luo H, Li M, Wu J, Wu X, Chen L, Gan Y, Guan F, Li M, Su Z, Chen J, Liu Y. β-patchoulene improves lipid metabolism to alleviate non-alcoholic fatty liver disease via activating AMPK signaling pathway. Biomed Pharmacother 2020; 134:111104. [PMID: 33341045 DOI: 10.1016/j.biopha.2020.111104] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has been a leading cause of chronic metabolic disease, seriously posing healthy burdens to the public, whereas interventions available for it are limited to date. Patchouli oil had been reported to attenuate hepatic steatosis in our previous study. β-patchoulene (β-PAE) is a representative component separated from patchouli oil with multiple activities, but its effect against NAFLD is still unknown. To investigate the effect and potential mechanism of β-PAE on NAFLD, we used high fat diet (HFD) in vivo and free fatty acid (FFA) in vitro to induce hepatic steatosis in rats and L02 cells, respectively. Histological examination was evaluated via Hematoxylin-eosin and oil red O staining. The parameters for hepatic steatosis were estimated via biochemical kits, western blotting and quantitative real-time PCR. Compound C, the inhibitor of AMPK, was applied further to examine the precise mechanism of β-PAE on NAFLD. Our results indicated that β-PAE significantly attenuated HFD-induced weight gain, hepatic injury, lipid deposition in serum and hepatic tissue as well as FFA induced-lipid accumulation. Besides, β-PAE markedly improved the expression of AMP-activated protein kinase (AMPK) and its downstream factors which correlate with hepatic lipid synthesis and oxidation in vivo and in vitro. Nevertheless, Compound C abrogated the benefits derived from β-PAE in L02 cells. In conclusion, these results suggest that β-PAE exerts AMPK agonist-like effect to regulate hepatic lipid synthesis and oxidation, eventually prevent NAFLD progression.
Collapse
Affiliation(s)
- Nan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huijuan Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Minyao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jiazhen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xue Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Yuxuan Gan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Fengkun Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Mengyao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan, 523808, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan, 523808, China.
| |
Collapse
|
21
|
Pogostemon cablin Triggered ROS-Induced DNA Damage to Arrest Cell Cycle Progression and Induce Apoptosis on Human Hepatocellular Carcinoma In Vitro and In Vivo. Molecules 2020; 25:molecules25235639. [PMID: 33266043 PMCID: PMC7731310 DOI: 10.3390/molecules25235639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/27/2020] [Accepted: 11/27/2020] [Indexed: 12/18/2022] Open
Abstract
The purpose of the study was to elucidate the anti-hepatoma effects and mechanisms of Pogostemon cablin essential oils (PPa extract) in vitro and in vivo. PPa extract exhibited an inhibitory effect on hepatocellular carcinoma (HCC) cells and was less cytotoxic to normal cells, especially normal liver cells, than it was to HCC cells, exerting a good selective index. Additionally, PPa extract inhibited HCC cell growth by blocking the cell cycle at the G0/G1 phase via p53 dependent or independent pathway to down regulated cell cycle regulators. Moreover, PPa extract induced the FAS-FASL-caspase-8 system to activate the extrinsic apoptosis pathway, and it increased the bax/bcl-2 ratio and reduced ΔΨm to activate the intrinsic apoptosis pathway that might be due to lots of reactive oxygen species (ROS) production which was induced by PPa extract. In addition, PPa extract presented to the potential to act synergistically with sorafenib to effectively inhibit HCC cell proliferation through the Akt/mTOR pathway and reduce regrowth of HCC cells. In an animal model, PPa extract suppressed HCC tumor growth and prolonged lifespan by reducing the VEGF/VEGFR axis and inducing tumor cell apoptosis in vivo. Ultimately, PPa extract demonstrated nearly no or low system-wide, physiological, or pathological toxicity in vivo. In conclusion, PPa extract effectively inhibited HCC cell growth through inducing cell cycle arrest and activating apoptosis in vitro and in vivo. Furthermore, PPa extract exhibits less toxicity toward normal cells and organs than it does toward HCC cells, which might lead to fewer side effects in clinical applications. PPa extract may be developed into a clinical drug to suppress tumor growth or functional food to prevent HCC initiation or chemoprotection of HCC recurrence.
Collapse
|
22
|
Serafim C, Araruna ME, Júnior EA, Diniz M, Hiruma-Lima C, Batista L. A Review of the Role of Flavonoids in Peptic Ulcer (2010-2020). Molecules 2020; 25:molecules25225431. [PMID: 33233494 PMCID: PMC7699562 DOI: 10.3390/molecules25225431] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Peptic ulcers are characterized by erosions on the mucosa of the gastrointestinal tract that may reach the muscle layer. Their etiology is multifactorial and occurs when the balance between offensive and protective factors of the mucosa is disturbed. Peptic ulcers represent a global health problem, affecting millions of people worldwide and showing high rates of recurrence. Helicobacter pylori infection and the use of non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most important predisposing factors for the development of peptic ulcers. Therefore, new approaches to complementary treatments are needed to prevent the development of ulcers and their recurrence. Natural products such as medicinal plants and their isolated compounds have been widely used in experimental models of peptic ulcers. Flavonoids are among the molecules of greatest interest in biological assays due to their anti-inflammatory and antioxidant properties. The present study is a literature review of flavonoids that have been reported to show peptic ulcer activity in experimental models. Studies published from January 2010 to January 2020 were selected from reference databases. This review refers to a collection of flavonoids with antiulcer activity in vivo and in vitro models.
Collapse
Affiliation(s)
- Catarina Serafim
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051900, Paraiba, Brazil; (C.S.); (M.E.A.); (E.A.J.)
| | - Maria Elaine Araruna
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051900, Paraiba, Brazil; (C.S.); (M.E.A.); (E.A.J.)
| | - Edvaldo Alves Júnior
- Postgraduate Program in Natural Products and Bioactive Synthetic, Health Sciences Center, Federal University of Paraiba, João Pessoa 58051900, Paraiba, Brazil; (C.S.); (M.E.A.); (E.A.J.)
| | - Margareth Diniz
- Department of Pharmacy, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051900, Paraiba, Brazil;
| | - Clélia Hiruma-Lima
- Department of Structural and Functional Biology (Physiology), Institute of Biosciences, São Paulo State University, Botucatu 18618970, São Paulo, Brazil;
| | - Leônia Batista
- Department of Pharmacy, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051900, Paraiba, Brazil;
- Correspondence: ; Tel.: +55-83-32167003; Fax: +55-83-32167502
| |
Collapse
|
23
|
Mohan S, Hobani YH, Shaheen E, Abou-Elhamd AS, Abdelhaleem A, Alhazmi HA, Abdelwahab SI. Ameliorative effect of Boesenbergin A, a chalcone isolated from Boesenbergia rotunda (Fingerroot) on oxidative stress and inflammation in ethanol-induced gastric ulcer in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113104. [PMID: 32565307 DOI: 10.1016/j.jep.2020.113104] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Boesenbergia rotunda (L.) Mansf. Kulturpfl., previously known as Boesenbergia pandurata (Family: Zingiberaceae) is a ginger species, locally known as fingerroot. It is an integral part of Southeast Asian traditional medicine in alleviating many gastrointestinal disorders such as flatulence, carminative, stomach ache, dyspepsia, and peptic ulcer. AIM OF THE STUDY Earlier we have investigated the cytoprotective effect of Boesenbergia rotunda extract. In the present study, we investigated the gastroprotection activity of Boesenbergin A (BA), a chalcone isolated from Boesenbergia rotunda extract in ethanol-induced ulcer model in rats. Besides, the contribution of anti-inflammatory and anti-oxidant ability of BA as probable mechanisms involved in the anti-ulcer activity, also been studied. MATERIALS AND METHODS BA was orally administered in rats before ulcer induction with ethanol. The lesions of the gastric mucosa were evaluated macroscopically and histopathologically. The efficiency of BA in mucus production, NO production, PGE2 synthesis, mucosal nonprotein sulphydryls, glutathione (GSH) level, and lipid peroxidation (MDA) level were studied. The involvement of the anti-inflammatory capacity of BA was analyzed by using the measurement of cytokines such as TNF-α and IL-6. Finally, the expression of biomarkers such as HSP 70 and iNOS was analyzed at the transcriptional and translational levels. RESULTS We confirmed the protective capacity of BA via the reduction of ulcerated and haemorrhagic areas. It has induced the protection through lowering GSH, MDA and increased NP-SH level. The plasma NO levels were significantly less in BA treated rats. Both cytokines TNF-α and IL-6 were decreased together with elevated PGE2. Upregulation of HSP and downregulation of iNOS were determined in immunohistochemical and gene expression studies CONCLUSIONS: The current results suggest that the prophylactic effect found with BA is due to (i) boosting of gastric mucus production and suppression of inflammatory mediators, via pro-inflammatory cytokines and (ii) modulating the oxidative stress response. The usefulness of Boesenbergia rotunda in folk medicine in treating ulcers partially could be due to the presence of this chalcone.
Collapse
Affiliation(s)
- Syam Mohan
- Substance Abuse and Toxicology Research Center, Jazan University, PO Box 114, Jazan, Saudi Arabia.
| | - Yahya Hasan Hobani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science, Jazan University, PO Box 114, Jazan, Saudi Arabia
| | - Emad Shaheen
- Medical Research Centre, Jazan University, PO Box 114, Jazan, Saudi Arabia
| | - Alaa Sayed Abou-Elhamd
- Department of Respiratory Care, Faculty of Applied Medical Science, PO Box 114, Jazan, Saudi Arabia; Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Egypt
| | - Aymen Abdelhaleem
- Medical Research Centre, Jazan University, PO Box 114, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, PO Box 114, Jazan, Saudi Arabia
| | | |
Collapse
|
24
|
Yi L, Lu Y, Yu S, Cheng Q, Yi L. Formononetin inhibits inflammation and promotes gastric mucosal angiogenesis in gastric ulcer rats through regulating NF-κB signaling pathway. J Recept Signal Transduct Res 2020; 42:16-22. [PMID: 33100111 DOI: 10.1080/10799893.2020.1837873] [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] [Indexed: 12/15/2022]
Abstract
To investigate the effects of formononetin on rats with gastric ulcer and further to explore its possible mechanism. Rats were randomly divided into sham operation group (Sham), model group (Model), omeprazole control group (Omeprazole) and formononetin in different dose groups (FOR-L, FOR-M, FOR-H). Rats model with gastric ulcer were established by 100% glacial acetic acid. Hematoxylin-eosin (H&E) staining was used to observe the pathological morphology of gastric mucosa. Immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) were used to detect the level of inflammatory and angiogenesis related factors. The expressions of nuclear factor kappa-B (NF-κB) signaling pathway-related proteins were detected by western blot. Formononetin and omeprazole could ameliorate the pathological morphology of gastric mucosa in gastric ulcer rats. Compared with Model group, the levels of tumor necrosis factor (TNF)-α, Interleukin (IL)-1β, IL-6, myeloperoxidase (MPO), human endothelin (ET)-1 and p-P65 protein in formononetin treatment and omeprazole groups were significantly decreased (p < 0.05). Moreover, formononetin could increase the content of vascular endothelial growth factor (VEGF), nitric oxide (NO) and the levels of CD34, tight junction proteins (ZO-1 and occludin) and p-IκBα in a dose-dependent manner. Formononetin can ameliorate gastric ulcer in rats by inhibiting inflammation and promoting gastric mucosal angiogenesis, and its mechanism maybe related to NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lanjie Yi
- Research Office of Clinical Documentation, Nanjing Institute of Traditional Chinese Medicine, Nanjing, China
| | - Yan Lu
- Department of Rheumatology, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Shun Yu
- Research Office of Clinical Documentation, Nanjing Institute of Traditional Chinese Medicine, Nanjing, China
| | - Qian Cheng
- Nanjing University of Traditional Chinese Medicine Library, Nanjing, China
| | - Lanjuan Yi
- Department of Gastroenterology, Yantai Shan Hospital, Yantai, China
| |
Collapse
|
25
|
Yang M, Yan T, Yu M, Kang J, Gao R, Wang P, Zhang Y, Zhang H, Shi L. Advances in understanding of health‐promoting benefits of medicine and food homology using analysis of gut microbiota and metabolomics. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.49] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Minmin Yang
- College of Life Sciences Shaanxi Normal University Xi'an China
| | - Tao Yan
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Meng Yu
- The Institute of Medicinal Plant Development Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jie Kang
- Physical Education Institute Shaanxi Normal University Xi'an China
| | - Ruoxi Gao
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Peng Wang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Yuhuan Zhang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Huafeng Zhang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
- Internatinal Joint Research Center of Shaanxi Province for Food and Health Science Shaanxi Normal University Xi'an China
| | - Lin Shi
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
- Internatinal Joint Research Center of Shaanxi Province for Food and Health Science Shaanxi Normal University Xi'an China
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| |
Collapse
|
26
|
Lian YZ, Lin IH, Yang YC, Chao JCJ. Gastroprotective effect of Lycium barbarum polysaccharides and C-phycocyanin in rats with ethanol-induced gastric ulcer. Int J Biol Macromol 2020; 165:1519-1528. [PMID: 33058973 DOI: 10.1016/j.ijbiomac.2020.10.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022]
Abstract
This study investigated the gastroprotective effect of Lycium barbarum polysaccharides (LBP) and C-phycocyanin (C-PC) in rats with ethanol-induced gastric ulcer. Rats were divided into 5 groups: normal, ulcer, ulcer treated with 100 mg/kg bw LBP, ulcer treated with 50 mg/kg bw C-PC, and ulcer treated with 50 mg/kg bw LBP and 25 mg/kg bw C-PC. Pretreatment with LBP and/or C-PC was given a week before ulcer induction. Ulcer induction was produced by 50% ethanol administration orally every other day for 4 weeks. After 5-week treatment, the histopathological observation showed that LBP or C-PC attenuated the severity of gastric mucosal damage. LBP decreased serum malondialdehyde (MDA) levels and gastric interleukin-6 (IL-6), intercellular adhesion molecule-1 (ICAM-1) levels, and myeloperoxidase (MPO) activity. C-PC decreased serum MDA levels and gastric tumor necrosis factor-α (TNF-α), IL-1β, IL-6, ICAM-1 levels, and MPO activity. Combined LBP and C-PC decreased serum MDA levels and gastric TNF-α, IL-1β, IL-6, and ICAM-1 levels. LBP and/or C-PC increased gastric heat shock protein 70 and non-protein sulfhydryl compounds. Rats with ulcer and treatment had enriched with the family Bacillaceae. Therefore, pretreatment with LBP and/or C-PC attenuated ethanol-induced gastric ulcer in rats via suppressing oxidation and inflammation and increasing gastroprotection.
Collapse
Affiliation(s)
- Yu Zhi Lian
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan.
| | - I-Hsuan Lin
- Research Center of Translational Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Yu-Chen Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, 110, Taiwan.
| | - Jane C-J Chao
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan; Master Program in Global Health and Development, Taipei Medical University, Taipei 110, Taiwan; Nutrition Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
27
|
Song H, Hou X, Zeng M, Chen X, Chen X, Yang T, Xu F, Peng J, Peng Q, Cai X, Yu R. Traditional Chinese Medicine Li-Zhong-Tang accelerates the healing of indomethacin-induced gastric ulcers in rats by affecting TLR-2/MyD88 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 259:112979. [PMID: 32442585 DOI: 10.1016/j.jep.2020.112979] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/25/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Li-Zhong-Tang (LZT) is a well-known Chinese herbal formulation first described in one of traditional Chinese medicine (TCM) scriptures, Treatise on Febrile Diseases. LZT has been commonly prescribed for the treatment of various gastrointestinal diseases for over 1800 years, and has demonstrated pronounced therapeutic effects on patients with gastric ulcers. AIM OF THE STUDY The present study aimed to scientifically evaluate protective effects of LZT on indomethacin (IND)-induced gastric injury in rats and to elucidate whether LZT exerts its gastro-protective effects via enhancing mucosal immunity by regulating TLR-2/MyD88 signaling pathway. MATERIAL AND METHODS Gastric ulcers were induced in male Sprague-Dawley (SD) rats with a single oral dose of 150 mg/kg IND. Ulcer index (UI) and curative index (CI) were evaluated. Histopathological examinations were performed and microscopic score (MS) was macroscopically calculated. The volume of gastric juice, free acidity, total acidity, and gastric pH was measured. The gastroprotective and inflammatory biomarkers including levels of nitric oxide (NO), tumor necrosis factor-α (TNF-α), prostaglandin E2 (PGE2), and malondialdehyde (MDA) were determined. Expression levels of TLR-2 and MyD88 mRNA were assessed by qRT-PCR. The expression, distribution, and co-localization of TLR-2 and MyD88 protein were determined by Western blot, immunohistochemistry, and immunofluorescence, respectively. RESULTS Induction of gastric ulcers in rats resulted in very significantly increased UI and elevated volume and acidity of gastric juice, which were markedly attenuated by LZT treatment. Microscopic examinations of the IND-induced gastric ulcers revealed severe gastric hemorrhagic necrosis, submucosal edema, and destruction of epithelial cells, which were significantly attenuated in LZT-treated rats. Moreover, treatment with LZT remarkably increased gastric mucosal levels of PGE2 and NO, and lowered highly elevated levels of TNF-α and MDA in gastric ulcerative rats. Mechanistically, LZT inhibited mRNA and protein expression of TLR-2 and MyD88 and enhanced immune function in gastric mucosa. Immunohistochemical analyses and immunofluorescent detection further confirmed a markedly decreased co-localization of TLR-2 and MyD88 protein in the gastric mucosa of LZT-treated rats as compared to that of gastric ulcerative rats. CONCLUSIONS These findings indicate that LZT alleviates serious gastric mucosal ulcerations induced by IND. Protective effects of LZT on gastric ulcers are believed to be associated with the intensification of the anti-oxidative defense system, mitigation of proinflammatory cytokines, stimulation of the production of cytoprotective mediators, and improvement of the mucosal immunity through TLR-2/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Houpan Song
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xueqin Hou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai' an, Shandong, 271016, China.
| | - Meiyan Zeng
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xiaojuan Chen
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xinyi Chen
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Tao Yang
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Fuping Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Jun Peng
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Qinghua Peng
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Xiong Cai
- Hunan Provincial Key Laboratory of Diagnostic Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China; Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| | - Rong Yu
- Hunan Provincial Key Laboratory of Translational Research in TCM Prescriptions and Zheng, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.
| |
Collapse
|
28
|
Hong SJ, Cho J, Boo CG, Youn MY, Pan JH, Kim JK, Shin EC. Inhalation of Patchouli ( Pogostemon Cablin Benth.) Essential Oil Improved Metabolic Parameters in Obesity-Induced Sprague Dawley Rats. Nutrients 2020; 12:E2077. [PMID: 32668680 PMCID: PMC7400805 DOI: 10.3390/nu12072077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
This study investigated effects of patchouli essential oil (PEO) inhalation on metabolic parameters. First, to characterize aromatic compounds in PEO, solid-phase microextraction-gas chromatography/mass spectrometric detection was employed in which 19 aromatic compounds were identified. In GC-olfactometry analysis, linalool, α-patchoulene, and β-patchoulene were found to be the constituents exhibiting the highest similarity to the aromatic compounds in patchouli. In an animal experiment using Sprague Darley rats, groups with PEO inhalation had a reduced food intake compared to the control group. Additionally, body weight was lower in the obesity-induced animal model exposed to PEO inhalation than the group without PEO. However, we found no significant difference in organ weights between groups. In our serum analysis, high-density lipoprotein cholesterol was significantly higher in the PEO inhalation groups, while low-density lipoprotein cholesterol content was highest in the positive control group, suggesting that inhalation of the aromatic compounds present in patchouli may improve cholesterol profile. In addition, leptin levels were reduced in the groups treated with PEO inhalation, which explains the differences in food intake and body weight gains. Last, animal groups exposed to PEO inhalation showed a relatively lower systolic blood pressure which suggests that inhalation of PEO (or aromatic compounds therein) may assist in regulating blood pressure. Collectively, our data demonstrate that the inhalation of PEO influenced certain markers related to metabolic diseases, hence provide basic data for future research as to preventive/therapeutic applications of PEO as well as their aromatic constituents.
Collapse
Affiliation(s)
- Seong Jun Hong
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea; (S.J.H.); (J.C.); (C.G.B.); (M.Y.Y.)
| | - Jinju Cho
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea; (S.J.H.); (J.C.); (C.G.B.); (M.Y.Y.)
| | - Chang Guk Boo
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea; (S.J.H.); (J.C.); (C.G.B.); (M.Y.Y.)
| | - Moon Yeon Youn
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea; (S.J.H.); (J.C.); (C.G.B.); (M.Y.Y.)
| | - Jeong Hoon Pan
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA; (J.H.P.); (J.K.K.)
| | - Jae Kyeom Kim
- Department of Behavioral Health and Nutrition, University of Delaware, Newark, DE 19716, USA; (J.H.P.); (J.K.K.)
| | - Eui-Cheol Shin
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Korea; (S.J.H.); (J.C.); (C.G.B.); (M.Y.Y.)
| |
Collapse
|
29
|
Transcriptomic Analysis of Fuzi Lizhong Decoction for the Treatment of Stomach Ulcers. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3291853. [PMID: 32148538 PMCID: PMC7054793 DOI: 10.1155/2020/3291853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/31/2019] [Accepted: 01/17/2020] [Indexed: 12/22/2022]
Abstract
This study aims to understand the treatment of stomach ulcers with FLD and to identify its potential target genes as well as related pathways by transcriptomic analysis. Rat stomach mRNA from a blank group (BG), a model group (MG), an untreated-model group (u-MG), and a treated group (TG) was sequenced. A partial least-squares discriminant analysis (PLS-DA) was used to differentiate the MG from the BG, and the Deseq2 R Package was used to identify differentially expressed genes between these groups. Furthermore, t-tests based on transcripts per million (TPM) were used to select different genes between MG and BG and significantly retrieved genes in TG, except for self-retrieved genes in u-MG. Finally, pathways regulated by retrieved genes were analyzed with KEGG database. Results showed that 327 of the 32,623 total detected genes were different (p < 0.05) between the MG and BG. Among these genes, eighteen genes were significantly retrieved after rats were treated with FLD in TG, and they were considered as target genes on which FLD acted. In conclusion, FLD was deduced to cure stomach ulcers by affecting glycerolipid metabolism, fatty acid degradation, circadian entrainment, circadian rhythm, and dopaminergic synapse.
Collapse
|
30
|
Huang P, Tang W, Shen R, Ju X, Shao G, Xu X, Jiang A, Qian X, Chen M, Zhou Z, Ren C. Analysis of candidate biomarkers and related transcription factors involved in the development and restoration of stress-induced gastric ulcer by transcriptomics. Cell Stress Chaperones 2020; 25:265-275. [PMID: 32088906 PMCID: PMC7058781 DOI: 10.1007/s12192-020-01070-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 01/01/2023] Open
Abstract
Stress-induced gastric ulcer is one of the common complications affecting patients after trauma, mainly leading to gastrointestinal bleeding and perforation, and severe cases may be life-threatening. However, the molecular mechanism of stress-induced gastric ulcer remains unclear. In the present study, RNA-sequencing was performed on gastric tissues of normal rats (C), stress-induced gastric ulcer rats (T0), and rats recovered from gastric ulcer for 3 days (T3), and bioinformatics analysis was performed to determine changes in gene expression and biological pathways. The protein-protein interaction (PPI) networks of differentially expressed genes (DEGs) were constructed by STRING and visualized by the Cytoscape software. The associated transcriptional factor (TFs)-gene regulatory network of the hub DEGs was also constructed. Pairwise comparisons obtained 103 (T0_C), 127 (T3_T0), and 13 (T3_C) DEGs, respectively. Gene ontology (GO) enrichment analysis indicated DEGs in T0_C and T3_T0 were significantly enriched in response to oxygen-containing compound, response to organic substance, and response to external stimulus. Pathway analysis suggested that DEGs were enriched in TNF signaling pathway, PPAR signaling pathway, apoptosis, and IL-17 signaling pathway. Seven hub genes (Fos, Jun, Nfkbia, Dusp1, Pim3, Junb, and Fosb) were obtained from the PPI networks of T0_C and T3_T0. Key TFs with close interactions, such as Fos, Jun, Nfkbia, Junb, Egr1, and Fosb, were screened This study used RNA-sequencing and bioinformatics analysis to screen out genes associated with gastric ulcer, which can help reveal the molecular mechanism of gastric ulcer development and restoration, and provide reference for the treatment of human gastric ulcers.
Collapse
Affiliation(s)
- Pan Huang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Weihong Tang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Rong Shen
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Genbao Shao
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Xiao Xu
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Anqi Jiang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Xiaobin Qian
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Miao Chen
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Zhengrong Zhou
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| | - Caifang Ren
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu People’s Republic of China
| |
Collapse
|
31
|
Quality assessment and differentiation of Aucklandiae Radix and Vladimiriae Radix based on GC-MS fingerprint and chemometrics analysis: basis for clinical application. Anal Bioanal Chem 2020; 412:1535-1549. [DOI: 10.1007/s00216-019-02380-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/06/2019] [Accepted: 12/27/2019] [Indexed: 12/18/2022]
|
32
|
Chen H, Nie Q, Xie M, Yao H, Zhang K, Yin J, Nie S. Protective effects of β-glucan isolated from highland barley on ethanol-induced gastric damage in rats and its benefits to mice gut conditions. Food Res Int 2019; 122:157-166. [DOI: 10.1016/j.foodres.2019.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
|
33
|
Zhang FB, Wang JP, Zhang HX, Fan GM, Cui X. Effect of β-patchoulene on cerebral ischemia-reperfusion injury and the TLR4/NF-κB signaling pathway. Exp Ther Med 2019; 17:3335-3342. [PMID: 30988709 PMCID: PMC6447785 DOI: 10.3892/etm.2019.7374] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 01/11/2019] [Indexed: 12/14/2022] Open
Abstract
β-patchoulene (β-PAE), an active constituent of the Pogostemon cablin, is well known for its anti-inflammatory and antioxidative functions in various diseases. However, little is known about the impact of β-PAE on the cerebral ischemia-reperfusion (I/R) injury. The current study aimed to determine the neuroprotective effect of β-PAE and the underlying mechanisms on cerebral I/R injury. Following pretreatment with β-PAE (10 mg/kg body weight) by tail intravenous injection for 1 h, Sprague-Dawley rats were subjected to middle cerebral artery occlusion for 2 h and reperfusion for 24 h. The results indicated that pretreatment with β-PAE could diminish the infarct volume, decrease the brain water content, reduce the neurological deficit score and restore the mitochondrial membrane potential, compared with the untreated I/R injury group. Furthermore, cell apoptosis was markedly suppressed by β-PAE, and this effect was associated with the decreased apoptosis regulator BAX/apoptosis regulator Bcl-2 expression ratio and caspase-3 activity. In addition, β-PAE significantly inhibited the release of proinflammatory factors, including tumor necrosis factor-α, interleukin (IL)-1β and IL-6. Superoxide generation and malondialdehyde levels were reduced while the levels of glutathione peroxidase and superoxide dismutase were elevated following treatment with β-PAE, indicating the antioxidative role of β-PAE in cerebral I/R injury. Furthermore, the Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling pathway was inhibited by β-PAE, as demonstrated by the decreased TLR4 expression and nuclear translocation of p65, and increased IκBα level. Taken together, the results suggested that β-PAE may exhibit a neuroprotective effect on cerebral I/R injury in rats through inactivating the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fu-Bo Zhang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jian-Ping Wang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hong-Xia Zhang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Gui-Mei Fan
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xin Cui
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|