1
|
Balkrishna A, Sharma S, Gohel V, Singh R, Tomer M, Dev R, Sinha S, Varshney A. Fevogrit, a polyherbal medicine, mitigates endotoxin (lipopolysaccharide)-induced fever in Wistar rats by regulating pro-inflammatory cytokine levels. Animal Model Exp Med 2025; 8:728-738. [PMID: 39021318 PMCID: PMC12008449 DOI: 10.1002/ame2.12472] [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: 05/06/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Fever is characterized by an upregulation of the thermoregulatory set-point after the body encounters any pathological challenge. It is accompanied by uncomfortable sickness behaviors and may be harmful in patients with other comorbidities. We have explored the impact of an Ayurvedic medicine, Fevogrit, in an endotoxin (lipopolysaccharide)-induced fever model in Wistar rats. METHODS Active phytoconstituents of Fevogrit were identified and quantified using ultra-high-performance liquid chromatography (UHPLC) platform. For the in-vivo study, fever was induced in male Wistar rats by the intraperitoneal administration of lipopolysaccharide (LPS), obtained from Escherichia coli. The animals were allocated to normal control, disease control, Paracetamol treated and Fevogrit treated groups. The rectal temperature of animals was recorded at different time points using a digital thermometer. At the 6-h time point, levels of TNF-α, IL-1β and IL-6 cytokines were analyzed in serum. Additionally, the mRNA expression of these cytokines was determined in hypothalamus, 24 h post-LPS administration. RESULTS UHPLC analysis of Fevogrit revealed the presence of picroside I, picroside II, vanillic acid, cinnamic acid, magnoflorine and cordifolioside A, as bioactive constituents with known anti-inflammatory properties. Fevogrit treatment efficiently reduces the LPS-induced rise in the rectal temperature of animals. The levels and gene expression of TNF-α, IL-1β and IL-6 in serum and hypothalamus, respectively, was also significantly reduced by Fevogrit treatment. CONCLUSION The findings of the study demonstrated that Fevogrit can suppress LPS-induced fever by inhibiting peripheral or central inflammatory signaling pathways and could well be a viable treatment for infection-induced increase in body temperatures.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
- Department of Allied and Applied SciencesUniversity of PatanjaliHaridwarIndia
- Patanjali UK TrustGlasgowUK
- Patanjali Yogpeeth Nepal, MandikhatarKathmanduNepal
| | - Sonam Sharma
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Vivek Gohel
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Rani Singh
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Meenu Tomer
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Rishabh Dev
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Sandeep Sinha
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
| | - Anurag Varshney
- Drug Discovery and Development DivisionPatanjali Research FoundationHaridwarIndia
- Department of Allied and Applied SciencesUniversity of PatanjaliHaridwarIndia
- Special Centre for Systems MedicineJawaharlal Nehru UniversityNew DelhiIndia
| |
Collapse
|
2
|
Vithalkar MP, Sandra KS, Bharath HB, Krishnaprasad B, Fayaz SM, Sathyanarayana B, Nayak Y. Network Pharmacology-driven therapeutic interventions for Interstitial Lung Diseases using Traditional medicines: A Narrative Review. Int Immunopharmacol 2025; 147:113979. [PMID: 39746273 DOI: 10.1016/j.intimp.2024.113979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/06/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
This review explores the progressive domain of network pharmacology and its potential to revolutionize therapeutic approaches for Interstitial Lung Diseases (ILDs), a collective term encompassing Interstitial Pneumonia, Pneumoconiosis, Connective Tissue Disease-related ILDs, and Sarcoidosis. The exploration focuses on the profound legacy of traditional medicines, particularly Ayurveda and Traditional Chinese Medicines (TCM), and their largely unexplored capacity in ILD treatment. These ancient healing systems, characterized by their holistic methodologies and multifaceted treatment modalities, offer a promising foundation for discovering innovative therapeutic strategies. Moreover, the review underscores the amalgamation of artificial intelligence (AI) and machine learning (ML) methodologies with bioinformatics, creating a computational synergy capable of deciphering the intricate biological networks associated with ILDs. Network pharmacology has tailored the hypothesis from the conventional "one target, one drug" towards a "network target, multi-component therapeutics" approach. The fusion of traditional literature and computational technology can unveil novel drugs, targets, and pathways, augmenting effective therapies and diminishing adverse effects related to current medications. In conclusion, this review provides a comprehensive exposition of how Network Pharmacology tools can leverage the insights of Ayurveda and TCM to craft efficacious therapeutic solutions for ILDs. It sets the stage for future investigations in this captivating interdisciplinary domain, validating the use of traditional medicines worldwide.
Collapse
Affiliation(s)
- Megh Pravin Vithalkar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - K S Sandra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - H B Bharath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - B Krishnaprasad
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - S M Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - B Sathyanarayana
- Muniyal Institute of Ayurveda Medical Sciences, Manipal, Karnataka 576104, India
| | - Yogendra Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
3
|
Guo Y, Zhang J, Yuan T, Yang C, Zhou Q, Shaukat A, Deng G, Wang X. Luteolin Alleviates Inflammation Induced by Staphylococcus aureus in Bovine Mammary Epithelial Cells by Attenuating NF-κB and MAPK Activation. Vet Sci 2025; 12:96. [PMID: 40005856 PMCID: PMC11861667 DOI: 10.3390/vetsci12020096] [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: 11/19/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
The internalization of S. aureus in bMECs is a major pathogenic mechanism leading to mastitis, causing significant economic losses in the dairy industry. Numerous plants contain Lut, a natural flavonoid with anti-inflammatory and antioxidant properties. However, little is known about Lut's ability to reduce inflammation caused by S. aureus in bMECs. This research aimed to evaluate the mechanism by which Lut reduces S. aureus-induced inflammation in bMECs. Through GO and KEGG enrichment analysis, researchers analyzed the differentially expressed genes in bMECs infected with S. aureus in NCBI GEO (GSE139612) and also analyzed the targets of Lut predicted by various online platforms. These studies identified two overlapping signaling pathways, the NF-κB and the MAPK pathways. We stimulated bMECs with S. aureus for two hours and then added Lut for ten hours, with a total duration of twelve hours. The expression levels of TLR2-MyD88-TRAF6 components, inflammatory cytokines, and protein phosphorylation associated with the MAPK and NF-κB signaling pathways were then assessed. Based on all of the results, Lut inhibited the generation of inflammatory cytokines in bMECs that were induced by S. aureus through the TLR2, NF-κB, and MAPK signaling pathways. This process might account for the anti-inflammatory properties of Lut.
Collapse
Affiliation(s)
- Yingfang Guo
- School of Physical Education, Wuhan Business University, Wuhan 430056, China;
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430056, China; (J.Z.); (C.Y.); (G.D.)
| | - Jinxin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430056, China; (J.Z.); (C.Y.); (G.D.)
| | - Ting Yuan
- College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing 404155, China;
| | - Cheng Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430056, China; (J.Z.); (C.Y.); (G.D.)
| | - Qingqing Zhou
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China;
| | - Aftab Shaukat
- Department of College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China;
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430056, China; (J.Z.); (C.Y.); (G.D.)
| | - Xiaoyan Wang
- College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing 404155, China;
| |
Collapse
|
4
|
Mao N, Yu Y, Lu X, Yang Y, Liu Z, Wang D. Preventive effects of matrine on LPS-induced inflammation in RAW 264.7 cells and intestinal damage in mice through the TLR4/NF-κB/MAPK pathway. Int Immunopharmacol 2024; 143:113432. [PMID: 39447411 DOI: 10.1016/j.intimp.2024.113432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Matrine is a tetracyclic quinolizidine alkaloid with diverse bioactive properties, including anti-inflammatory and neuroprotective properties. However, the underlying anti-inflammatory mechanisms remain unclear. PURPOSE This study aimed to explore how matrine reduces Lipopolysaccharide (LPS)-induced inflammation in RAW 264.7 cells and to assess its protective effects against LPS-induced intestinal damage. METHODS The effect of matrine on cell viability was assessed using the cell counting kit-8 (CCK-8) assay. Additionally, its impact on inflammatory cytokines and macrophage polarization was assessed using enzyme-linked immunosorbent assay (ELISA), flow cytometry, and quantitative real-time polymerase chain reaction (qRT-PCR) analyses. The effects on intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP), nitric oxide (NO) production, and oxidative stress were evaluated using 2',7'-dichlorodihydrofluorescein diacetate staining and JC-1 and Griess assays. Immunofluorescence staining was used to observe the translocation of the NF-κB p65 subunit. Western blotting (WB) and qRT-PCR were employed to analyze the expression levels of proteins related to the toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB)/mitogen-activated protein kinase (MAPK) pathway. An LPS-induced mouse model was established to study the intestinal inflammation and barrier injury. Mouse feces characteristics, colon length, and disease activity index (DAI) were recorded. Hematoxylin-eosin (H&E) and alcian blue/periodic acid schiff (AB/PAS) staining were used to observe morphological changes and barrier damage in the duodenum, jejunum, ileum, and colon and to measure villus length, crypt depth, goblet cell count, and positive areas in the duodenum, jejunum, and ileum. The content of short-chain fatty acids (SCFAs) in the colon was determined using gas chromatography (GC). RESULTS Matrine inhibited LPS-induced inflammatory cytokine levels, suppressed macrophage M1 polarization, and promoted M2 macrophage polarization. Matrine reduced LPS-induced increases in ROS and NO levels and regulates oxidative stress. Additionally, matrine inhibited the nuclear translocation of the NF-κB p65 subunit and exerted anti-inflammatory effects by suppressing the activation of the TLR4/NF-κB/MAPK pathway. In vivo experiments indicated that matrine significantly alleviated LPS-induced diarrhea, increased DAI, and shortened the colon. Matrine reduced the production of the pro-inflammatory cytokine interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α and the pro-inflammatory mediator NO in mouse intestinal tissues while promoting the content of the anti-inflammatory cytokine IL-10. Furthermore, it improved intestinal tissue structure and alleviated LPS-induced intestinal barrier damage. Finally, matrine increased the SCFA levels in the intestine. CONCLUSION Matrine exerted its anti-inflammatory effects and protects against intestinal injury through the TLR4/NF-κB/MAPK signaling pathway.
Collapse
Affiliation(s)
- Ningning Mao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaming Yu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xuanqi Lu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yang Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Zhenguang Liu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Deyun Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
5
|
Abd Elkarim AS, Mohamed SH, Ali NA, Elsayed GH, Aly MS, Elgamal AM, Elsayed WM, El-Newary SA. The Phytochemical Profile of the Petroleum Ether Extract of Purslane Leaves and Its Anticancer Effect on 4-(Methylnitrosamino)-1-(3-pyridyl)-1-buta-4 None (NNK)-Induced Lung Cancer in Rats. Int J Mol Sci 2024; 25:13024. [PMID: 39684736 DOI: 10.3390/ijms252313024] [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: 10/06/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/18/2024] Open
Abstract
Lung cancer is a prevalent and very aggressive sickness that will likely claim 1.8 million lives by 2022, with an estimated 2.2 million additional cases expected worldwide. The goal of the current investigation was to determine whether petroleum ether extract of purslane leaf could be used to treat lung cancer induced by 4-(Methylnitrosamino)-1-(3-pyridyl)-1-buta-4 none (NNK) in rats. In the in vitro extract recorded, promising anticancer effects in A540 cell lines with IC50 were close to the reference drug, doxorubicin (14.3 and 13.8 μg/mL, respectively). A dose of 500 mg/kg/day orally for 20 weeks exhibited recovery effects on NNK-induced lung cancer with a good safety margin, where Intercellular Adhesion Molecule-1 (ICAM-1), the lung cancer biomarker, was significantly reduced by about 18.75% compared to cancer control. Purslane exhibited many anticancer mechanisms, including (i) anti-proliferation as a significant reduction in Ki67 level (20.42%), (ii) anti-angiogenesis as evident by a considerable decrease in Matrix metalloproteinase-9 (MMP-9) expression (79%), (iii) anti-inflammation as a remarked decline in Insulin-like growth factor 1 (IGF-1) expression (62%), (iv) pro-apoptotic effect as a significant activation in Forkhead box protein O1 (FOXO1) expression (262%), and (v) anti-oxidation as remarkable activation on antioxidant biomarkers either non-enzymatic or enzymatic concurrent with considerable depletion on oxidative stress biomarker, in comparison to cancer control. The histopathological examination revealed that Purslane extract showed markedly improved tissue structure and reduced pathological changes across all examined organs caused by NNK. The anti-lung cancer effect exhibited by the extract may be linked to the active ingredients of the extract that were characterized by LC-MS, such as α-linolenic acid, linoleic acid, palmitic acid, β-sitosterol, and alkaloids (berberine and magnoflorine).
Collapse
Affiliation(s)
- Asmaa S Abd Elkarim
- Chemistry of Tanning Materials and Leather Technology Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Safaa H Mohamed
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
| | - Naglaa A Ali
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
| | - Ghada H Elsayed
- Hormones Department, National Research Centre, 33 El-Bouhoths St., Dokki, Giza 12622, Egypt
- Stem Cells Lab, Centre of Excellence for Advanced Sciences, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Mohamed S Aly
- Department of Animal Reproduction and Artificial Insemination, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Abdelbaset M Elgamal
- Department of Chemistry of Microbial and Natural Products, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Wael M Elsayed
- Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| | - Samah A El-Newary
- Medicinal and Aromatic Plants Research Department, National Research Centre, 33 El-Bohouth St., Dokki, Giza 12622, Egypt
| |
Collapse
|
6
|
Zhang M, Xu L, Zhu C, Zhang Y, Luo R, Ren J, Yu J, Zhang Y, Liang G, Zhang Y. Magnoflorine ameliorates hepatic fibrosis and hepatic stellate cell activation by regulating ferroptosis signaling pathway. Heliyon 2024; 10:e39892. [PMID: 39634391 PMCID: PMC11615489 DOI: 10.1016/j.heliyon.2024.e39892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Liver fibrosis is a chronic liver disease that brings a heavy economic burden to the world and has attracted global attention. Although the pathological mechanisms and treatment strategies of liver fibrosis have been extensively studied, there are currently no effective targeted drugs for the prevention and treatment of liver fibrosis in clinical practice. Therefore, it is imperative to seek and develop effective treatment strategies and drugs for liver fibrosis. Magnoflorine (MAG) is a natural product with multiple pharmacological activities. Thus, in this study, we will explore the effect of MAG on alleviating liver fibrosis in mice and its mechanism of action. Our study indicates that MAG can alleviate liver damage, improve liver collagen deposition, and significantly reduced the expression levels of hepatic stellate cells (HSCs) activation markers in vivo. Additionally, the findings of this study indicate that MAG can inhibit the transforming growth factor-beta (TGF-β)/Smad signaling pathway. Bioinformatics analysis suggests that the alleviating effect of MAG on liver fibrosis may be associated with ferroptosis. Interestingly, in vitro experiments have demonstrated that MAG slows down the progression of liver fibrosis by inhibiting the activation of HSCs, and further confirms that MAG promotes ferroptosis in ROS-mediated activated HSCs. In short, MAG has a good alleviating effect on liver fibrosis and will be a potential candidate drug for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Lenan Xu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Chengkai Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Yawen Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Ruixiang Luo
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Juan Ren
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jie Yu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Yanmei Zhang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| |
Collapse
|
7
|
Chen Y, Yang H, Hu X, Yang T, Zhao Y, Liu H, Fan H. Coenzyme Q10 ameliorates lipopolysaccharide-induced acute lung injury by attenuating oxidative stress and NLRP3 inflammation through regulating mitochondrial dynamics. Int Immunopharmacol 2024; 141:112941. [PMID: 39191119 DOI: 10.1016/j.intimp.2024.112941] [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: 05/21/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Increasing evidence has demonstrated that coenzyme Q10 (CoQ10) exhibits a range of biological properties. Herein, we explored the protective effect and potential molecular mechanism of CoQ10 on lipopolysaccharide (LPS)-induced acute lung injury (ALI). We found that medium (10 mg/kg) and high (50 mg/kg) doses of CoQ10 ameliorated LPS (50 µg/µL)-induced ALI to varying degrees, as demonstrated by reduced lung coefficient, lower wet/dry weight lung tissue ratio, decreased bronchoalveolar lavage fluid protein concentration, less anatomical and histopathological damage to the lung, and increased expression of proteins related to lung epithelial barrier structure. CoQ10 also alleviated LPS-induced oxidative stress and inflammation mediated by NOD-like receptor protein 3 (NLRP3) by reducing the reactive oxygen species (ROS), malondialdehyde, and mitochondrial ROS concentrations, increasing superoxide dismutase, glutathione, and catalase activity, and decreasing NLRP3 expression at the protein and mRNA levels. Moreover, CoQ10 alleviated structural and functional damage to the mitochondria, inhibited mitochondrial fission, and promoted mitochondrial fusion, mainly by inhibiting phosphorylation of dynamin-related protein 1 (Drp1) at Ser616 and Ser637. Correlation analysis revealed that mitochondrial fission (especially Drp1) was positively correlated with oxidative stress, NLRP3-mediated inflammation, and structural damage to the lung epithelial barrier. Molecular docking analysis showed that CoQ10 binds stably to Drp1, with a binding energy of -5.9 kcal/mol. Furthermore, the use of schaftoside (a Drp1 inhibitor) has further elucidated the mechanism of action of CoQ10. Together, these results suggest that CoQ10 alleviates LPS-induced ALI by regulating mitochondrial dynamics, attenuating oxidative stress, and decreasing NLRP3-medated inflammation, thereby promoting lung epithelial barrier structural remodeling.
Collapse
Affiliation(s)
- Yongping Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; Branch of Animal Husbandry and Veterinary of Heilong Jiang Academy of Agricultural Sciences, Qiqihar 161005, China
| | - Xueyuan Hu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Huanqi Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China.
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Balkrishna A, Tiwari A, Maity M, Tomer M, Varshney Y, Dev R, Sinha S, Varshney A. Co-administration of Ayurvedic medicines Arshogrit and Jatyadi Ghrit, attenuate croton oil-induced hemorrhoids in rat model of recto-anal inflammation by modulating TNF-α and IL-1β levels. Drug Dev Ind Pharm 2024; 50:938-951. [PMID: 39565131 DOI: 10.1080/03639045.2024.2432595] [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: 03/16/2024] [Revised: 10/25/2024] [Accepted: 11/17/2024] [Indexed: 11/21/2024]
Abstract
OBJECTIVE To study the efficacy of co-administration of Arshogrit (AG) and Jatyadi Ghrit (JG), two herb-based Ayurvedic medicines, in rat model of croton oil-induced hemorrhoids. SIGNIFICANCE Hemorrhoids refer to a pathological condition affecting the recto-anal region causing pain, swelling, bleeding and protrusion. The available therapies for hemorrhoids are symptomatic or invasive but are expensive and associated with adverse effects. Hence, there exists a need for efficacious and safer pharmacotherapies. METHODS Ultra high performance liquid chromatography detected nine phytocompounds in AG and seven in JG. Seven fatty acids were additionally identified in JG by Gas Chromatography-Mass Spectrometry analysis. The in-vivo efficacy of the co-administration of AG, which was administered orally at the doses of 20, 60 and 200 mg/kg/day and JG, which was topically applied (100 mg/animal/day) was evaluated in Wistar rats by inducing hemorrhoids development with the application of croton oil preparation (COP) in the recto-anal area. Prednisolone was employed as the standard drug and was administered orally at the dose of 1 mg/kg/day. RESULTS AG and JG were able to attenuate the croton oil-induced macro and microscopic anomalies. Gross pathological observation demonstrated remarkable decrease in croton oil-induced swelling, hemorrhage and formation of pseudomembrane, with the escalating doses of AG. Microscopic observation revealed alleviation in the histopathological lesions (necrosis, inflammation, hemorrhage/congestion, degeneration and dilatation of blood vessels). AG and JG additionally reduced COP-induced increase in the serum levels of pro-inflammatory cytokines. CONCLUSION This study convincingly demonstrates that co-administration of AG and JG is a potential therapy against hemorrhoids, warranting further investigations.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, India
- Patanjali UK Trust, Glasgow, United Kingdom
- Vedic Acharya Samaj Foundation, Inc., Groveland, FL, USA
| | - Aakanksha Tiwari
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Madhulina Maity
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Meenu Tomer
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Yash Varshney
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Rishabh Dev
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Sandeep Sinha
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Foundation, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Haridwar, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
9
|
Zhu M, Hu J, Pan Y, Jiang Q, Shu C. Magnoflorine attenuates Ang II-induced cardiac remodeling via promoting AMPK-regulated autophagy. Cardiovasc Diagn Ther 2024; 14:576-588. [PMID: 39263476 PMCID: PMC11384461 DOI: 10.21037/cdt-24-130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/27/2024] [Indexed: 09/13/2024]
Abstract
Background Heart failure (HF) remains one of the most common events in the progression of hypertension. Magnoflorine (MNF) has been shown beneficial effects on the cardiovascular system. However, the action of MNF on angiotensin (Ang) II-induced cardiac remodeling and its underlying mechanisms have not yet been characterised. Here, we assessed the action of MNF in the development of hypertension-related HF. Methods C57BL/6 male mice were subjected to Ang II through a micro-osmotic pump infusion continuously for 4 weeks to induce hypertensive HF. MNF (10 and 20 mg/kg) was administered in the final 2 weeks. Ang II content was measured by enzyme-linked immunosorbent assay (ELISA) kit. Values of ejection fraction (EF) and fractional shortening (FS) were detected using an ultrasound diagnostic instrument. The mRNA levels of hypertrophic and fibrotic genes were determined by real-time quantitative polymerase chain reaction (RT-qPCR). Haematoxylin and eosin (H&E), wheat germ agglutinin (WGA), Masson trichrome, and Sirius Red staining were used to analyse pathologic changes in heart tissues. The expression levels of phosphorylated AMP-activated protein kinase (AMPK), light chain 3 microtubule associated protein II (LC3 II) to LC3 I, and p62 were detected by western blot assay. Results MNF significantly improved cardiac dysfunction and the content of creatine kinase-MB without altering blood pressure in Ang II-challenged mice. MNF obviously corrected the phenotypes of cardiac hypertrophy and fibrosis, including the high mRNA levels of atrial natriuretic peptide (Anp), brain natriuretic peptide (Bnp), collagen1a (Col1a1), transforming growth factor beta (Tgfb1), enlarged myocardial areas, and increased positive areas of Masson trichrome and Sirius Red staining. In addition, MNF alleviated oxidative injury, reflected by the upregulation of glutathione and the downregulation of reactive oxygen species and malondialdehyde. The activation of AMPK was elevated accompanied by an increased level of autophagy by MNF in hypertensive heart tissues. The therapeutic action of MNF was confirmed in Ang II-challenged H9c2 cells. Specifically, the AMPK inhibitor could eliminate the autophagy pathway in which MNF is involved. Conclusions MNF has benefits in hypertension-induced cardiac remodeling, which was partially associated with the improvement of oxidative stress via the mediation of the AMPK/autophagy axis.
Collapse
Affiliation(s)
- Meili Zhu
- Department of Rehabilitation Medicine, the First People's Hospital of Yongkang, Yongkang, China
| | - Jiangbiao Hu
- Department of Rehabilitation Medicine, the First People's Hospital of Yongkang, Yongkang, China
| | - Yifan Pan
- Department of Cardiology, the First People's Hospital of Yongkang, Yongkang, China
| | - Qian Jiang
- Department of Rehabilitation Medicine, the First People's Hospital of Yongkang, Yongkang, China
| | - Chang Shu
- Department of Medical, the First People's Hospital of Yongkang, Yongkang, China
| |
Collapse
|
10
|
Guo X, Yang J, Wang W, Gong Y, Zhang P, Wu M, Zheng Y, Wang C. Pharmacokinetic and tissue distribution analysis of bioactive compounds from Fuke Qianjin capsules in rats by a validated UPLCMS/MS method. J Pharm Biomed Anal 2024; 243:116069. [PMID: 38460275 DOI: 10.1016/j.jpba.2024.116069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/11/2024]
Abstract
Fuke Qianjin capsules (FKQJ) exhibit obvious advantages and characteristics in the treatment of pelvic inflammatory disease. At present, information regarding the in vivo process of FKQJ is lacking, which has become a bottleneck in further determining the therapeutic effect of this traditional Chinese medicine. In the present study, a sensitive, simple and reliable method was developed and validated for the simultaneous quantification of 12 main components (4 flavonoids, 4 alkaloids, 2 phthalides and 2 diterpene lactones) in plasma and seven tissues of rats to study the pharmacokinetic and distribution characteristics of these components in vivo by using ultra performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) for the first time. Plasma and tissue were prepared by protein precipitation with acetonitrile and methanol, followed by its separation on a Waters Acquity UPLC BEH C18 column. The quantification was performed via multiple reaction monitoring (MRM) by a triple quadrupole mass spectrometer under positive electrospray ionization (ESI) mode. The method was validated to demonstrate its selectivity, linearity, accuracy, precision, recovery, matrix effect and stability. For 12 analytes, the low limit of quantification (LLOQs) reached 0.005-2.44 ng/mL, and all calibration curves showed good linearity (r2 ≥ 0.990) in linear ranges. The intra-day and inter-day precision (relative standard deviation) for all analytes was less than 14.96%, and the accuracies were in the range of 85.29%-114.97%. Extraction recoveries and matrix effects of analytes were acceptable. The pharmacokinetic results showed that the main components could be absorbed quickly, had a short residence time, and were eliminated quickly in vivo. At different time points, the 12 components were widely distributed with uneven characteristics in the body, which tended to be distributed in the liver, kidney and lung and to a lesser extent in the uterus, brain and heart. The pharmacokinetic process and tissue distribution characteristics of FKQJ were expounded in this study, which can provide a scientific theory for in-depth development of FKQJ and guide FKQJ use in the clinic.
Collapse
Affiliation(s)
- Xiujie Guo
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Jiaying Yang
- Taizhou Medical City Guokehuawu Bio-Pharm Co., Ltd, Taizhou 225300, China
| | - Wei Wang
- Taizhou Medical City Guokehuawu Bio-Pharm Co., Ltd, Taizhou 225300, China
| | - Yun Gong
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412000, China
| | - Peng Zhang
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412000, China.
| | - Mengyao Wu
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412000, China
| | - Yuanqing Zheng
- Zhuzhou Qianjin Pharmaceutical Co., Ltd., Zhuzhou 412000, China
| | - Chaoran Wang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
11
|
Cheng Y, Lu Z, Mao T, Song Y, Qu Y, Chen X, Chen K, Liu K, Zhang C. Magnoflorine Ameliorates Chronic Kidney Disease in High-Fat and High-Fructose-Fed Mice by Promoting Parkin/PINK1-Dependent Mitophagy to Inhibit NLRP3/Caspase-1-Mediated Pyroptosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12775-12787. [PMID: 38776285 DOI: 10.1021/acs.jafc.3c09634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2024]
Abstract
Excessive intake of fat and fructose in Western diets has been confirmed to induce renal lipotoxicity, thereby driving the progression of chronic kidney disease (CKD). This study was conducted to evaluate the efficacy of magnoflorine in a CKD mouse model subjected to high-fat and high-fructose diets. Our results demonstrated that magnoflorine treatment ameliorated abnormal renal function indices (serum creatinine, urea nitrogen, uric acid, and urine protein) in high-fat- and high-fructose-fed mice. Histologically, renal tubular cell steatosis, lipid deposition, tubular dilatation, and glomerular fibrosis were significantly reduced by the magnoflorine treatment in these mice. Mechanistically, magnoflorine promotes Parkin/PINK1-mediated mitophagy, thereby inhibiting NLRP3/Caspase-1-mediated pyroptosis. Consistent findings were observed in the palmitic acid-incubated HK-2 cell model. Notably, both silencing of Parkin and the use of a mitophagy inhibitor reversed the inhibitory effect of magnoflorine on NLRP3 inflammasome activation in vitro. Therefore, the present study provides compelling evidence that magnoflorine improves renal injury in high-fat- and high-fructose-fed mice by promoting Parkin/PINK1-dependent mitophagy to inhibit NLRP3 inflammasome activation and pyroptosis. Our findings suggest that dietary supplementation with magnoflorine and magnoflorine-rich foods (such as magnolia) might be an effective strategy for the prevention of CKD.
Collapse
Affiliation(s)
- Ye Cheng
- Pharmaceutical Department, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, China
| | - Zhengjie Lu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Tongyun Mao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yingying Song
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yaqin Qu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xin Chen
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Kaiqi Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kexin Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| |
Collapse
|
12
|
Och A, Lemieszek MK, Cieśla M, Jedrejek D, Kozłowska A, Pawelec S, Nowak R. Berberis vulgaris L. Root Extract as a Multi-Target Chemopreventive Agent against Colon Cancer Causing Apoptosis in Human Colon Adenocarcinoma Cell Lines. Int J Mol Sci 2024; 25:4786. [PMID: 38732003 PMCID: PMC11084310 DOI: 10.3390/ijms25094786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Berberis vulgaris L. (Berberidaceae) is a shrub that has been widely used in European folk medicine as an anti-inflammatory and antimicrobial agent. The purpose of our study was to elucidate the mechanisms of the chemopreventive action of the plant's methanolic root extract (BVR) against colon cancer cells. Studies were conducted in human colon adenocarcinoma cell lines (LS180 and HT-29) and control colon epithelial CCD841 CoN cells. According to the MTT assay, after 48 h of cell exposure, the IC50 values were as follows: 4.3, 46.1, and 50.2 µg/mL for the LS180, HT-29, and CCD841 CoN cells, respectively, showing the greater sensitivity of the cancer cells to BVR. The Cell Death Detection ELISAPLUS kit demonstrated that BVR induced programmed cell death only against HT-29 cells. Nuclear double staining revealed the great proapoptotic BVR properties in HT-29 cells and subtle effect in LS180 cells. RT-qPCR with the relative quantification method showed significant changes in the expression of genes related to apoptosis in both the LS180 and HT-29 cells. The genes BCL2L1 (126.86-421.43%), BCL2L2 (240-286.02%), CASP3 (177.19-247.83%), and CASP9 (157.99-243.75%) had a significantly elevated expression, while BCL2 (25-52.03%) had a reduced expression compared to the untreated control. Furthermore, in a panel of antioxidant tests, BVR showed positive effects (63.93 ± 0.01, 122.92 ± 0.01, and 220.29 ± 0.02 mg Trolox equivalents (TE)/g in the DPPH•, ABTS•+, and ORAC assays, respectively). In the lipoxygenase (LOX) inhibition test, BVR revealed 62.60 ± 0.87% of enzyme inhibition. The chemical composition of BVR was determined using a UHPLC-UV-CAD-MS/MS analysis and confirmed the presence of several known alkaloids, including berberine, as well as other alkaloids and two derivatives of hydroxycinnamic acid (ferulic and sinapic acid hexosides). The results are very promising and encourage the use of BVR as a comprehensive chemopreventive agent (anti-inflammatory, antioxidant, and pro-apoptotic) in colorectal cancer, and were widely discussed alongside data from the literature.
Collapse
Affiliation(s)
- Anna Och
- Department of Pharmaceutical Botany, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland;
| | | | - Marek Cieśla
- College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland;
| | - Dariusz Jedrejek
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation—State Rsearch Institute, Czartoryskich 8 Street, 24-100 Puławy, Poland; (D.J.); (S.P.)
| | - Aleksandra Kozłowska
- Department of Radiotherapy, Medical University of Lublin, 13 Radziwiłłowska St., 20-080 Lublin, Poland;
| | - Sylwia Pawelec
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation—State Rsearch Institute, Czartoryskich 8 Street, 24-100 Puławy, Poland; (D.J.); (S.P.)
| | - Renata Nowak
- Department of Pharmaceutical Botany, Medical University of Lublin, 1 Chodźki St., 20-093 Lublin, Poland;
| |
Collapse
|
13
|
Zeng Z, Fu Y, Li M, Shi Y, Ding Q, Chen S. Guben Qingfei decoction attenuates LPS-induced acute lung injury by modulating the TLR4/NF-κB and Keap1/Nrf2 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117674. [PMID: 38154525 DOI: 10.1016/j.jep.2023.117674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acute lung injury (ALI) is a life-threatening and widespread disease, with exceptionally high morbidity and mortality rates. Unfortunately, effective drugs for ALI treatment are currently lacking. Guben Qingfei decoction (GBQF) is a Chinese herbal compound known for its efficacy in treating viral pneumonia, yet the precise underlying mechanisms remain unknown. AIM OF THE STUDY This study aimed to validate the mitigating effect of GBQF on ALI and to further investigate its mechanism. MATERIALS AND METHODS An ALI mice model was established by infusing LPS into the endotracheal tube. The effects of GBQF on ALI were investigated by measuring lung W/D; MPO; BALF total protein concentration; total number of cells; TNF-α, IL-1β, and IL-6 levels; pathological changes in lung tissue, and oxidation products. Immunohistochemistry and Western Blotting were performed to verify the underlying mechanisms. MH-S and BEAS-2B cells were induced by LPS, and the effects of GBQF were confirmed by RT-PCR and immunofluorescence. RESULTS GBQF significantly reduced LPS-induced ALI in mice, improved lung inflammation, reduced the production of oxidative products, increased the activity of antioxidant enzymes, and reduced the degree of lung tissue damage. GBQF prevents MH-S cells from releasing inflammatory factors and reduces oxidative damage to BEAS-2B cells. In vivo studies have delved deeper into the mechanism of action of GBQF, revealing its correlation with the TLR4/NF-κB and Keap1/Nrf2 pathways. CONCLUSIONS Our study demonstrates that GBQF is an effective treatment for ALI, providing a new perspective on medication development for ALI treatment.
Collapse
Affiliation(s)
- Ziyuan Zeng
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China
| | - Yuchen Fu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Minfang Li
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China; Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Qi Ding
- Shenzhen Research Institute, Beijing University of Chinese Medicine, Shenzhen, China.
| | - Sheng Chen
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China.
| |
Collapse
|
14
|
Joshi P, Patel S, Paliwal A, Jain S, Verma K, Dwivedi J, Sharma S. Tinospora cordifolia ameliorates paclitaxel-induced neuropathic pain in albino rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117559. [PMID: 38072294 DOI: 10.1016/j.jep.2023.117559] [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: 09/07/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tinospora cordifolia (T. cordifolia) (Willd.) Miers, a member of the Menispermaceae, family documented in the ancient textbooks of the Ayurveda System of Medicine, has been used in the management of sciatica pain and diabetic neuropathy. AIM The study has been designed to evaluate the antinociceptive potential of various extracts of T. cordifolia stem in Paclitaxel (PT)-generated neuropathic pain model in albino rats and explore its possible mechanism employing molecular docking studies. METHODS Stems of T. cordifolia were shade dried, grinded in fine powder, and extracted separately with different solvents viz. ethanol, water & hydro-alcoholic and characterized using LCMS/MS. The antinociceptive property of T. cordifolia stem (200 and 400 mg/kg) was examined in albino rats using a PT-induced neuropathic pain model. Further, the effect of these extracts was also observed using different behavioral assays viz. cold allodynia, mechanical hyperalgesia (pin-prick test), locomotor activity test, walking track test, and Sciatic Functional Index (SFI) in rats. Tissue lysate of the sciatic nerve was used to determine various biochemical markers such as GSH, SOD, TBARS, tissue protein, and nitrite. Further to explore the possible mechanism of action, the most abundant and therapeutically active compounds available in aqueous extract were analyzed for binding affinity towards soluble epoxide hydrolase (sEH) enzyme (PDB ID: 3wk4) employing molecular docking studies. RESULTS The results of the LCMS/MS study of different extracts of T. cordifolia indicated presence of alkaloids, glycosides, terpenoids, sterols and sugars such as amritoside A, tinocordin, magnoflorine, N-methylcoclaurine, coridine, 20β-hydroxyecdysone and menaquinone-7 palmatin, cordifolioside A and tinosporine etc. Among all the three extracts, the hydroalcoholic extract (400 mg/kg) showed the highest response followed by aqueous and ethanolic extracts as evident in in vivo behavioral and biochemical evaluations. Furthermore, docking studies also exposed that these compounds viz. N-methylcoclaurine tinosporin, palmatine, tinocordin, 20β-hydroxyecdysone, and coridine exhibited well to excellent affinity towards target sEH protein. CONCLUSION T. cordifolia stem could alleviate neuropathic pain via soluble epoxide hydrolase inhibitory activity.
Collapse
Affiliation(s)
- Priyanka Joshi
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India; R&D, Patanjali Ayurved Ltd, Patanjali Food and Herbal Park, Haridwar, Uttarakhand, India
| | - Saraswati Patel
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Ajita Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, Rajasthan, India.
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India.
| |
Collapse
|
15
|
Dong J, Ji B, Jiang Y, Liu K, Guo L, Cui L, Wang H, Li B, Li J. Autophagy activation alleviates the LPS-induced inflammatory response in endometrial epithelial cells in dairy cows. Am J Reprod Immunol 2024; 91:e13820. [PMID: 38332507 DOI: 10.1111/aji.13820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024] Open
Abstract
PROBLEM Endometritis is a common disease that affects dairy cow reproduction. Autophagy plays a vital role in cellular homeostasis and modulates inflammation by regulating interactions with innate immune signaling pathways. However, little is known about the regulatory relationship between autophagy and inflammation in bovine endometrial epithelial cells (BEECs). Thus, we aimed to determine the role of autophagy in the inflammatory response in BEECs. METHODS OF STUDY In the present study, the expression levels of proinflammatory cytokines were measured by quantitative real-time polymerase chain reaction. Changes in the nuclear factor-κB (NF-κB) pathway and autophagy were determined using immunoblotting and immunocytochemistry. The induction of autophagosome formation was visualized by transmission electron microscopy. RESULTS Our results demonstrated that autophagy activation was inhibited in LPS-treated BEECs, while activation of the NF-κB pathway and the mRNA expression of IL-6, IL-8, and TNF-α were increased. Furthermore, blocking autophagy with the inhibitor chloroquine increased NF-κB signaling pathway activation and proinflammatory factor expression in LPS-treated BEECs. Conversely, activation of autophagy with the agonist rapamycin inhibited the NF-κB signaling pathway and downregulated proinflammatory factors. CONCLUSIONS These data indicated that LPS-induced inflammation was related to the inhibition of autophagy in BEECs. Thus, the activation of autophagy may represent a novel therapeutic strategy for eliminating inflammation in BEECs.
Collapse
Affiliation(s)
- Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Bowen Ji
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | | | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Bichun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| |
Collapse
|
16
|
Wang L, Li P, Zhou Y, Gu R, Lu G, Zhang C. Magnoflorine Ameliorates Collagen-Induced Arthritis by Suppressing the Inflammation Response via the NF-κB/MAPK Signaling Pathways. J Inflamm Res 2023; 16:2271-2296. [PMID: 37265745 PMCID: PMC10231344 DOI: 10.2147/jir.s406298] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023] Open
Abstract
Objective Magnoflorine (Mag) has been reported to have anxiolytics, anti-cancer, and anti-inflammatory properties. In this study, we aim to investigate the effects of Mag on the rheumatoid arthritis (RA) and explore the underlying mechanism using a collagen-induced arthritis (CIA) mouse model and a lipopolysaccharide (LPS)-stimulated macrophage inflammation model. Methods The in vivo effects of Mag on CIA were studied by inducing CIA in a mouse model using DBA/1J mice followed by treatment with vehicle, methotrexate (MTX, 1 mg/kg/d), and Mag (5 mg/kg/d, 10 mg/kg/d, and 20 mg/kg/d), and the in vitro effects of Mag on macrophages were examined by stimulation of RAW264.7 cells line and peritoneal macrophages (PMs) by LPS in the presence of different concentrations of Mag. Network pharmacology and molecular docking was then performed to predict the the binding ability between Mag and its targets. Inflammatory mediators were assayed by quantitative real-time PCR and enzyme linked immunosorbent assay (ELISA). Signaling pathway changes were subsequently determined by Western blotting and immunohistochemistry (IHC). Results In vivo experiments demonstrated that Mag decreased arthritis severity scores, joints destruction, and macrophages infiltration into the synovial tissues of the CIA mice. Network pharmacology analysis revealed that Mag interacted with TNF-α, IL-6, IL-1β, and MCP-1. Consistent with this, analysis of the serum, synovial tissue of the CIA mice, and the supernatant of the cultured RAW264.7 cells and PMs showed that Mag suppressed the expression of TNF-α, IL-6, IL-1β, MCP-1, iNOS, and IFN-β. Furthermore, Mag attenuated the phosphorylation of p65, IκBα, ERK, JNK, and p38 MAPKs in the synovial tissues of the CIA mice and LPS-stimulated RAW 264.7 cells. Conclusion Mag may exert anti-arthritic and anti-inflammatory effects by inhibiting the activation of NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Lei Wang
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Pengfei Li
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Yu Zhou
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Renjun Gu
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Ge Lu
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Chunbing Zhang
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
- Department of Clinical Laboratory, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
17
|
Li D, Yang L, Wang W, Song C, Xiong R, Pan S, Li N, Geng Q. Eriocitrin attenuates sepsis-induced acute lung injury in mice by regulating MKP1/MAPK pathway mediated-glycolysis. Int Immunopharmacol 2023; 118:110021. [PMID: 36966548 DOI: 10.1016/j.intimp.2023.110021] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/18/2023] [Accepted: 03/08/2023] [Indexed: 03/28/2023]
Abstract
Metabolic reprogramming has been shown to aggravate sepsis-induced acute lung injury. In particular, enhanced glycolysis is closely associated with inflammation and oxidative stress. Eriocitrin (ERI) is a natural flavonoid found in citrus fruit that exhibits various pharmacological activities, with antioxidant, anti-inflammatory, anti-diabetic, and anti-tumor properties. However, the role of ERI in lung injury is not well understood. We established a septic mouse model of acute lung injury (ALI) using lipopolysaccharide (LPS) for induction. Primary peritoneal macrophages were isolated to verify the relevant molecular mechanism. Tissues were assessed for lung pathology, pro-inflammatory cytokines, markers of oxidative stress, and protein and mRNA expression levels. In vivo experiments showed that ERI effectively alleviated LPS-induced pathological injury, suppress the inflammatory response (TNF-α, IL-1β, IL-6 levels) and decreased oxidative stress (MDA, ROS) in murine lung tissue. In vitro, ERI increased the resistance of LPS-treated cells to excessive inflammation and oxidative stress by inhibiting the enhancement of glycolysis (indicated by expression levels of HIF-1α, HK2, LDHA, PFKFB3, and PKM2). Specifically, the beneficial effects of ERI following LPS-induced lung injury occurred through promoting the expression of MKP1, which mediates the inactivation of the MAPK pathway to inhibit enhanced glycolysis. These results demonstrate that ERI has a protective effect on sepsis-induced ALI by regulating MKP1/MAPK pathway mediated-glycolysis. Hence, ERI is a promising candidate against ALI via inhibiting glycolysis.
Collapse
Affiliation(s)
- Donghang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liu Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shize Pan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
18
|
El-Sayed EK, Ibrahim RR, Ahmed AA, Khattab MA, Chen LY, Lai KH, Shaarawy FSE, Tawfik NF, Moharram FA. Quercus coccinea Münchh leaves polyphenols: Appraisal acute lung injury induced by lipopolysaccharide in mice. Biomed Pharmacother 2023:114765. [PMID: 37246132 DOI: 10.1016/j.biopha.2023.114765] [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: 04/04/2023] [Accepted: 04/21/2023] [Indexed: 05/30/2023] Open
Abstract
Genus Quercus is a well-known source for its polyphenolic content and important biological activity. Plants belonging to the Quercus genus were traditionally used in asthma, inflammatory diseases, wound healing, acute diarrhea, and hemorrhoid. Our work intended to study the polyphenolic profile of the Q. coccinea (QC) leaves and to assess the protective activity of its 80% aqueous methanol extract (AME) against lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Together, the potential molecular mechanism was investigated. Nineteen polyphenolic compounds (1-18), including tannins, flavone, and flavonol glycosides. Phenolic acids and aglycones were purified and identified from the AME of QC leaves. Treatment with AME of QC showed an anti-inflammatory effect evidenced by a remarkable decline in the count of white blood cells and neutrophils which was in harmony with decreasing the levels of high mobility group box-1, nuclear factor kappa B, tumor necrosis factor-α, and interleukin 1 beta. In addition, the antioxidant activity of QC was documented through the significant reduction in malondialdehyde level and elevation of reduced glutathione level and superoxide dismutase activity. Furthermore, the mechanism involved in the pulmonary protective effect of QC involved the downregulation of the TLR4/MyD88 pathway. The AME of QC showed a protective effect against LPS-induced ALI through the powerful anti-inflammatory and antioxidant activities which are linked to its abundancy with polyphenols.
Collapse
Affiliation(s)
- Elsayed K El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Reham R Ibrahim
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Asmaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt
| | - Mohamed A Khattab
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lo-Yun Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Fatheya S El Shaarawy
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Nashwa F Tawfik
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Fatma A Moharram
- Pharmacognosy Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
19
|
Shen Y, Fan X, Qu Y, Tang M, Huang Y, Peng Y, Fu Q. Magnoflorine attenuates inflammatory responses in RA by regulating the PI3K/Akt/NF-κB and Keap1-Nrf2/HO-1 signalling pathways in vivo and in vitro. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154339. [PMID: 35870375 DOI: 10.1016/j.phymed.2022.154339] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/02/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND As a prolonged autoimmune disorder, rheumatoid arthritis (RA) is characterised by synovial hyperplasia and the erosion of bone and cartilage. Magnoflorine (MAG) is the main component purified from Clematis manshurica Rupr. Recent studies have shown that MAG has anti-inflammatory, antioxidant, and immunosuppressive effects, which are relevant to anti-RA activities. OBJECTIVE The current investigation was conducted to explore the anti-RA effects of MAG and to discover the possible molecular mechanisms. METHODS In vitro experiments, CCK-8, wound healing, and transwell assays were utilized to evaluate the anti-proliferative, anti-migratory, and anti-invasive activities of MAG, respectively. The rate of cell distribution and cell apoptosis were evaluated by flow cytometry. ROS generation was detected by DCFH-DA staining. Western blotting, quantitative real-time polymerase chain reaction assay, and immunofluorescent staining were employed to test the anti-RA effect of MAG as well as to explore the potential mechanisms by evaluating related gene and protein expression. For in vivo experiments, an adjuvant-induced arthritis (AIA) rat model was established. The related parameters were measured in rats. Then, rats were sacrificed, and ankle joints were collected for histopathological analysis and observation. RESULTS MAG significantly decreased the proliferation, migration, invasion, and reactive oxygen species levels in IL-1β-treated MH7A cells. Furthermore, MAG promoted cell apoptosis by increasing Bax levels and decreasing Bcl-2 levels. MAG also induced cell cycle arrest. Inflammatory cytokines (iNOS, COX-2, IL-6, and IL-8) and MMPs (MMP-1, 2, 3, 9, and 13) were reduced by MAG treatment. Molecular analysis revealed that MAG exerted anti-RA effects by partly inhibiting the PI3K/Akt/NF-κB signalling axis and activating the Keap1-Nrf2/HO-1 signalling pathway. In vivo studies have revealed that MAG treatment substantially improved severe symptoms in AIA rats, and these curative effects were linked to the attenuation of inflammatory responses. CONCLUSION These results first suggested that MAG exhibits anti-arthritic effects in IL-1β-treated MH7A cells and AIA rat models. Thus, MAG may be used as a new drug to treat RA clinically.
Collapse
Affiliation(s)
- Yue Shen
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Xinting Fan
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Yuhan Qu
- School of Food and Biological engineering, Chengdu university, Chengdu 610106, China
| | - Min Tang
- School of Food and Biological engineering, Chengdu university, Chengdu 610106, China
| | - Yuehui Huang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Yi Peng
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China
| | - Qiang Fu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu university, Chengdu 610106, China.
| |
Collapse
|
20
|
Wu YC, Hsu SP, Hu MC, Lan YT, Yeh ETH, Yang FM. PEP-sNASP Peptide Alleviates LPS-Induced Acute Lung Injury Through the TLR4/TRAF6 Axis. Front Med (Lausanne) 2022; 9:832713. [PMID: 35386914 PMCID: PMC8977741 DOI: 10.3389/fmed.2022.832713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/22/2022] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) is a severe inflammatory lung disease associated with macrophages. Somatic nuclear autoantigenic sperm protein (sNASP) is a negative regulator of Toll-like receptor (TLR) signaling that targets tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) in macrophages, which is required to maintain homeostasis of the innate immune response. In the present study, we generated a cell permeable PEP-sNASP peptide using the sNASP protein N-terminal domain, and examined its potential therapeutic effect in a mouse model of ALI induced by the intranasal administration of lipopolysaccharide (LPS) and elucidated the underlying molecular mechanisms in RAW 264.7 cells. In vivo, PEP-sNASP peptide treatment markedly ameliorated pathological injury, reduced the wet/dry (W/D) weight ratio of the lungs and the production of proinflammatory cytokines (interleukin (IL)-1β, IL-6, and TNF-α). In vitro, we demonstrated that when the PEP-sNASP peptide was transduced into RAW 264.7 cells, it bound to TRAF6, which markedly decreased LPS-induced proinflammatory cytokines by inhibiting TRAF6 autoubiquitination, nuclear factor (NF)-κB activation, reactive oxygen species (ROS) and cellular nitric oxide (NO) levels. Furthermore, the PEP-sNASP peptide also inhibited NLR family pyrin domain containing 3 (NLRP3) inflammasome activation. Our results therefore suggest that the PEP-sNASP may provide a potential protein therapy against oxidative stress and pulmonary inflammation via selective TRAF6 signaling.
Collapse
Affiliation(s)
- Yu-Chih Wu
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Chun Hu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Ting Lan
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Edward T H Yeh
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AK, United States.,Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AK, United States
| | - Feng-Ming Yang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
21
|
Okumo T, Takayama Y, Maruyama K, Kato M, Sunagawa M. Senso-Immunologic Prospects for Complex Regional Pain Syndrome Treatment. Front Immunol 2022; 12:786511. [PMID: 35069559 PMCID: PMC8767061 DOI: 10.3389/fimmu.2021.786511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Complex regional pain syndrome (CRPS) is a chronic pain syndrome that occurs in tissue injuries as the result of surgery, trauma, or ischemia. The clinical features of this severely painful condition include redness and swelling of the affected skin. Intriguingly, it was recently suggested that transient receptor potential ankyrin 1 (TRPA1) is involved in chronic post-ischemia pain, a CRPS model. TRPA1 is a non-selective cation channel expressed in calcitonin gene-related peptide (CGRP)-positive primary nociceptors that becomes highly activated in ischemic conditions, leading to the generation of pain. In this review, we summarize the history of TRPA1 and its involvement in pain sensation, inflammation, and CRPS. Furthermore, bone atrophy is also thought to be a characteristic clinical sign of CRPS. The altered bone microstructure of CRPS patients is thought to be caused by aggravated bone resorption via enhanced osteoclast differentiation and activation. Although TRPA1 could be a target for pain treatment in CRPS patients, we also discuss the paradoxical situation in this review. Nociceptor activation decreases the risk of bone destruction via CGRP secretion from free nerve endings. Thus, TRPA1 inhibition could cause severe bone atrophy. However, the suitable therapeutic strategy is controversial because the pathologic mechanisms of bone atrophy in CRPS are unclear. Therefore, we propose focusing on the remission of abnormal bone turnover observed in CRPS using a recently developed concept: senso-immunology.
Collapse
Affiliation(s)
- Takayuki Okumo
- Department of Physiology, Showa University School of Medicine, Shinagawa, Japan
| | - Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, Shinagawa, Japan
| | - Kenta Maruyama
- Department of Physiology, Showa University School of Medicine, Shinagawa, Japan.,Division of Cell Signaling, National Institute for Physiological Sciences, Natural Institutes for Natural Sciences, Okazaki, Japan
| | - Mami Kato
- Department of Physiology, Showa University School of Medicine, Shinagawa, Japan.,Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Sunagawa
- Department of Physiology, Showa University School of Medicine, Shinagawa, Japan
| |
Collapse
|
22
|
Zhong R, Xia T, Wang Y, Ding Z, Li W, Chen Y, Peng M, Li C, Zhang H, Shu Z. Physalin B ameliorates inflammatory responses in lipopolysaccharide-induced acute lung injury mice by inhibiting NF-κB and NLRP3 via the activation of the PI3K/Akt pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114777. [PMID: 34737012 DOI: 10.1016/j.jep.2021.114777] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/12/2021] [Accepted: 10/22/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Physalin B (PB) is an active constituent of Physalis alkekengi L. var. Franchetii, which is a traditional medicine for clearing heat and detoxification, resolving phlegm, and diuresis. It has been commonly applied to treat sore throat, phlegm-heat, cough, dysuria, pemphigus, and eczema. AIM OF STUDY Physalin B has shown efficacy as an anti-acute lung injury (ALI) agent previously; however, its mechanisms of action remain unclear. In the present study, we established a lipopolysaccharide-induced septic ALI model using BALB/c mice to further confirm the therapeutic potential of PB and to assess the underlying molecular mechanisms. MATERIALS AND METHODS We used 75% ethanol and macroporous resin for extraction, separation, and enrichment of PB. The LPS-induced ALI mouse model was used to determine anti-inflammatory effects of PB. The severity of acute lung injury was evaluated by hematoxylin and eosin staining, wet/dry lung ratio, and myeloperoxidase (MPO) activity in lung tissue. An automatic analyzer was used to measure the arterial blood gas index. Protein levels of pro-inflammatory cytokines in serum, bronchoalveolar lavage fluid (BALF), and lung tissue was measured using an ELISA. Quantitative RT-PCR was used to measure changes in RNA levels of pro-inflammatory cytokines in the lungs. A fluorometric assay kit was used for determination of apoptosis-related factors to assess anti-apoptotic effects of PB. Western blotting was used to assess levels of key pathway proteins and apoptosis-related proteins. Connections between the pathways were tested through inhibitor experiments. RESULTS Pretreatment with PB (15 mg kg-1 d-1, i.g.) significantly reduced lung wet/dry weight ratios and MPO activity in blood and BALF of ALI mice, and it alleviated LPS-induced inflammatory cell infiltration in lung tissue. The levels of pro-inflammatory factors TNF-α, IL-6, and IL-1β and their mRNA levels in blood, BALF, and lung tissue were reduced following PB pretreatment. PB pretreatment also downregulated the apoptotic factors caspase-3, caspase-9, and apoptotic protein Bax, and it upregulated apoptotic protein Bcl-2. The NF-κB and NLRP3 pathways were inhibited through activation of the PI3K/Akt pathway due to PB pretreatment, whereas administration of PI3K inhibitors increased activation of these pathways. CONCLUSIONS Taken together, our results suggest that the anti-ALI properties of PB are closely associated with the inactivation of NF-κB and NLRP3 by altering the PI3K/Akt pathway. Furthermore, our findings provide a novel strategy for application of PB as a potential agent for treating patients with ALI. To the best of our knowledge, this is the first study to elucidate the underlying mechanism of action of PB against ALI.
Collapse
Affiliation(s)
- Renxing Zhong
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Tianyi Xia
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Yi Wang
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Zihe Ding
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Wei Li
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Ying Chen
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Mingming Peng
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Chuanqiu Li
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Han Zhang
- School of Pharmacy, Jiamusi University, Jiamusi, 154007, PR China
| | - Zunpeng Shu
- Guangdong Standardized Processing Engineering Technology Research Center of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China; The College of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
23
|
Poldip2/Nox4 Mediates Lipopolysaccharide-Induced Oxidative Stress and Inflammation in Human Lung Epithelial Cells. Mediators Inflamm 2022; 2022:6666022. [PMID: 35140544 PMCID: PMC8818432 DOI: 10.1155/2022/6666022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 11/10/2021] [Accepted: 12/30/2021] [Indexed: 12/05/2022] Open
Abstract
NADPH oxidase 4 (Nox4) is an important source of reactive oxygen species (ROS) production, and its expression is increased in lipopolysaccharide- (LPS-) stimulated lung epithelial cells. Polymerase δ-interacting protein 2 (Poldip2) has been proved to bind Nox4 and participates in oxidative stress and inflammation. However, the role of Poldip2/Nox4 in LPS-induced oxidative stress and inflammation in lung epithelial cells remains unclear. Cell viability was measured via MTT assays. The expression of Poldip2, Nox4, heme oxygenase-1 (HO-1), cyclooxygenase-2 (COX-2), AKT, and p-AKT was detected by Western blotting and/or immunofluorescence. Poldip2 and Nox4 interaction was analyzed via coimmunoprecipitation (Co-IP) assay. NADPH enzymatic activity and production of ROS, prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) were assessed simultaneously. The small interfering RNA (siRNA) or plasmid targeting Nox4 was used to downregulate or upregulate Nox4, and the lentiviral vector encoding Poldip2 was used to downregulate or upregulate Poldip2. The present study demonstrated that LPS stimulation significantly increased the protein levels of Poldip2 and Nox4 and proved that Poldip2 interacted with Nox4 proved by Co-IP. Importantly, Poldip2 acted as an upstream regulator of Nox4. The increased expression of Nox4 and COX-2; NADPH enzymatic activity; production of ROS, PGE2, TNF-α, and IL-1β; and decreased HO-1 expression were significantly suppressed by lentiviral Poldip2 shRNA downregulation but were increased by lentiviral upregulation of Poldip2. Furthermore, inhibiting of PI3K-AKT signaling notably attenuated LPS-induced Poldip2/Nox4 activation. Our study demonstrated that Poldip2 mediates LPS-induced oxidative stress and inflammation via interaction with Nox4 and was regulated by the PI3K-AKT signaling. Targeting Poldip2 could be a beneficial therapeutic strategy for the treatment of ALI.
Collapse
|
24
|
Lv X, Yao T, He R, He Y, Li M, Han Y, Zhang Y, Long L, Jiang G, Cheng X, Xie Y, Huang L, Peng Z, Hu G, Li Q, Tao L, Meng J. Protective Effect of Fluorofenidone Against Acute Lung Injury Through Suppressing the MAPK/NF-κB Pathway. Front Pharmacol 2022; 12:772031. [PMID: 34987397 PMCID: PMC8721041 DOI: 10.3389/fphar.2021.772031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a severe disease that presents serious damage and excessive inflammation in lungs with high mortality without effective pharmacological therapy. Fluorofenidone (AKFPD) is a novel pyridone agent that has anti-fibrosis, anti-inflammation, and other pharmacological activities, while the effect of fluorofenidone on ALI is unclarified. Here, we elucidated the protective effects and underlying mechanism of fluorofenidone on lipopolysaccharide (LPS)-induced ALI. In this study, fluorofenidone alleviated lung tissue structure injury and reduced mortality, decreased the pulmonary inflammatory cell accumulation and level of inflammatory cytokines IL-1β, IL-6, and TNF-α in the bronchoalveolar lavage fluid, and attenuated pulmonary apoptosis in LPS-induced ALI mice. Moreover, fluorofenidone could block LPS-activated phosphorylation of ERK, JNK, and P38 and further inhibited the phosphorylation of IκB and P65. These results suggested that fluorofenidone can significantly contrast LPS-induced ALI through suppressing the activation of the MAPK/NF-κB signaling pathway, which indicates that fluorofenidone could be considered as a novel therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Xin Lv
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Tingting Yao
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Rongling He
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yijun He
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Mengyu Li
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Han
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Zhang
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Lingzhi Long
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Guoliang Jiang
- Department of Respirology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun Cheng
- Department of Respirology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China
| | - Ling Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Gaoyun Hu
- Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qianbin Li
- Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| | - Jie Meng
- Department of Respirology, Third Xiangya Hospital, Central South University, Changsha, China.,Organ Fibrosis Key Laboratory of Hunan Province, Changsha, China.,National International Collaborative Research Center for Medical Metabolomics, Changsha, China
| |
Collapse
|
25
|
Zhu L, Wu H, Ma Z, Dong D, Yang Z, Tian J. Astaxanthin ameliorates lipopolysaccharide-induced acute lung injury via inhibition of inflammatory reactions and modulation of the SOCS3/JAK2/STAT3 signaling pathways in mice. Food Funct 2022; 13:11638-11651. [DOI: 10.1039/d2fo02182j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The results showed that astaxanthin had a protective effect on LPS-induced acute lung injury in mice, and its protective mechanism was through activating the SOCS3/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Li Zhu
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Huihui Wu
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Zhenbo Ma
- Medical Imaging Center, Taian City Central Hospital, No. 29, Longtan Road, Taian 271000, China
| | - Decheng Dong
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Ze Yang
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Jing Tian
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| |
Collapse
|
26
|
Umar T, Ma X, Yin B, Umer S, Zahoor A, Akhtar M, Umar Z, Shaukat A, Deng G. miR-424-5p overexpression inhibits LPS-stimulated inflammatory response in bovine endometrial epithelial cells by targeting IRAK2. J Reprod Immunol 2021; 150:103471. [PMID: 35032930 DOI: 10.1016/j.jri.2021.103471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 11/26/2022]
Abstract
Endometritis is inflammation of endometrium due to various factors and is a common cause of infertility. Several remedies used for endometritis like antibiotics, hormones, and herbs. Studies confirm that microRNAs play a significant role in various inflammatory diseases. However, the role of miR-424-5p in endometritis is not clear. In our study, histopathology, real-time quantitative polymerase chain reaction, Western blot analysis, immunofluorescence, ELISA, and dual-luciferase reporter assay were used to elucidate the effect of miR-424-5p in lipopolysaccharide (LPS)-primed inflammatory response in bovine endometrial epithelial cells (BEECs) and clarify the potential mechanism. Our results revealed that miR-424-5p mimics noticeably decrease the production of proinflammatory cytokines (IL-1β, IL-6, and TNF-α), while miR-424-5p inhibitors have inverse effects in BEECs. Moreover, overexpression of miR-424-5p on BEECs cells also suppressed NF-κB p65 activation. Afterwards, we verified that miR-424-5p inhibited Interleukin 1 Receptor Associated Kinase 2 (IRAK2) expression by binding to the 3'-UTR of IRAK2 mRNA. Further, co-transfection of miR-424-5p inhibitors and siRNA-IRAK2 revealed that negative regulation of miR-424-5p on LPS-induced inflammatory response in BEECs was mediated by IRAK2.Mutually, miR-424-5p pharmacologic stabilization represents an entirely unique medical aid for cow endometritis and other inflammation-related diseases.
Collapse
Affiliation(s)
- Talha Umar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Xiaofei Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Baoyi Yin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Saqib Umer
- Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, PR China; Department of Theriogenology, Faculty of Veterinary Science, University of Agriculture, Faisalabad, 38000 Punjab, Pakistan
| | - Arshad Zahoor
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Muhammad Akhtar
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Zaima Umar
- Department of Anatomy and Histology, Faculty of Veterinary Science, University of Agriculture, Faisalabad, 38000 Punjab, Pakistan
| | - Aftab Shaukat
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.
| |
Collapse
|
27
|
Yang W, Jiang X, Liu J, Qi D, Luo Z, Yu G, Li X, Sen M, Chen H, Liu W, Liu Y, Wang G. Integrated Strategy From In Vitro, In Situ, In Vivo to In Silico for Predicting Active Constituents and Exploring Molecular Mechanisms of Tongfengding Capsule for Treating Gout by Inhibiting Inflammatory Responses. Front Pharmacol 2021; 12:759157. [PMID: 34912220 PMCID: PMC8666879 DOI: 10.3389/fphar.2021.759157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/07/2023] Open
Abstract
The study of screening active constituents from traditional Chinese medicine (TCM) is important for explicating the mechanism of action of TCM and further evaluating the safety and efficacy effectively. However, detecting and identifying the active constituents from complicated biological samples still remain a challenge. Here, a practical, quick, and novel integrated strategy from in vitro, in situ, in vivo to in silico for rapidly screening the active constituents was developed. Firstly, the chemical profile of TCM in vitro was identified using UPLC-Q Exactive-Orbitrap HRMS. Secondly, the in situ intestinal perfusion with venous sampling (IPVS) method was used to investigate the intestinal absorption components. Thirdly, after intragastric administration of the TCM extract, the in vivo absorbed prototype components were detected and identified. Finally, the target network pharmacology approach was applied to explore the potential targets and possible mechanisms of the absorbed components from TCM. The reliability and availability of this approach was demonstrated using Tongfengding capsule (TFDC) as an example of herbal medicine. A total of 141 compounds were detected and identified in TFDC, and among them, 64 components were absorbed into the plasma. Then, a total of 35 absorbed bioactive components and 50 related targets shared commonly by compounds and gout were integrated via target network pharmacology analysis. Ultimately, the effects of the absorbed components on metabolism pathways were verified by experiments. These results demonstrated that this original method may provide a practical tool for screening bioactive compounds from TCM treating particular diseases. Furthermore, it also can clarify the potential mechanism of action of TCM and rationalize the application of TFDC as an effective herbal therapy for gout.
Collapse
Affiliation(s)
- Wenning Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoquan Jiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingtong Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Dongying Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiqiang Luo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Guohua Yu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xueyan Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Muli Sen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongjiao Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guopeng Wang
- Zhongcai Health (Beijing) Biological Technology Development Co., Ltd., Beijing, China
| |
Collapse
|
28
|
Alhassen L, Dabbous T, Ha A, Dang LHL, Civelli O. The Analgesic Properties of Corydalis yanhusuo. Molecules 2021; 26:molecules26247498. [PMID: 34946576 PMCID: PMC8704877 DOI: 10.3390/molecules26247498] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/29/2022] Open
Abstract
Corydalis yanhusuo extract (YHS) has been used for centuries across Asia for pain relief. The extract is made up of more than 160 compounds and has been identified as alkaloids, organic acids, volatile oils, amino acids, alcohols, and sugars. However, the most crucial biological active constituents of YHS are alkaloids; more than 80 have been isolated and identified. This review paper aims to provide a comprehensive review of the phytochemical and pharmacological effects of these alkaloids that have significant ties to analgesia.
Collapse
Affiliation(s)
- Lamees Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92697, USA; (L.A.); (T.D.); (A.H.); (L.H.L.D.)
| | - Travis Dabbous
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92697, USA; (L.A.); (T.D.); (A.H.); (L.H.L.D.)
| | - Allyssa Ha
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92697, USA; (L.A.); (T.D.); (A.H.); (L.H.L.D.)
| | - Leon Hoang Lam Dang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92697, USA; (L.A.); (T.D.); (A.H.); (L.H.L.D.)
| | - Olivier Civelli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California-Irvine, Irvine, CA 92697, USA; (L.A.); (T.D.); (A.H.); (L.H.L.D.)
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California-Irvine, Irvine, CA 92697, USA
- Correspondence:
| |
Collapse
|
29
|
Kim SM, Min JH, Kim JH, Choi J, Park JM, Lee J, Goo SH, Oh JH, Kim SH, Chun W, Ahn KS, Kang S, Lee JW. Methyl p‑hydroxycinnamate exerts anti‑inflammatory effects in mouse models of lipopolysaccharide‑induced ARDS. Mol Med Rep 2021; 25:37. [PMID: 34859262 PMCID: PMC8669673 DOI: 10.3892/mmr.2021.12553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Methyl p-hydroxycinnamate (MH), an esterified derivative of p-Coumaric acid exerts anti-inflammatory effects on lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Based on these effects, the present study investigated the protective role of MH in a mouse model of LPS-induced acute respiratory distress syndrome (ARDS). The results demonstrated that administration of LPS (5 mg/kg intranasally) markedly increased the neutrophil/macrophage numbers and levels of inflammatory molecules (TNF-α, IL-6, IL-1β and reactive oxygen species) in the bronchoalveolar lavage fluid (BALF) of mice. On histological examination, the presence of inflammatory cells was observed in the lungs of mice administered LPS. LPS also notably upregulated the secretion of monocyte chemoattractant protein-1 and protein content in BALF as well as expression of inducible nitric oxide synthase in the lungs of mice; it also caused activation of p38 mitogen-activated protein kinase (MAPK) and NF-κB signaling. However, MH treatment significantly suppressed LPS-induced upregulation of inflammatory cell recruitment, inflammatory molecule levels and p38MAPK/NF-κB activation, and also led to upregulation of heme oxygenase-1 (HO-1) expression in the lungs of mice. In addition, the ability of MH to induce HO-1 expression was confirmed in RAW264.7 macrophages. Taken together, the findings of the present study indicated that MH may exert protective effects against airway inflammation in ARDS mice by inhibiting inflammatory cell recruitment and the production of inflammatory molecules.
Collapse
Affiliation(s)
- Seong-Man Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Jae-Hong Min
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Jung Hee Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Jinseon Choi
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Jin-Mi Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Juhyun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Soo Hyeon Goo
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Jae Hoon Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Seung-Ho Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| | - Sukmo Kang
- Biotoxtech Co., Ltd., Ochang, Cheongju, Chungcheongbuk‑do 28115, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju, Chungcheongbuk‑do 28116, Republic of Korea
| |
Collapse
|
30
|
Yuan Y, Dong FX, Liu X, Xiao HB, Zhou ZG. Liquid Chromatograph-Mass Spectrometry-Based Non-targeted Metabolomics Discovery of Potential Endogenous Biomarkers Associated With Prostatitis Rats to Reveal the Effects of Magnoflorine. Front Pharmacol 2021; 12:741378. [PMID: 34790120 PMCID: PMC8591080 DOI: 10.3389/fphar.2021.741378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022] Open
Abstract
Magnoflorine (Mag) has multiple pharmacological activities for the prevention and treatment of prostatitis. However, its molecular mechanisms andpharmacological targets are not clear. In this study, the ultra-performance liquid tandem mass spectrometry-based metabolomics method was used to clarify the intervention of Mag against prostatitis and the biological mechanism. A total of 25 biomarkers associated with the prostatitis model were identified by metabolomics, and a number of metabolic pathways closely related to the model were obtained by MetPA analysis. After given Mag treatment, the results of each indicator were shown that Mag alkaloid could inhibit the development of prostatitis effectively. We found that Mag had regulative effects on potential biomarkers of prostatitis model, which can regulate them to the control group. Our results indicated that alkaloids have an effective intervention therapy for prostatitis, and five types of metabolic pathways closely related to prostatitis model were obtained, including phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, tyrosine metabolism, arginine and proline metabolism, glycine, serine and threonine metabolism, alanine, aspartate and glutamate metabolism. This study has provided the basic experimental data for the development of Mag in the prevention and treatment of prostatitis.
Collapse
Affiliation(s)
- Yin Yuan
- Department of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fei-Xue Dong
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Liu
- Department of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hong-Bin Xiao
- Department of Basic Medicine, College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhong-Guang Zhou
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
31
|
Wang S, Jia D, Lu H, Qu X. Paeoniflorin improves myocardial injury via p38 MAPK/NF-KB p65 inhibition in lipopolysaccharide-induced mouse. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1449. [PMID: 34734001 PMCID: PMC8506776 DOI: 10.21037/atm-21-4049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/07/2021] [Indexed: 12/19/2022]
Abstract
Background Paeoniflorin (Pae) is an active compound with a variety of pharmacological effects. This aim was to investigate how Pae protects against myocardial injury and to explore its potential mechanism. Methods We established a BALB/c mouse model that was intraperitoneal injection (i.p.) of RvE1 (25 µg/kg) or Pae (20 mg/kg) for 3 days, and then treated with lipopolysaccharide (LPS, 10 mg/kg, i.p.). The mice were randomly divided into the sham group, the LPS group, the LPS + RvE1 group, the LPS + Pae group (n=8). Cardiac dysfunction was detected by HE staining and ELISA assay. The oxidative stress, mitochondrial membrane potential (MMP), mitochondrial permeability transition pore (mPTP) and apoptosis were assessed. Furthermore, western blotting (WB) assay were employed to analyze the protective mechanisms. Results Pae improved LPS-induced cardiac function and impeded apoptosis. Pae significantly reduced the release of inflammatory cytokines such as interleukin (IL)-6, tumor necrosis factor-α (TNF-α), and IL-1β. Furthermore, Pae decreased malondialdehyde (MDA), glutathione (GSH), and reactive oxygen species (ROS), and increased superoxide dismutase (SOD). In addition, Pae attenuated the mPTP opening and MMP depolarization. Notably, Pae treatment inhibited the activation of p38 MAPK and NF-κB p65. Conclusions It was confirmed that Pae alleviated LPS-induced myocardial injury. Pae might be as a new drug candidate for myocardial ischaemic complications.
Collapse
Affiliation(s)
- Shaojun Wang
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dong Jia
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haimiao Lu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiufen Qu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Singh B, Nathawat S, Sharma RA. Ethnopharmacological and phytochemical attributes of Indian Tinospora species: A comprehensive review. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103381] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
33
|
Li R, Ai X, Hou Y, Lai X, Meng X, Wang X. Amelioration of diabetic retinopathy in db/db mice by treatment with different proportional three active ingredients from Tibetan medicine Berberis dictyophylla F. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114190. [PMID: 33964362 DOI: 10.1016/j.jep.2021.114190] [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: 02/07/2021] [Revised: 04/09/2021] [Accepted: 05/02/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Berberis dictyophylla F., a famous Tibetan medicine, has been used to prevent and treat diabetic retinopathy (DR) for thousands of years in clinic. However, its underlying mechanisms remain unclear. AIM OF THE STUDY The present study was designed to probe the synergistic protection and involved mechanisms of berberine, magnoflorine and berbamine from Berberis dictyophylla F. on the spontaneous retinal damage of db/db mice. MATERIALS AND METHODS The 14-week spontaneous model of DR in db/db mice were randomly divided into eight groups: model group, calcium dobesilate (CaDob, 0.23 g/kg) group and groups 1-6 (different proportional three active ingredients from Berberis dictyophylla F.). All mice were intragastrically administrated for a continuous 12 weeks. Body weight and fasting blood glucose (FBG) were recorded and measured. Hematoxylin-eosin and periodic acid-Schiff (PAS) stainings were employed to evaluate the pathological changes and abnormal angiogenesis of the retina. ELISA was performed to assess the levels of IL-6, HIF-1α and VEGF in the serum. Immunofluorescent staining was applied to detect the protein levels of CD31, VEGF, p-p38, p-JNK, p-ERK and NF-κB in retina. In addition, mRNA expression levels of VEGF, Bax and Bcl-2 in the retina were monitored by qRT-PCR analysis. RESULTS Treatment with different proportional three active ingredients exerted no significant effect on the weight, but decreased the FBG, increased the number of retinal ganglionic cells and restored internal limiting membrane. The results of PAS staining demonstrated that the drug treatment decreased the ratio of endothelial cells to pericytes while thinned the basal membrane of retinal vessels. Moreover, these different proportional active ingredients can markedly downregulate the protein levels of retinal CD31 and VEGF, and serum HIF-1α and VEGF. The gene expression of retinal VEGF was also suppressed. The levels of retinal p-p38, p-JNK and p-ERK proteins were decreased by drug treatment. Finally, drug treatment reversed the proinflammatory factors of retinal NF-κB and serum IL-6, and proapoptotic Bax gene expression, while increased antiapoptotic Bcl-2 gene expression. CONCLUSIONS These results indicated that DR in db/db mice can be ameliorated by treatment with different proportional three active ingredients from Berberis dictyophylla F. The potential vascular protection mechanisms may be involved in inhibiting the phosphorylation of the MAPK signaling pathway, thus decreasing inflammatory and apoptotic events.
Collapse
Affiliation(s)
- Rui Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaopeng Ai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ya Hou
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianrong Lai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xiaobo Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
34
|
Zhao F, Guo Z, Hou F, Fan W, Wu B, Qian Z. Magnoflorine Alleviates "M1" Polarized Macrophage-Induced Intervertebral Disc Degeneration Through Repressing the HMGB1/Myd88/NF-κB Pathway and NLRP3 Inflammasome. Front Pharmacol 2021; 12:701087. [PMID: 34366853 PMCID: PMC8343137 DOI: 10.3389/fphar.2021.701087] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Intervertebral disc degeneration (IDD) is related to the deterioration of nucleus pulposus (NP) cells due to hypertrophic differentiation and calcification. The imbalance of pro-inflammatory (M1 type) and anti-inflammatory (M2 type) macrophages contributes to maintaining tissue integrity. Here, we aimed to probe the effect of Magnoflorine (MAG) on NP cell apoptosis mediated by “M1” polarized macrophages. THP-1 cells were treated with lipopolysaccharide (LPS) to induce “M1” polarized macrophages. Under the treatment with increasing concentrations of MAG, the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, IL-18), high mobility group box protein 1 (HMGB1), as well as myeloid differentiation factor 88 (MyD88), nuclear factor kappa B (NF-κB) and NOD-like receptor 3 (NLRP3) inflammasomes in THP-1 cells were determined. What’s more, human NP cells were treated with the conditioned medium (CM) from THP-1 cells. The NP cell viability and apoptosis were evaluated. Western blot (WB) was adopted to monitor the expression of apoptosis-related proteins (Bax, Caspase3, and Caspase9), catabolic enzymes (MMP-3, MMP-13, ADAMTS-4, and ADAMTS-5), and extracellular matrix (ECM) compositions (collagen II and aggrecan) in NP cells. As a result, LPS evidently promoted the expression of pro-inflammatory cytokines and HMGB1, the MyD88-NF-κB activation, and the NLRP3 inflammasome profile in THP-1 cells, while MAG obviously inhibited the "M1″ polarization of THP-1 cells. After treatment with “M1” polarized THP-1 cell CM, NP cell viability was decreased, while cell apoptosis, the pro-inflammatory cytokines, apoptosis-related proteins, and catabolic enzymes were distinctly up-regulated, and ECM compositions were reduced. After treatment with MAG, NP cell damages were dramatically eased. Furthermore, MAG dampened the HMGB1 expression and inactivated the MyD88/NF-κB pathway and NLRP3 inflammasome in NP cells. In conclusion, this study confirmed that MAG alleviates “M1” polarized macrophage-mediated NP cell damage by inactivating the HMGB1-MyD88-NF-κB pathway and NLRP3 inflammasome, which provides a new reference for IDD treatment.
Collapse
Affiliation(s)
- Feng Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhenye Guo
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Fushan Hou
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wei Fan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Binqiang Wu
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhonglai Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
35
|
Herbal Active Ingredients: Potential for the Prevention and Treatment of Acute Lung Injury. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5543185. [PMID: 34258266 PMCID: PMC8245226 DOI: 10.1155/2021/5543185] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a life-threatening clinical syndrome with high morbidity and mortality. The main pathological features of ALI are increased alveolar-capillary membrane permeability, edema, uncontrolled migration of neutrophils to the lungs, and diffuse alveolar damage, resulting in acute hypoxemic respiratory failure. Glucocorticoids, aspirin, and other anti-inflammatory drugs are commonly used to treat ALI. Respiratory supports, such as a ventilator, are used to alleviate hypoxemia. Many treatment methods are available, but they cannot significantly ameliorate the quality of life of patients with ALI and reduce mortality rates. Herbal active ingredients, such as flavonoids, terpenoids, saponins, alkaloids, and quinonoids, exhibit advantages for ALI prevention and treatment, but the underlying mechanism needs further study. This paper summarizes the role of herbal active ingredients in anti-ALI therapy and progresses in the understanding of their mechanisms. The work also provides some references and insights for the discovery and development of novel drugs for ALI prevention and treatment.
Collapse
|
36
|
A comprehensive review of natural products against atopic dermatitis: Flavonoids, alkaloids, terpenes, glycosides and other compounds. Biomed Pharmacother 2021; 140:111741. [PMID: 34087696 DOI: 10.1016/j.biopha.2021.111741] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/24/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022] Open
Abstract
Atopic dermatitis (AD) is considered a great challenge for human communities and imposes both physiological and mental burdens on patients. Natural products have widely been used to treat a wide range of diseases, including cancer, gastrointestinal diseases, asthma, neurological disorders, and infections. To seek potential natural products against AD, in the current review, we searched the terms "atopic dermatitis" and "natural product" in Pubmed, Medline, Web of Science,Science Direct, Embase, EBSCO, CINAHL, ACS. The results show that many natural products, especially puerarin, ferulic acid and ginsenosides, cound protect against AD. Meanwhile, we discussed the therapeutic mechanisms and showed that the natural products exert their anti-inflammatory effects by suppressing the quantity and activity of many inflammatory cell types and cytokines, including neutrophils, monocytes, lymphocytes, Langerhans cells, interleukins (ILs, including IL-1α, IL-1β, IL-4), TNF-α, and TSLP, IgE. via inhibition of JAK/STAT, MAPKs and NF-κB signaling pathways, thereby, halting the inflammatory cascade. Future investigations should focus on studies with more reflective of the clinical characteristics and demographics, so as to develop natural products that will be hopefully available for the treatment of human AD disease.
Collapse
|
37
|
Activation of TREM-1 induces endoplasmic reticulum stress through IRE-1α/XBP-1s pathway in murine macrophages. Mol Immunol 2021; 135:294-303. [PMID: 33957479 DOI: 10.1016/j.molimm.2021.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that endoplasmic reticulum (ER) stress activates several pro-inflammatory signaling pathways in many diseases, including acute lung injury (ALI). We have reported that blocking triggering receptor expressed on myeloid cells 1 (TREM-1) protects against ALI by suppressing pulmonary inflammation in mice with ALI induced by lipopolysaccharides (LPS). However, the molecular mechanism underlying the TREM-1-induced pro-inflammatory microenvironment in macrophages remains unclearly. Herein, we aimed to determine whether TREM-1 regulates the inflammatory responses induced by LPS associated with ER stress activation. We found that the activation of TREM-1 by a monoclonal agonist antibody (anti-TREM-1) increased the mRNA and protein levels of IL-1β, TNF-α, and IL-6 in primary macrophages. Treatment of the anti-TREM-1 antibody increased the expression of ER stress markers (ATF6, PERK, IRE-1α, and XBP-1s) in primary macrophages. While pretreatment with 4-PBA, an inhibitor of ER stress, significantly inhibited the expression of ER stress markers and pro-inflammatory cytokines and reduced LDH release. Furthermore, inhibiting the activity of the IRE-1α/XBP-1s pathway by STF-083010 significantly mitigated the increased levels of IL-1β, TNF-α, and IL-6 in macrophages treated by the anti-TREM-1 antibody. XBP-1 silencing attenuated pro-inflammatory microenvironment evoked by activation of TREM-1. Besides, we found that blockade of TREM-1 with LR12 ameliorated ER stress induced by LPS in vitro and in vivo. In conclusion, we conclude that TREM-1 activation induces ER stress through the IRE-1α/XBP-1s pathway in macrophages, contributing to the pro-inflammatory microenvironment.
Collapse
|
38
|
Huang Z, Xie N, Illes P, Di Virgilio F, Ulrich H, Semyanov A, Verkhratsky A, Sperlagh B, Yu SG, Huang C, Tang Y. From purines to purinergic signalling: molecular functions and human diseases. Signal Transduct Target Ther 2021; 6:162. [PMID: 33907179 PMCID: PMC8079716 DOI: 10.1038/s41392-021-00553-z] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/24/2021] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Purines and their derivatives, most notably adenosine and ATP, are the key molecules controlling intracellular energy homoeostasis and nucleotide synthesis. Besides, these purines support, as chemical messengers, purinergic transmission throughout tissues and species. Purines act as endogenous ligands that bind to and activate plasmalemmal purinoceptors, which mediate extracellular communication referred to as "purinergic signalling". Purinergic signalling is cross-linked with other transmitter networks to coordinate numerous aspects of cell behaviour such as proliferation, differentiation, migration, apoptosis and other physiological processes critical for the proper function of organisms. Pathological deregulation of purinergic signalling contributes to various diseases including neurodegeneration, rheumatic immune diseases, inflammation, and cancer. Particularly, gout is one of the most prevalent purine-related disease caused by purine metabolism disorder and consequent hyperuricemia. Compelling evidence indicates that purinoceptors are potential therapeutic targets, with specific purinergic agonists and antagonists demonstrating prominent therapeutic potential. Furthermore, dietary and herbal interventions help to restore and balance purine metabolism, thus addressing the importance of a healthy lifestyle in the prevention and relief of human disorders. Profound understanding of molecular mechanisms of purinergic signalling provides new and exciting insights into the treatment of human diseases.
Collapse
Grants
- National Key R&D Program of China (2019YFC1709101,2020YFA0509400, 2020YFC2002705), the National Natural Science Foundation of China (81821002, 81790251, 81373735, 81972665), Guangdong Basic and Applied Basic Research Foundation (2019B030302012), the Project First-Class Disciplines Development of Chengdu University of Traditional Chinese Medicine (CZYHW1901), São Paulo Research Foundation (FAPESP 2018/07366-4), Russian Science Foundation grant 20-14-00241, NSFC-BFBR;and Science and Technology Program of Sichuan Province, China (2019YFH0108)
- National Key R&D Program of China (2020YFA0509400, 2020YFC2002705), the National Natural Science Foundation of China (81821002, 81790251).
- National Key R&D Program of China (2020YFA0509400, 2020YFC2002705), the National Natural Science Foundation of China (81821002, 81790251), Guangdong Basic and Applied Basic Research Foundation (2019B030302012).
- the Project First-Class Disciplines Development of Chengdu University of Traditional Chinese Medicine (CZYHW1901) and Science and Technology Program of Sichuan Province, China (2019YFH0108).
- the Project First-Class Disciplines Development of Chengdu University of Traditional Chinese Medicine (CZYHW1901), and Science and Technology Program of Sichuan Province, China (2019YFH0108).
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Peter Illes
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universitaet Leipzig, Leipzig, Germany
| | | | - Henning Ulrich
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Alexey Semyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexei Verkhratsky
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Sechenov First Moscow State Medical University, Moscow, Russia
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Beata Sperlagh
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Shu-Guang Yu
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yong Tang
- International Collaborative Centre on Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
39
|
Balkrishna A, Khandrika L, Varshney A. Giloy Ghanvati ( Tinospora cordifolia (Willd.) Hook. f. and Thomson) Reversed SARS-CoV-2 Viral Spike-Protein Induced Disease Phenotype in the Xenotransplant Model of Humanized Zebrafish. Front Pharmacol 2021; 12:635510. [PMID: 33953674 PMCID: PMC8091047 DOI: 10.3389/fphar.2021.635510] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/26/2021] [Indexed: 12/11/2022] Open
Abstract
The current Severe Acute Respiratory Syndrome disease caused by Coronavirus-2 (SARS-CoV-2) has been a serious strain on the healthcare infrastructure mainly due to the lack of a reliable treatment option. Alternate therapies aimed at symptomatic relief are currently prescribed along with artificial ventilation to relieve distress. Traditional medicine in the form of Ayurveda has been used since ancient times as a holistic treatment option rather than targeted therapy. The practice of Ayurveda has several potent herbal alternatives for chronic cough, inflammation, and respiratory distress which are often seen in the SARS-CoV-2 infection. In this study we have used the aqueous extracts of Tinospora cordifolia (willd.) Hook. f. and Thomson in the form of Giloy Ghanvati, as a means of treatment to the SARS-CoV-2 spike-protein induced disease phenotype in a humanized zebrafish model. The introduction of spike-protein in the swim bladder transplanted with human lung epithelial cells (A549), caused an infiltration of pro-inflammatory immune cells such as granulocytes and macrophages into the swim bladder. There was also an increased systemic damage as exemplified by renal tissue damage and increased behavioral fever in the disease induction group. These features were reversed in the treatment group, fed with three different dosages of Giloy Ghanvati. The resultant changes in the disease phenotype were comparable to the group that were given the reference compound, Dexamethasone. These findings correlated well with various phyto-compounds detected in the Giloy Ghanvati and their reported roles in the viral disease phenotype amelioration.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, India
| | | | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, Haridwar, India
- Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Haridwar, India
| |
Collapse
|
40
|
Lee HC, Liu FC, Tsai CN, Chou AH, Liao CC, Yu HP. Esculetin Ameliorates Lipopolysaccharide-Induced Acute Lung Injury in Mice Via Modulation of the AKT/ERK/NF-κB and RORγt/IL-17 Pathways. Inflammation 2021; 43:962-974. [PMID: 32170603 DOI: 10.1007/s10753-020-01182-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Esculetin, a coumarin derivative from various natural plants, has an anti-inflammatory property. In the present study, we examined if esculetin has any salutary effects against lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Acute lung injury (ALI) was induced via the intratracheal administration of LPS, and esculetin (20 and 40 mg/kg) was given intraperitoneally 30 min before LPS challenge. After 6 h of LPS administration, lung tissues were collected for analysis. Pretreatment with esculetin significantly attenuated histopathological changes, inflammatory cell infiltration, and production of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, in the lung tissue. Furthermore, esculetin inhibited the protein kinase B (AKT), extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B (NF-κB) pathways and downregulated the expression of RORγt and IL-17 in LPS-induced ALI. Our results indicated that esculetin possesses anti-inflammatory and protective effects against LPS-induced ALI via inhibition of the AKT/ERK/NF-κB and RORγt/IL-17 pathways.
Collapse
Affiliation(s)
- Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
41
|
Li J, Lu K, Sun F, Tan S, Zhang X, Sheng W, Hao W, Liu M, Lv W, Han W. Panaxydol attenuates ferroptosis against LPS-induced acute lung injury in mice by Keap1-Nrf2/HO-1 pathway. J Transl Med 2021; 19:96. [PMID: 33653364 PMCID: PMC7927246 DOI: 10.1186/s12967-021-02745-1] [Citation(s) in RCA: 276] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/08/2021] [Indexed: 12/27/2022] Open
Abstract
Background Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) induces uncontrolled and self-amplified pulmonary inflammation, and has high morbidity and mortality rates in critically ill patients. In recent years, many bioactive ingredients extracted from herbs have been reported to effectively ameliorate ALI/ARDS via different mechanisms. Ferroptosis, categorized as regulated necrosis, is more immunogenic than apoptosis and contributes to the progression of ALI. In this study, we examined the impact of panaxydol (PX), isolated from the roots of Panax ginseng, on lipopolysaccharide (LPS)-induced ALI in mice. Methods In vivo, the role of PX on LPS-induced ALI in mice was tested by determination of LPS-induced pulmonary inflammation, pulmonary edema and ferroptosis. In vitro, BEAS-2B cells were used to investigate the molecular mechanisms by which PX functions via determination of inflammation, ferroptosis and their relationship. Results Administration of PX protected mice against LPS-induced ALI, including significantly ameliorated lung pathological changes, and decreased the extent of lung edema, inflammation, and ferroptosis. In vitro, PX inhibited LPS-induced ferroptosis and inflammation in bronchial epithelial cell line BEAS-2B cells. The relationship between ferroptosis and inflammation was investigated. The results showed that ferroptosis mediated inflammation in LPS-treated BEAS-2B cells, and PX might ameliorate LPS-induced inflammation via inhibiting ferroptosis. Meanwhile, PX could upregulate Keap1-Nrf2/HO-1 pathway, and selective inhibition of Keap1-Nrf2/HO-1 pathway significantly abolished the anti-ferroptotic and anti-inflammatory functions of PX in LPS-treated cells. Conclusion PX attenuates ferroptosis against LPS-induced ALI via Keap1-Nrf2/HO-1 pathway, and is a promising novel therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Jiucui Li
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Kongmiao Lu
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Fenglan Sun
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Shanjuan Tan
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Xiao Zhang
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Wei Sheng
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Wanming Hao
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Min Liu
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China.
| | - Weihong Lv
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China.
| | - Wei Han
- Qingdao Municipal Hospital, School of Medicine, Qingdao University, No. 1, Jiaozhou Road, Qingdao, 266011, Shandong, China.
| |
Collapse
|
42
|
Sharma P, Gupta S, Goel N, Gupta A, Saini V, Sharma N. A review: novel coronavirus (COVID-19): an evidence-based approach. BIOMEDICAL ENGINEERING TOOLS FOR MANAGEMENT FOR PATIENTS WITH COVID-19 2021. [PMCID: PMC8192331 DOI: 10.1016/b978-0-12-824473-9.00007-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The World Health Organization in China was informed about the cases of pneumonia of unknown antecedent ailments. Since then, there have been over 141 million cases globally of 2019 novel coronavirus (Covid-19), 3.01 million deaths, and over 80.4 million recovered. Clinical research of novel agents represent opportunities to inform real-time public health action. In 2018 there was a systematic review to identify priority research questions for Severe Acute Respiratory Syndrome-related coronavirus and Middle East Respiratory Syndrome-related coronavirus. Here, we review information available on COVID-19 and provide evidenced-based approaches in clinical research for the current COVID-19 outbreak.
Collapse
|
43
|
Chang L, Wang Q, Ju J, Li Y, Cai Q, Hao L, Zhou Y. Magnoflorine Ameliorates Inflammation and Fibrosis in Rats With Diabetic Nephropathy by Mediating the Stability of Lysine-Specific Demethylase 3A. Front Physiol 2020; 11:580406. [PMID: 33414721 PMCID: PMC7785030 DOI: 10.3389/fphys.2020.580406] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) represents one of the most devastating complications for patients with diabetes. The anti-diabetic activities of Magnoflorine (MF) were reported, with underlying mechanism unknown. Lysine-specific demethylase 3A (KDM3A) was identified in the renal injuries. In the current study, we investigated the functional role of MF in DN progression with the involvement of KDM3A. We reported that in the animal model of DN induced by streptozotocin (STZ) injection, MF attenuated inflammatory response and fibrosis in the kidneys. In cultured mesangial cells, MF similarly ameliorated abnormal proliferation and lowered the expression of inflammation- and fibrosis-related factors stimulated by high glucose (HG) treatment. Upon MF treatment, there was a decline in KDM3A-positive cells in renal tissues of rats, accompanying an augment in KDM3A ubiquitination. KDM3A upregulation in vitro by a proteasome inhibitor MG132 comparably dampened the inhibitory role of MF in inflammatory response and fibrosis. Further analyses revealed that MF increased transforming growth factor β-induced factor 1 (TGIF1) transcriptional activity by promoting ubiquitination and degradation of KDM3A, thus inhibiting the activation of TGF-β1/Smad2/3 signaling pathway. TGIF1 silencing weakened the repressive role of MF in mesangial cells as well. In conclusion, MF contributes to TGIF1 transcription via an epigenetic mechanism.
Collapse
Affiliation(s)
- Liang Chang
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Wang
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiannan Ju
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Li
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiao Cai
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lirong Hao
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Zhou
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Park JW, Ryu HW, Ahn HI, Min JH, Kim SM, Kim MG, Kwon OK, Hwang D, Kim SY, Choi S, Zamora N, Rosales K, Oh SR, Lee JW, Ahn KS. The Anti-Inflammatory Effect of Trichilia martiana C. DC. in the Lipopolysaccharide-Stimulated Inflammatory Response in Macrophages and Airway Epithelial Cells and in LPS-Challenged Mice. J Microbiol Biotechnol 2020; 30:1614-1625. [PMID: 32876073 PMCID: PMC9728236 DOI: 10.4014/jmb.2006.06042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022]
Abstract
A number of species of the genus Trichilia (Meliaceae) exhibit anti-inflammatory effects. However, the effect of Trichilia martiana C. DC. (TM) on lipopolysaccharide (LPS)-induced inflammation has not, to the best of our knowledge, yet been determined. Therefore, in the present study, the antiinflammatory effect of TM on LPS-stimulated RAW264.7 macrophages was evaluated. The ethanol extract of TM (TMEE) significantly inhibited LPS-induced nitric oxide (NO), prostaglandin 2 (PGE2), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). TMEE also reduced the levels of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-1β and IL-6. The upregulation of mitogen-activated protein kinases (MAPKs) and NF-κB activation was revealed to be downregulated following TMEE pretreatment. Furthermore, TMEE was indicated to lead to the nucleus translocation of nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and the expression of heme oxygenase-1 (HO-1). In H292 airway epithelial cells, the pretreatment of TMEE significantly downregulated the production of LPS-stimulated IL-1β, and TMEE was indicated to increase the expression of HO-1. In animal models exhibiting LPS-induced acute lung injury (ALI), treatment with TMEE reduced the levels of macrophages influx and TNF-α production in the bronchoalveolar lavage fluid (BALF) of ALI mice. Additionally, TMEE significantly downregulated the activation of ERK, JNK and IκB, and upregulated the expression of HO-1 in the lungs of ALI mice. In conclusion, the results of the current study demonstrated that TMEE could exert a regulatory role in the prevention or treatment of the endotoxin-mediated inflammatory response.
Collapse
Affiliation(s)
- Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea
| | - Hyung Won Ryu
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea
| | - Hye In Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea
| | - Jae-Hong Min
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,College of Pharmacy, Chungbuk National University, Cheongju 8160, Republic of Korea
| | - Seong-Man Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,College of Pharmacy, Chungnam National University, Daejeon 414, Republic of Korea,
| | - Min-Gu Kim
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,College of Pharmacy, Chungbuk National University, Cheongju 8160, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea
| | - Daseul Hwang
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,College of Pharmacy, Chungbuk National University, Cheongju 8160, Republic of Korea
| | - Soo-Yong Kim
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 311, Republic of Korea
| | - Sangho Choi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 311, Republic of Korea
| | - Nelson Zamora
- Bioprospecting Research Unit, National Biodiversity Institute, Santo Domingo, Heredia 22-3100, Costa Rica
| | - Kattia Rosales
- Bioprospecting Research Unit, National Biodiversity Institute, Santo Domingo, Heredia 22-3100, Costa Rica
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea
| | - Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,Corresponding authors J-W.Lee Phone : +82-43-240-6135 Fax : +82-43-240-6129 E-mail:
| | - Kyung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 286, Republic of Korea,K-S.Ahn Phone : +82-43-240-6113 Fax : +82-43-240-6129 E-mail:
| |
Collapse
|
45
|
Chen H, Shi Z, Xing Y, Li X, Fu F. Fangchinoline attenuates cardiac dysfunction in rats with endotoxemia via the inhibition of ERK1/2 and NF-κB p65 phosphorylation. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1167. [PMID: 33241016 PMCID: PMC7576032 DOI: 10.21037/atm-20-5669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Cardiac dysfunction is a complication commonly encountered by patients with endotoxemia. Fangchinoline (Fan) is a natural bisbenzylisoquinoline alkaloid. This study aimed to investigate the cardioprotective effect of Fan against lipopolysaccharide (LPS)-induced acute cardiac dysfunction. Methods Rats were administered with Baicalin (100 mg/kg) and Fan (30 or 60 mg/kg) via intraperitoneal injection (i.p.) for 3 days, followed by LPS treatment (10 mg/kg, i.p.). The rats were randomly grouped (n=10): the control group, the LPS group, the LPS + Baicalin group, the LPS + Fan groups. Echocardiography and hematoxylin and eosin (HE) staining were performed to detect cardiac dysfunction. Cardiac function were also determined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), ELISA, and western blot, respectively. The protective mechanisms of Fan were analyzed by western blot and qRT-PCR. Results LPS induced the depression of cardiac function, myocardial inflammation, and apoptosis. These changes were associated with decreased GRP78 and GADD34, increased C/EBP-homologous protein (CHOP) and cleaved caspase-12. Fan significantly reduced the release of inflammatory cytokines such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-18, and IL-6. Furthermore, Fan treatment increased superoxide dismutase (SOD) and decreased malondialdehyde (MDA. Notably, Fan inhibited myocardial apoptosis following ER stress in the LPS-induced rat model and stimulated phosphorylation activation of ERK1/2 and NF-κB p65 proteins. Conclusions Fan deficiency alleviated LPS-induced endotoxemia in rats. Therefore, Fan may be a new therapeutic approach for the treatment of cardiac dysfunction.
Collapse
Affiliation(s)
| | - Zonghua Shi
- CCU, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Xinwei Li
- Department of Basic Medicine, Louhe Medical College, Louhe, China
| | - Fengzhou Fu
- Department of Cardiology, Dongguan Songshan Lake Central Hospital, Dongguan, China
| |
Collapse
|
46
|
Euphorbia cuneata Represses LPS-induced Acute Lung Injury in Mice via its Antioxidative and Anti-inflammatory Activities. PLANTS 2020; 9:plants9111620. [PMID: 33233453 PMCID: PMC7700453 DOI: 10.3390/plants9111620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 11/16/2022]
Abstract
Euphorbia cuneata (EC; Euphorbiaceae), which widely grows in Saudi Arabia and Yemen, is used traditionally to treat pain and inflammation. This study aimed to evaluate the protective anti-inflammatory effect of a standardized extract of EC against lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice and the possible underlying mechanism(s) of this pharmacologic activity. ALI was induced in male Balb/c mice using intraperitoneal injection of LPS. A standardized total methanol extract of EC or dexamethasone was administered 5 days prior to LPS challenge. Bronchoalveolar fluid (BALF) and lung samples were collected for analysis. The results demonstrated the protective anti-inflammatory effect of EC against LPS-induced ALI in mice. Standardized EC contained 2R-naringenin-7-O-β-glucoside (1), kaempferol-7-O-β-glucoside (2), cuneatannin (3), quercetin (4), and 2R-naringenin (5) in concentrations of 6.16, 4.80, 51.05, 13.20, and 50.00 mg/g of extract, respectively. EC showed a protective effect against LPS-induced pulmonary damage. EC reduced lung wet/dry weight (W/D) ratio and total protein content in BALF, indicating attenuation of the pulmonary edema. Total and differential cell counts were decreased in EC-treated animals. Histopathological examination confirmed the protective effect of EC, as indicated by an amelioration of LPS-induced lesions in lung tissue. EC also showed a potent anti-oxidative property as it decreased lipid peroxidation and increased the antioxidants in lung tissue. Finally, the anti-inflammatory activity of EC was obvious through its ability to suppress the activation of nuclear factor-κB (NF-κB), and hence its reduction of the levels of downstream inflammatory mediators. In conclusion, these results demonstrate the protective effects of EC against LPS-induced lung injury in mice, which may be due to its antioxidative and anti-inflammatory activities.
Collapse
|
47
|
Zhou F, Liu Z, Cai H, Miao Z, Wei F, Song C. Role of microRNA-15a-5p/TNFAIP3-interacting protein 2 axis in acute lung injury induced by traumatic hemorrhagic shock. Exp Ther Med 2020; 20:2. [PMID: 32934667 PMCID: PMC7471858 DOI: 10.3892/etm.2020.9130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the role of microRNA (miR)-15a-5p in the pathogenesis of acute lung injury induced by traumatic hemorrhagic shock (THS), and to explore the underlying molecular mechanism. The expression level of miR-15a-5p was detected using reverse transcription-quantitative (RT-qPCR) and the association between miR-15a-5p and TNFAIP3-interacting protein 2 (TNIP2) was revealed using TargetScan and dual luciferase reporter assays. To investigate the effect of miR-15a-5p on THS-induced acute lung injury, a THS rat model was established. Lung capillary permeability and lung edema were then determined. Moreover, proinflammatory factors in the bronchoalveolar lavage fluid (BALF) and serum of the THS rat model were detected using ELISA. In addition, protein levels in the current study were measured via western blotting. It was revealed that miR-15a-5p was significantly upregulated in both patients with THS and samples from the THS rat model. TNIP2 represents a direct target of miR-15a-5p, and it was downregulated in both patients with THS and the THS rat model. Further analyses indicated that downregulation of miR-15a-5p significantly relieved acute lung injury induced by THS, evidenced by a decreased ratio of Evan's blue dye (EBD) in the BALF to EBD in plasma of THS rats, decreased lung permeability index and reduced lung wet/dry ratio. Inhibition of miR-15a-5p also decreased THS-induced upregulation of pro-inflammatory factors. Furthermore, the data revealed that THS-induced NF-κB activation in the lung tissues of rats was inhibited by miR-15a-5p knockdown. Moreover, it was demonstrated that all the effects of miR-15a-5p on THS rats were ablated following TNIP2 silencing. Taken together, the data of the current study indicate that miR-15a-5p downregulation serves a protective role in THS-induced acute lung injury via directly targeting TNIP2.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhizhen Liu
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Huazhong Cai
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Zhenjun Miao
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Faxing Wei
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Chao Song
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
48
|
Li C, Gao H, Feng X, Bi C, Zhang J, Yin J. Ginsenoside Rh2 impedes proliferation and migration and induces apoptosis by regulating NF-κB, MAPK, and PI3K/Akt/mTOR signaling pathways in osteosarcoma cells. J Biochem Mol Toxicol 2020; 34:e22597. [PMID: 32762018 DOI: 10.1002/jbt.22597] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/07/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Ginsenoside Rh2 is a primary bioactive compound obtained from ginseng that indicated anticancer activities against several malignant tumors. However, previous studies have reported little about the inhibitory effect of Rh2 on osteosarcoma (OS). This study aims to explore whether Rh2 could exert anticancer effects in OS cells and further investigate the proliferation, migration, and apoptosis mechanisms induced by Rh2 in human OS U20S cell line. The viability of U20S cells was obtained by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay. Cell migration property was analyzed by wound-healing assay. Apoptosis was visualized using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL), 4',6-diamidino-2-phenylindole (DAPI), and annexin V/propidium iodide (PI) staining. Relative protein expressed was confirmed through Western blot analysis. Mitochondrial membrane potential was evaluated by JC-1 staining. In this study, we used broad-spectrum anticancer drug cisplatin (CP) as a positive control. The results indicated that Rh2 remarkably inhibited cell viability of U20S cells in a dose- and time-dependent manner, and suppressed migration. TUNEL, DAPI, annexin V/PI, and JC-1 assay suggested that Rh2 could induce cellular apoptosis. Rh2 could reduce the levels of Bcl-2, caspase 3, and caspase 9, and promote the expression level of Bax in U20S cells. Moreover, Rh2 could induce apoptosis by promoting mitogen-activated protein kinase (MAPK) signaling pathway and inhibit PI3K/Akt/mTOR and nuclear factor-κB (NF-κB) signaling pathway in U20S cells. These findings indicated that Rh2 has an anticancer effect on U20S cells by regulating MAPK, PI3K/Akt/mTOR, and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chenchen Li
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xuemei Feng
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Chuyao Bi
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jing Zhang
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| | - Jianyuan Yin
- Department of Natural Products Chemistry, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
49
|
Wang Y, Shang G, Wang W, Qiu E, Pei Y, Zhang X. Magnoflorine inhibits the malignant phenotypes and increases cisplatin sensitivity of osteosarcoma cells via regulating miR-410-3p/HMGB1/NF-κB pathway. Life Sci 2020; 256:117967. [PMID: 32553931 DOI: 10.1016/j.lfs.2020.117967] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022]
Abstract
AIMS Magnoflorine is an essential type of alkaloid and possesses anti-tumor activity in multiple cancers. Recent studies have demonstrated that magnoflorine plays tumor-suppressive roles in gastric and breast cancers. However, its role in osteosarcoma (OS) tumorigenesis is enigmatic. This study aimed to investigate the role and mechanism of magnoflorine in OS. MATERIALS AND METHODS Two human OS cells (MG-63 and U-2 OS) were treated with different concentrations of magnoflorine. Cell viability and invasion were then detected by Cell Counting Kit-8 and Transwell assay, respectively. And the effects of magnoflorine on the epithelial-mesenchymal transition (EMT) and cisplatin sensitivity were also measured. To explore the potential mechanism, we assayed the influence of magnoflorine on the miR-410-3p/HMGB1/NF-κB signaling pathway. Additionally, rescue experiments were performed to further confirm the regulation mechanism of magnoflorine. KEY FINDINGS Magnoflorine inhibited the viability, invasion, and EMT of OS cells in a dose-dependent manner. And it increased the sensitivity of OS cells to cisplatin. Magnoflorine significantly suppressed HMGB1 expression and NF-κB activation, but upregulated miR-410-3p level. Overexpression of HMGB1 promoted NF-κB activation and reversed the effects of magnoflorine on the viability, invasion, EMT and cisplatin sensitivity of OS cells. miR-410-3p mimic inhibited the EMT of OS cells, which was restored by HMGB1 upregulation. And miR-410-3p inhibitor abrogated the influence of magnoflorine on HMGB1 expression in OS cells. SIGNIFICANCE Magnoflorine inhibited the malignant phenotypes and increased cisplatin sensitivity of OS cells via modulating miR-410-3p/HMGB1/NF-κB pathway. These results indicated that magnoflorine might be a novel drug for the treatment of OS.
Collapse
Affiliation(s)
- Yuming Wang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Guanning Shang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Wei Wang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Enduo Qiu
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Yi Pei
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Xiaojing Zhang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China.
| |
Collapse
|
50
|
Li Z, Qi F, Li F. Identification of drug targets and potential molecular mechanisms for Wantong Jingu Tablet extract in treatment of rheumatoid arthritis: bioinformatics analysis of fibroblast-like synoviocytes. Chin Med 2020; 15:59. [PMID: 32518584 PMCID: PMC7275334 DOI: 10.1186/s13020-020-00339-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/25/2020] [Indexed: 01/20/2023] Open
Abstract
Background Rheumatoid arthritis-fibroblast-like synoviocytes (RA-FLSs) play important roles in pathogenesis of rheumatoid arthritis (RA). Wantong Jingu Tablet (WJT), a mixture of traditional Chinese medicine, is a potentially effective therapy for RA, but its underlying mechanism is unclear. In this study, we explore the effects of WJT on human RA-FLSs and the underlying molecular mechanism. Methods The major components of WJT were determined using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-QTOF/MS). Cell proliferative ability was evaluated by CCK-8, colony formation assay, and EdU incorporation assay. Cell apoptotic capacity was examined by caspase-3 and caspase-9 activity test. Protein levels of Bax and Bcl-2 were investigated by western blotting. High-throughput sequencing and bioinformatics analysis were conducted to screen and identify targeted genes, followed by identification by qRT-PCR and western blotting. Results In this study, we have identified 346 compounds in WJT. Our results showed that WJT inhibited the RA-FLSs proliferation, and promoted apoptosis in a dose- and time-dependent manner. More importantly, 184 differentially expressed genes (DEGs) has been screened after WJT treatment based on DEGSeq2 and 278 DEGs was identified by DEGSeq2 combined with WGCNA. Then, 10 hub genes were identified based on two different analyses, while the expression levels of only SMC3, THOC1, BUB1, and STAG2 were decreased after WJT treatment, which was identical to the sequencing profiles. Conclusions WJT exerted its anti-proliferation and pro-apoptosis effects possibly through suppressing the expression of SMC3, THOC1, BUB1, and STAG2 in RA-FLSs. Thus, therapeutics targeting these genes may be a promising strategy for rescuing RA.
Collapse
Affiliation(s)
- Zhaodong Li
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China
| | - Fangyuan Qi
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China
| | - Fan Li
- Department of Pathogen Biology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, No. 126 Xinmin Street, Changchun, 130021 Jilin China.,The Key Laboratory for Bionics Engineering, Ministry of Education, China, Jilin University, Changchun, 130021 Jilin China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, 130021 Jilin China.,Key Laboratory for Biomedical Materials of Jilin Province, Jilin University, Changchun, 130021 Jilin China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang China
| |
Collapse
|