1
|
Zhao G, Xie Y, Lei X, Guo R, Cui N. mTOR aggravated CD4 + T cell pyroptosis by regulating the PPARγ-Nrf2 pathway in sepsis. Int Immunopharmacol 2024; 140:112822. [PMID: 39096877 DOI: 10.1016/j.intimp.2024.112822] [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: 05/05/2024] [Revised: 07/15/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome caused by a dysregulated host response to infection. CD4+T cell reduction is crucial to sepsis-induced immunosuppression. Pyroptosis, a programmed necrosis, is concerned with lymphocytopenia. Peroxisome proliferator-activated receptor gamma (PPARγ) regulated by upstream mTOR, exerts anti-pyroptosis effects. To investigate the potential effects of mTOR-PPARγ on sepsis-induced CD4+T cell depletion and the underlying mechanisms, we observed mTOR activation and pyroptosis with PPARγ-Nrf suppression through cecal ligation and puncture (CLP) sepsis mouse model. Further mechanism research used genetically modified mice with T cell-specific knockout mTOR or Tuberous Sclerosis Complex1 (TSC1). It revealed that mTOR mediated CD4 + T cell pyroptosis in septic mice by negatively regulating the PPARγ-Nrf2 signaling pathway. Taken together, mTOR-PPARγ-Nrf2 signaling mediated the CD4+ T cell pyroptosis in sepsis, contributing to CD4+T cell depletion and immunosuppression.
Collapse
Affiliation(s)
- Guoyu Zhao
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yawen Xie
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Xianli Lei
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Ran Guo
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Na Cui
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
2
|
Dey AD, Mannan A, Dhiman S, Singh TG. Unlocking new avenues for neuropsychiatric disease therapy: the emerging potential of Peroxisome proliferator-activated receptors as promising therapeutic targets. Psychopharmacology (Berl) 2024; 241:1491-1516. [PMID: 38801530 DOI: 10.1007/s00213-024-06617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
RATIONALE Peroxisome proliferator-activated receptors (PPARs) are transcription factors that regulate various physiological processes such as inflammation, lipid metabolism, and glucose homeostasis. Recent studies suggest that targeting PPARs could be beneficial in treating neuropsychiatric disorders by modulating neuronal function and signaling pathways in the brain. PPAR-α, PPAR-δ, and PPAR-γ have been found to play important roles in cognitive function, neuroinflammation, and neuroprotection. Dysregulation of PPARs has been associated with neuropsychiatric disorders like bipolar disorder, schizophrenia, major depression disorder, and autism spectrum disorder. The limitations and side effects of current treatments have prompted research to target PPARs as a promising novel therapeutic strategy. Preclinical and clinical studies have shown the potential of PPAR agonists and antagonists to improve symptoms associated with these disorders. OBJECTIVE This review aims to provide an overview of the current understanding of PPARs in neuropsychiatric disorders, their potential as therapeutic targets, and the challenges and future directions for developing PPAR-based therapies. METHODS An extensive literature review of various search engines like PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out with the keywords "PPAR, Neuropsychiatric disorders, Oxidative stress, Inflammation, Bipolar Disorder, Schizophrenia, Major depression disorder, Autism spectrum disorder, molecular pathway". RESULT & CONCLUSION Although PPARs present a hopeful direction for innovative therapeutic approaches in neuropsychiatric conditions, additional research is required to address obstacles and convert this potential into clinically viable and individualized treatments.
Collapse
Affiliation(s)
- Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | | |
Collapse
|
3
|
Cui C, Fan Z. Exploring the mechanism of Liang Xue Wu Hua Tang in the treatment of rosacea via network pharmacology and molecular docking. Medicine (Baltimore) 2024; 103:e38705. [PMID: 38941423 PMCID: PMC11466150 DOI: 10.1097/md.0000000000038705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/05/2024] [Indexed: 06/30/2024] Open
Abstract
Rosacea is a chronic and recurrent inflammatory skin disease affecting the center of the face that causes burning and itching sensations and changes in aesthetics. Liang Xue Wu Hua Tang (LXWHT) is a classic herbal formulation that is efficacious and has been widely used in the clinical treatment of rosacea; however, the pharmacological mechanisms remain unclear. The aim of the present study was to investigate the mechanism of action of LXWHT using network pharmacology and molecular docking. The Traditional Chinese Medicine System Pharmacology database was searched to identify the active ingredients and pharmacological targets of LXWHT, and the GeneCard, Disgenet, and Gene Expression Omnibus databases were applied to screen rosacea-related targets. Cytoscape software was used to visualize the protein-protein interaction network, and network topology analysis was used to identify core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed for the core targets. Molecular docking simulations and visualization were performed using Maestro and PyMOL, respectively. A total of 43 active compounds and 28 potential targets for LXWHT treatment of rosacea were selected for analysis. The Gene Ontology/Kyoto Encyclopedia of Genes and Genomes results indicated that LXWHT may exert therapeutic effects on rosacea by intervening in immune pathways including tumor necrosis factor pathway, interleukin-17 pathways, and Toll-like receptor signaling pathways. Chemokine ligand 2, interferon-γ, interleukin-1ß, peroxisome proliferator-activated receptor-γ, and matrix metallopeptidase 9 may be the core therapeutic target. Quercetin, stigmasterol, kaempferol, beta-sitosterol, luteolin, beta-carotene, baicalein, acetin, and isorhamnetin were predicted to be the key active ingredients. LXWHT may exert therapeutic effects in the treatment of rosacea by modulating immunity and angiogenesis, laying the foundation for further research.
Collapse
Affiliation(s)
- Can Cui
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhu Fan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Qin P, Li Y, Su Y, Wang Z, Wu R, Liang X, Zeng Y, Guo P, Yu Z, Huang X, Yang H, Zeng Z, Zhao X, Gong S, Han J, Chen Z, Xiao W, Chen A. Bifidobacterium adolescentis-derived hypaphorine alleviates acetaminophen hepatotoxicity by promoting hepatic Cry1 expression. J Transl Med 2024; 22:525. [PMID: 38822329 PMCID: PMC11143572 DOI: 10.1186/s12967-024-05312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/16/2024] [Indexed: 06/02/2024] Open
Abstract
Acetaminophen (APAP)-induced liver injury (AILI) is a pressing public health concern. Although evidence suggests that Bifidobacterium adolescentis (B. adolescentis) can be used to treat liver disease, it is unclear if it can prevent AILI. In this report, we prove that B. adolescentis significantly attenuated AILI in mice, as demonstrated through biochemical analysis, histopathology, and enzyme-linked immunosorbent assays. Based on untargeted metabolomics and in vitro cultures, we found that B. adolescentis generates microbial metabolite hypaphorine. Functionally, hypaphorine inhibits the inflammatory response and hepatic oxidative stress to alleviate AILI in mice. Transcriptomic analysis indicates that Cry1 expression is increased in APAP-treated mice after hypaphorine treatment. Overexpression of Cry1 by its stabilizer KL001 effectively mitigates liver damage arising from oxidative stress in APAP-treated mice. Using the gene expression omnibus (GEO) database, we verified that Cry1 gene expression was also decreased in patients with APAP-induced acute liver failure. In conclusion, this study demonstrates that B. adolescentis inhibits APAP-induced liver injury by generating hypaphorine, which subsequently upregulates Cry1 to decrease inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ping Qin
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yanru Li
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Nursing, Southern Medical University, Guangzhou, 510515, China
| | - Yangjing Su
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ze Wang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Rong Wu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoqi Liang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yunong Zeng
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Peiheng Guo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhichao Yu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xintao Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Hong Yang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Shenhai Gong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jiaochan Han
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Ali Chen
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
5
|
Liu CJ, Li HX, Chen ZH, Li JJ, Shi W, Zhang FX. A review of the ethnopharmacology, phytochemistry, pharmacology, pharmacokinetics and toxicology of Abri Herba (Ji-Gu-Cao). PHYTOCHEMISTRY 2024; 221:114064. [PMID: 38508326 DOI: 10.1016/j.phytochem.2024.114064] [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: 09/20/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/22/2024]
Abstract
Abri Herba (AH, known as 'Ji-Gu-Cao' in China) has a long-term medicinal history of treating cholecystitis, acute and chronic hepatitis and non-alcoholic fatty liver (NAFL) in China or other Asian countries. This review aimed to provide a comprehensive analysis of AH in terms of ethnopharmacology, phytochemistry, pharmacology, pharmacokinetics and toxicology. The information involved in the study was collected from a variety of electronic resources, and >100 scientific studies have been used since 1962. Until now, 95 chemical compounds have been isolated and identified from AH and the seeds of Abrus cantoniensis Hance (ACH), including 47 terpenoids, 26 flavonoids and 4 alkaloids. The pharmacological activities of AH extracts and their pure compounds have been explored in the aspects of anti-hyperlipidaemia, hepatoprotection, anti-tumour, anti-viral, anti-bacterial, anti-inflammatory and analgesic, immunomodulation, antioxidant and others. The pharmacokinetics and excretion kinetics of AH in vivo and 15 traditional and clinical prescriptions containing AH have been sorted out, and the potential therapeutic mechanism and drug metabolism pattern were also summarised. The pods of ACH are toxic, with a median lethal dose (LD50) of 10.01 ± 2.90 g/kg (i.g.) in mice. Interestingly, the toxicity of ACH's pods and seeds decreased after boiling. However, the toxicity mechanism of pods of ACH is unclear, limiting its clinical application. Clinical trials in the future should be used to explore its safety. Meanwhile, as one of the relevant pharmacological activities, the effects and mechanism of AH on anti-hyperlipidaemia and hepatoprotection should be further studied, which is of great significance for understanding its mechanism of action in the treatment of NAFL disease and improving its clinical application.
Collapse
Affiliation(s)
- Cheng-Jun Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Hong-Xin Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Zi-Hao Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Jin-Jin Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Wei Shi
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Feng-Xiang Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| |
Collapse
|
6
|
Song X, Liu G, Bin Y, Bai R, Liang B, Yang H. C1q/Tumor Necrosis Factor-Related Protein-9 Enhances Macrophage Cholesterol Efflux and Improves Reverse Cholesterol Transport via AMPK Activation. Biochem Genet 2024:10.1007/s10528-024-10761-1. [PMID: 38600398 DOI: 10.1007/s10528-024-10761-1] [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: 05/02/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
Cholesterol efflux from foam cells in atherosclerotic plaques is crucial for reverse cholesterol transport (RCT), an important antiatherogenic event. ATP-binding cassette (ABC) transporters, ABCA1 and ABCG1, are key receptors in the cholesterol efflux pathway. C1q/tumor necrosis factor-related protein-9 (CTRP9) is a newly discovered adipokine and exhibits an atheroprotective activity. However, the role of CTRP9 in RCT still remains unknown. In this work, we investigated the effect of subcutaneous administration of CTRP9 protein on RCT and atherosclerotic lesion formation in ApoE-/- mice fed with a high-fat diet. CTRP9-dependent regulation of cholesterol efflux and ABC transporters in RAW 264.7 foam cells was determined. Our results showed that CTRP9 protein decreased atherosclerotic lesions, increased cholesterol efflux, and upregulated liver ABCA1 and ABCG1 expression in ApoE-/- mice. CTRP9 treatment dose-dependently increased mRNA and protein expression of ABCA1, ABCG1, and LXR-α in RAW 264.7 foam cells. Moreover, the expression and phosphorylation of AMPK was potentiated upon CTRP9 treatment. Notably, CTRP9-induced cholesterol efflux and upregulation of ABCA, ABCG1, and LXR-α were impaired when AMPK was knocked down. AMPK depletion restored cholesterol accumulation in CTRP9-treated RAW 264.7 cells. Taken together, subcutaneous injection is an effective novel delivery route for CTRP9 protein, and exogenous CTRP9 can facilitate cholesterol efflux and promote RCT in an animal model of atherosclerosis. The atheroprotective activity of CTRP9 is mediated through the activation of AMPK signaling.
Collapse
Affiliation(s)
- Xiaosu Song
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Gaizhen Liu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Yunfei Bin
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Rui Bai
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China
| | - Huiyu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, China.
| |
Collapse
|
7
|
Liu S, Cao C, Wang Y, Hu L, Liu Q. Novel Therapies for ANCA-associated Vasculitis: Apilimod Ameliorated Endothelial Cells Injury through TLR4/NF-κB Pathway and NLRP3 Inflammasome. Curr Pharm Des 2024; 30:2325-2344. [PMID: 38910483 DOI: 10.2174/0113816128312530240607051608] [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: 03/12/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Antineutrophil cytoplasmic antibody-associated vasculitis (AAV) is a rapidly progressive form of glomerulonephritis for which effective therapeutic drugs are currently lacking, and its underlying mechanism remains unclear. AIMS This study aimed to investigate new treatment options for AAV through a combination of bioinformatics analysis and cell molecular experiments. METHODS The research utilized integrated bioinformatics analysis to identify genes with differential expression, conduct enrichment analysis, and pinpoint hub genes associated with AAV. Potential therapeutic compounds for AAV were identified using Connectivity Map and molecular docking techniques. In vitro experiments were then carried out to examine the impact and mechanism of apilimod on endothelial cell injury induced by MPO-ANCA-positive IgG. RESULTS The findings revealed a set of 374 common genes from differentially expressed genes and key modules of WGCNA, which were notably enriched in immune and inflammatory response processes. A proteinprotein interaction network was established, leading to the identification of 10 hub genes, including TYROBP, PTPRC, ITGAM, KIF20A, CD86, CCL20, GAD1, LILRB2, CD8A, and COL5A2. Analysis from Connectivity Map and molecular docking suggested that apilimod could serve as a potential therapeutic cytokine inhibitor for ANCA-GN based on the hub genes. In vitro experiments demonstrated that apilimod could mitigate tight junction disruption, endothelial cell permeability, LDH release, and endothelial activation induced by MPO-ANCA-positive IgG. Additionally, apilimod treatment led to a significant reduction in the expression of proteins involved in the TLR4/NF-κB and NLRP3 inflammasome-mediated pyroptosis pathways. CONCLUSION This study sheds light on the potential pathogenesis of AAV and highlights the protective role of apilimod in mitigating MPO-ANCA-IgG-induced vascular endothelial cell injury by modulating the TLR4/ NF-kB and NLRP3 inflammasome-mediated pyroptosis pathway. These findings suggest that apilimod may hold promise as a treatment for AAV and warrant further investigation.
Collapse
Affiliation(s)
- Siyang Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenlin Cao
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of the Second Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiru Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liu Hu
- Department of Health Management Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingquan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Mao S, Yao J, Zhang T, Zhang X, Tan W, Li C. Bilobalide attenuates lipopolysaccharide‑induced HepG2 cell injury by inhibiting TLR4‑NF‑κB signaling via the PI3K/Akt pathway. Exp Ther Med 2024; 27:24. [PMID: 38125341 PMCID: PMC10728898 DOI: 10.3892/etm.2023.12312] [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: 01/22/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2023] Open
Abstract
Inflammation is involved in the pathological process underlying a number of liver diseases. Bilobalide (BB) is a natural compound from Ginkgo biloba leaves that was recently demonstrated to exert hepatoprotective effects by inhibiting oxidative stress in the liver cancer cell line HepG2. The anti-inflammatory activity of BB has been reported in recent studies. The major objective of the present study was to investigate whether BB could attenuate inflammation-associated cell damage. HepG2 cells were cultured with lipopolysaccharide (LPS) and BB, and cell damage was evaluated by measuring cell viability using MTT assay. The activity of the NF-κB signaling pathway was assessed by measuring the levels of IκBα, NF-κB p65, phosphorylated (p)-IκBα, p-p65, p65 DNA-binding activity and inflammatory cytokines IL-1β, IL-6 and TNF-α. A toll-like receptor (TLR)4 inhibitor (CLI-095) was used to detect the involvement of TLR4 in cell injury caused by LPS. In addition, the PI3K/Akt inhibitor LY294002 was applied to explore the involvement of the PI3K/Akt axis in mediating the effects of BB. The results demonstrated that LPS induced HepG2 cell injury. LPS also elevated the levels of p-IκBα, p-p65, p65 DNA-binding activity and inflammatory cytokines. However, CLI-095 significantly attenuated the LPS-induced cell damage and inhibited the activation of NF-κB signaling. BB also dose-dependently attenuated the LPS-induced cell damage, activation of NF-κB signaling and TLR4 overexpression. Furthermore, it was observed that LY294002 diminished the cytoprotective effects of BB on cell injury, TLR4 expression and NF-κB activation. These findings indicated that BB could attenuate LPS-induced inflammatory injury to HepG2 cells by regulating TLR4-NF-κB signaling.
Collapse
Affiliation(s)
- Shumei Mao
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jinpeng Yao
- Department of Cardiology, Yantai Kaifaqu Hospital, Yantai, Shandong 264006, P.R. China
| | - Teng Zhang
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiang Zhang
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Wei Tan
- Department of Respiratory Medicine, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Chengde Li
- Department of Clinical Pharmacy, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
9
|
Zhang Y, Huang J, Gan L, Wu R, Jin J, Wang T, Sun S, Zhang Z, Li L, Zheng X, Zhang K, Sun L, Ma H, Li D. Hepatoprotective effects of Niudali ( Callerya speciosa) root aqueous extracts against tetrachloromethane-induced acute liver injury and inflammation. Food Sci Nutr 2023; 11:7026-7038. [PMID: 37970412 PMCID: PMC10630805 DOI: 10.1002/fsn3.3626] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 11/17/2023] Open
Abstract
Niudali (Callerya speciosa) is commonly grown in southeastern regions of China and consumed as a food ingredient. Although Niudali root extracts showed various biological activities, the hepatoprotective effects of Niudali root phytochemicals are not fully studied. Herein, we prepared two Niudali root aqueous extracts, namely, c and Niudali polysaccharides-enriched extract (NPE), and identified an alkaloid, (hypaphorine) in NEW. The hepatoprotective effects of NWE, NPE, and hypaphorine were evaluated in an acute liver injury model induced by carbon tetrachloride (CCl4) in mice. Pathohistological examination and blood chemistry assays showed that treatment of NWE, NPE, and hypaphorine alleviated CCl4-induced liver damage by lowering the liver injury score (by 75.51%, 80.01%, and 41.22%) and serum aspartate and alanine transaminases level (by 63.24%, 85.22%, and 49.74% and by 78.73%, 80.08%, and 81.70%), respectively. NWE, NPE, and hypaphorine also reduced CCl4-induced hepatic oxidative stresses in the liver tissue by decreasing the levels of malondialdehyde (by 40.00%, 51.25%, and 28.75%) and reactive oxygen species (by 30.22%, 36.14%, and 33.54%) while increasing the levels of antioxidant enzymes including superoxide dismutase (by 21.36%, 21.64%, and 8.90%), catalase (by 22.13%, 33.33%, and 5.39%), and glutathione (by 84.87%, 90.65%, and 80.53%), respectively. Mechanistic assays showed that NWE, NPE, and hypaphorine alleviated liver damage by mediating inflammatory biomarkers (e.g., pro-inflammatory cytokines) via the signaling pathways of mitogen-activated protein kinases and nuclear factor-κB. Findings from our study extend the understanding of Niudali's hepatoprotective effects, which is useful for its development as a dietary intervention for liver inflammation.
Collapse
Affiliation(s)
- Yizi Zhang
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
| | - Jinwen Huang
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
| | - Lishe Gan
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
- International Healthcare Innovation Institute (Jiangmen)JiangmenChina
| | - Rihui Wu
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
- International Healthcare Innovation Institute (Jiangmen)JiangmenChina
| | - Jingwei Jin
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
- International Healthcare Innovation Institute (Jiangmen)JiangmenChina
| | - Tinghan Wang
- Bioactive Botanical Research Laboratory, Biomedical and Pharmaceutical Sciences, College of PharmacyUniversity of Rhode IslandKingstonRhode IslandUSA
| | - Shili Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & UtilizationGuangzhouChina
| | - Zhenbiao Zhang
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & UtilizationGuangzhouChina
| | - Liya Li
- Institute of Microbial Pharmaceuticals, College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Xi Zheng
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
| | - Kun Zhang
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
| | - Lingli Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Key Laboratory of Tea Resources Innovation & UtilizationGuangzhouChina
| | - Hang Ma
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
- International Healthcare Innovation Institute (Jiangmen)JiangmenChina
- Bioactive Botanical Research Laboratory, Biomedical and Pharmaceutical Sciences, College of PharmacyUniversity of Rhode IslandKingstonRhode IslandUSA
| | - Dongli Li
- School of Biotechnology and Health SciencesWuyi UniversityJiangmenChina
- International Healthcare Innovation Institute (Jiangmen)JiangmenChina
| |
Collapse
|
10
|
Luan J, Ji X, Liu L. PPARγ in Atherosclerotic Endothelial Dysfunction: Regulatory Compounds and PTMs. Int J Mol Sci 2023; 24:14494. [PMID: 37833942 PMCID: PMC10572723 DOI: 10.3390/ijms241914494] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
The formation of atherosclerotic plaques is one of the main sources of cardiovascular disease. In addition to known risk factors such as dyslipidemia, diabetes, obesity, and hypertension, endothelial dysfunction has been shown to play a key role in the formation and progression of atherosclerosis. Peroxisome proliferator-activated receptor-gamma (PPARγ), a transcription factor belonging to the steroid superfamily, is expressed in the aorta and plays a critical role in protecting endothelial function. It thereby serves as a target for treating both diabetes and atherosclerosis. Although many studies have examined endothelial cell disorders in atherosclerosis, the role of PPARγ in endothelial dysfunction is still not well understood. In this review, we summarize the possible mechanisms of action behind PPARγ regulatory compounds and post-translational modifications (PTMs) of PPARγ in the control of endothelial function. We also explore the potential use of endothelial PPARγ-targeted agents in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
| | | | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200082, China
| |
Collapse
|
11
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
12
|
Liu CH, Jheng PR, Rethi L, Godugu C, Lee CY, Chen YT, Nguyen HT, Chuang EY. P-Selectin mediates targeting of a self-assembling phototherapeutic nanovehicle enclosing dipyridamole for managing thromboses. J Nanobiotechnology 2023; 21:260. [PMID: 37553670 PMCID: PMC10408148 DOI: 10.1186/s12951-023-02018-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/23/2023] [Indexed: 08/10/2023] Open
Abstract
Thrombotic vascular disorders, specifically thromboembolisms, have a significant detrimental effect on public health. Despite the numerous thrombolytic and antithrombotic drugs available, their efficacy in penetrating thrombus formations is limited, and they carry a high risk of promoting bleeding. Consequently, the current medication dosage protocols are inadequate for preventing thrombus formation, and higher doses are necessary to achieve sufficient prevention. By integrating phototherapy with antithrombotic therapy, this study addresses difficulties related to thrombus-targeted drug delivery. We developed self-assembling nanoparticles (NPs) through the optimization of a co-assembly engineering process. These NPs, called DIP-FU-PPy NPs, consist of polypyrrole (PPy), dipyridamole (DIP), and P-selectin-targeted fucoidan (FU) and are designed to be delivered directly to thrombi. DIP-FU-PPy NPs are proposed to offer various potentials, encompassing drug-loading capability, targeted accumulation in thrombus sites, near-infrared (NIR) photothermal-enhanced thrombus management with therapeutic efficacy, and prevention of rethrombosis. As predicted, DIP-FU-PPy NPs prevented thrombus recurrence and emitted visible fluorescence signals during thrombus clot penetration with no adverse effects. Our co-delivery nano-platform is a simple and versatile solution for NIR-phototherapeutic multimodal thrombus control.
Collapse
Affiliation(s)
- Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 11031, Taiwan
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering Graduate Institute of Biomedical Optomechatronics, Research Center of Biomedical Device, Innovation Entrepreneurship Education Center, College of Interdisciplinary Studies, Taipei Medical University, Taipei, 11031, Taiwan
| | - Lekha Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering Graduate Institute of Biomedical Optomechatronics, Research Center of Biomedical Device, Innovation Entrepreneurship Education Center, College of Interdisciplinary Studies, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chandraiah Godugu
- National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad, Hyderabad, India
| | - Ching Yi Lee
- Department of Neurosurgery, Chang Gung Memorial Hospital Linkou Main Branch and School of Medicine, College of Medicine, Chang Gung University, Taoyuan, 33305, Taiwan
| | - Yan-Ting Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering Graduate Institute of Biomedical Optomechatronics, Research Center of Biomedical Device, Innovation Entrepreneurship Education Center, College of Interdisciplinary Studies, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hieu Trung Nguyen
- Department of Orthopedics and Trauma, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh City, 700000, Viet Nam
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering Graduate Institute of Biomedical Optomechatronics, Research Center of Biomedical Device, Innovation Entrepreneurship Education Center, College of Interdisciplinary Studies, Taipei Medical University, Taipei, 11031, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
- Precision Medicine and Translational Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
| |
Collapse
|
13
|
Cheng T, Wang H, Hu Y. The causal effects of genetically determined human blood metabolites on the risk of atrial fibrillation. Front Cardiovasc Med 2023; 10:1211458. [PMID: 37564907 PMCID: PMC10410273 DOI: 10.3389/fcvm.2023.1211458] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/14/2023] [Indexed: 08/12/2023] Open
Abstract
Background Blood metabolites have been found related to atrial fibrillation (AF), but the causal role is still unclear. Mendel randomization (MR) can give information about the causality between blood metabolites and AF. Methods Two-sample MR analysis was used to evaluate the causality between 486 blood metabolites and AF. Firstly, the genome-wide association study (GWAS) data for AF (from Nielsen et al.) was analyzed and some metabolites were identified. Then another GWAS data for AF (from Roselli et al.) was repeatedly analyzed to verify the results. Inverse variance weighted method was mainly used to determine the causality, and MR-egger, Weighted Median, and MR-PRESSO models were used as supplements of MR. Cochran's Q test was used to assess heterogeneity. And MR-Egger intercept and MR-PRESSO global test were performed to measure pleiotropy. Results The study used Bonferroni's corrected P value (P < 1.03 × 10-4) as the significance threshold. After MR analysis and replication analysis, we found two overlapped metabolites. Among which tryptophan betaine was the most significant causal metabolite in both AF GWAS data (from Nielsen et al.) (odds ratio (OR) = 0.83, 95% confidence interval (CI) = 0.76-0.90, P = 9.37 × 10-6) and AF GWAS data (from Roselli et al.) (OR = 0.82, 95% CI = 0.76-0.88, P = 2.00 × 10-7), while uridine was nominally significant metabolites in both AF GWAS data (from Nielsen et al.) (OR = 0.58, 95% CI = 0.40-0.84, P = 0.004) and AF GWAS data (from Roselli et al.) (OR = 0.56, 95% CI = 0.35-0.88, P = 0.01). And the results of sensitivity analysis showed that none of them had obvious heterogeneity or pleiotropy. Conclusion The study identified several blood metabolites that were causally related to AF, which may provide new perspectives on the pathogenesis of AF.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Beijing University of ChineseMedicine, Beijing, China
| | - Huan Wang
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
14
|
Kim H, Appel LJ, Lichtenstein AH, Wong KE, Chatterjee N, Rhee EP, Rebholz CM. Metabolomic Profiles Associated With Blood Pressure Reduction in Response to the DASH and DASH-Sodium Dietary Interventions. Hypertension 2023; 80:1494-1506. [PMID: 37161796 PMCID: PMC10262995 DOI: 10.1161/hypertensionaha.123.20901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/10/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND The DASH (Dietary Approaches to Stop Hypertension) diets reduced blood pressure (BP) in the DASH and DASH-Sodium trials, but the underlying mechanisms are unclear. We identified metabolites associated with systolic BP or diastolic BP (DBP) changes induced by dietary interventions (DASH versus control arms) in 2 randomized controlled feeding studies-the DASH and DASH-Sodium trials. METHODS Metabolomic profiling was conducted in serum and urine samples collected at the end of diet interventions: DASH (n=219) and DASH-Sodium (n=395). Using multivariable linear regression models, associations were examined between metabolites and change in systolic BP and DBP. Tested for interactions between diet interventions and metabolites were the following comparisons: (1) DASH versus control diets in the DASH trial (serum), (2) DASH high-sodium versus control high-sodium diets in the DASH-Sodium trial (urine), and (3) DASH low-sodium versus control high-sodium diets in the DASH-Sodium trial (urine). RESULTS Sixty-five significant interactions were identified (DASH trial [serum], 12; DASH high sodium [urine], 35; DASH low sodium [urine], 18) between metabolites and systolic BP or DBP. In the DASH trial, serum tryptophan betaine was associated with reductions in DBP in participants consuming the DASH diets but not control diets (P interaction, 0.023). In the DASH-Sodium trial, urine levels of N-methylglutamate and proline derivatives (eg, stachydrine, 3-hydroxystachydrine, N-methylproline, and N-methylhydroxyproline) were associated with reductions in systolic BP or DBP in participants consuming the DASH diets but not control diets (P interaction, <0.05 for all tests). CONCLUSIONS We identified metabolites that were associated with BP lowering in response to dietary interventions. REGISTRATION URL: https://www. CLINICALTRIALS gov/ct2/show/NCT03403166; Unique identifier: NCT03403166 (DASH trial). URL: https://www. CLINICALTRIALS gov/ct2/show/NCT00000608; Unique identifier: NCT00000608 (DASH-Sodium trial).
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Epidemiology (H.K., L.J.A., C.M.R.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD (H.K., L.J.A., C.M.R.)
| | - Lawrence J. Appel
- Department of Epidemiology (H.K., L.J.A., C.M.R.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD (L.J.A., C.M.R.)
| | - Alice H. Lichtenstein
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD (H.K., L.J.A., C.M.R.)
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA (A.H.L.)
| | - Kari E. Wong
- Metabolon, Research Triangle Park, Morrisville, NC (K.E.W.)
| | - Nilanjan Chatterjee
- Department of Biostatistics (N.C.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, MA (E.P.R.)
| | - Casey M. Rebholz
- Department of Epidemiology (H.K., L.J.A., C.M.R.), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD (H.K., L.J.A., C.M.R.)
| |
Collapse
|
15
|
Ivanov IA, Siniavin AE, Palikov VA, Senko DA, Shelukhina IV, Epifanova LA, Ojomoko LO, Belukhina SY, Prokopev NA, Landau MA, Palikova YA, Kazakov VA, Borozdina NA, Bervinova AV, Dyachenko IA, Kasheverov IE, Tsetlin VI, Kudryavtsev DS. Analogs of 6-Bromohypaphorine with Increased Agonist Potency for α7 Nicotinic Receptor as Anti-Inflammatory Analgesic Agents. Mar Drugs 2023; 21:368. [PMID: 37367693 DOI: 10.3390/md21060368] [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: 04/20/2023] [Revised: 06/03/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Hypaphorines, tryptophan derivatives, have anti-inflammatory activity, but their mechanism of action was largely unknown. Marine alkaloid L-6-bromohypaphorine with EC50 of 80 μM acts as an agonist of α7 nicotinic acetylcholine receptor (nAChR) involved in anti-inflammatory regulation. We designed the 6-substituted hypaphorine analogs with increased potency using virtual screening of their binding to the α7 nAChR molecular model. Fourteen designed analogs were synthesized and tested in vitro by calcium fluorescence assay on the α7 nAChR expressed in neuro 2a cells, methoxy ester of D-6-iodohypaphorine (6ID) showing the highest potency (EC50 610 nM), being almost inactive toward α9α10 nAChR. The macrophages cytometry revealed an anti-inflammatory activity, decreasing the expression of TLR4 and increasing CD86, similarly to the action of PNU282987, a selective α7 nAChR agonist. 6ID administration in doses 0.1 and 0.5 mg/kg decreased carrageenan-induced allodynia and hyperalgesia in rodents, in accord with its anti-inflammatory action. Methoxy ester of D-6-nitrohypaphorine demonstrated anti-oedemic and analgesic effects in arthritis rat model at i.p. doses 0.05-0.26 mg/kg. Tested compounds showed excellent tolerability with no acute in vivo toxicity in dosages up to 100 mg/kg i.p. Thus, combining molecular modelling and natural product-inspired drug design improved the desired activity of the chosen nAChR ligand.
Collapse
Affiliation(s)
- Igor A Ivanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Andrei E Siniavin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ivanovsky Institute of Virology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Victor A Palikov
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Dmitry A Senko
- Center Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Irina V Shelukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Lyubov A Epifanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Lucy O Ojomoko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Svetlana Y Belukhina
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
| | - Nikita A Prokopev
- Department of Biology, M.V. Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Mariia A Landau
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Yulia A Palikova
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Vitaly A Kazakov
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Natalia A Borozdina
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Arina V Bervinova
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Igor A Dyachenko
- The Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 142290 Pushchino, Russia
| | - Igor E Kasheverov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Denis S Kudryavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| |
Collapse
|
16
|
PPARs and the Kynurenine Pathway in Melanoma-Potential Biological Interactions. Int J Mol Sci 2023; 24:ijms24043114. [PMID: 36834531 PMCID: PMC9960262 DOI: 10.3390/ijms24043114] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors involved in various physiological and pathological processes within the skin. PPARs regulate several processes in one of the most aggressive skin cancers, melanoma, including proliferation, cell cycle, metabolic homeostasis, cell death, and metastasis. In this review, we focused not only on the biological activity of PPAR isoforms in melanoma initiation, progression, and metastasis but also on potential biological interactions between the PPAR signaling and the kynurenine pathways. The kynurenine pathway is a major pathway of tryptophan metabolism leading to nicotinamide adenine dinucleotide (NAD+) production. Importantly, various tryptophan metabolites exert biological activity toward cancer cells, including melanoma. Previous studies confirmed the functional relationship between PPAR and the kynurenine pathway in skeletal muscles. Despite the fact this interaction has not been reported in melanoma to date, some bioinformatics data and biological activity of PPAR ligands and tryptophan metabolites may suggest a potential involvement of these metabolic and signaling pathways in melanoma initiation, progression, and metastasis. Importantly, the possible relationship between the PPAR signaling pathway and the kynurenine pathway may relate not only to the direct biological effect on melanoma cells but also to the tumor microenvironment and the immune system.
Collapse
|
17
|
Tan Z, Chen S, Zhang M, Qu X, Li T, Zhang A, He Y, Ou M, Long L, Chen L, Wu F. An ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry identification and characterization of the active constituents from Abrus mollis Hance. J Sep Sci 2023; 46:e2200311. [PMID: 36349515 DOI: 10.1002/jssc.202200311] [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: 04/19/2022] [Revised: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Abstract
Abrus mollis Hance is a traditional Chinese medicine that is widely used to treat acute and chronic hepatitis, steatosis, and fibrosis. Its therapeutic qualities of it have long been acknowledged, although the active ingredients responsible for its efficacy and the mechanisms of its action are unknown. In this study, the chemical constituents absorbed into the blood from Abrus mollis Hance were assessed by using liquid chromatography-quadrupole-time-of-flight mass spectrometry and the data was analyzed with the UNIFI screening platform. The results obtained were compared to existing chromatographic-mass spectrometry information, including retention times and molecular weights as well as known reference compounds. 41 chemical constituents were found in Abrus mollis Hance, and these included 16 flavonoids, 13 triterpenoids, five organic acids, and two alkaloids. Experimentally it was found that Abrus mollis Hance had a therapeutic benefit when treating α-naphthalene isothiocyanate-induced acute liver injury in rats. In addition, 11 blood prototypical constituents, including six flavonoids, three triterpenoids, and two alkaloids, were found in serum samples following intragastric administration of Abrus mollis Hance extracts to rats. This novel study can be used for the quality control and pharmacodynamic assessment of Abrus mollis Hance in order to assess its efficacy in the therapeutic treatment of patients.
Collapse
Affiliation(s)
- Zhien Tan
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Shimin Chen
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China.,Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, P. R. China
| | - Mengli Zhang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Xiaosheng Qu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Taiping Li
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Aihua Zhang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Yanmei He
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Chinmedomics Research Center of State Administration of TCM, Laboratory of Metabolomics, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Min Ou
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Lihuo Long
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Lu Chen
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| | - Fangfang Wu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plants, Nanning, P. R. China
| |
Collapse
|
18
|
Hou B, Wen Y, Zhu X, Qi M, Cai W, Du B, Sun H, Qiu L. Preparation and characterization of vaccarin, hypaphorine and chitosan nanoparticles and their promoting effects on chronic wounds healing. Int J Biol Macromol 2022; 221:1580-1592. [PMID: 35961560 DOI: 10.1016/j.ijbiomac.2022.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/25/2022] [Accepted: 08/07/2022] [Indexed: 11/18/2022]
Abstract
Chronic wounds have become an important factor hindering human health, affecting tens of millions of people worldwide, especially diabetic wounds. Based on the antibacterial properties of chitosan, the angiogenesis promoting effect of vaccarin (VAC) and the anti-inflammatory effect of hypaphorine (HYP), nanoparticles with high bioavailability were prepared. VAC, HYP and chitosan nanoparticles (VAC + HYP-NPS) were used to the treatment of chronic wounds. Transmission electron microscopy (TEM) images showed the nanoparticles were spherical. ZetaPALS showed the potential of nanoparticles were -12.8 ± 5.53 mV and the size were 166.8 ± 29.95 nm. Methyl thiazolyl tetrazolium (MTT) assay showed that VAC + HYP-NPS had no toxicity and the biocompatibility was satisfactory. In the treatment of chronic wounds in diabetic rats, VAC + HYP-NPS significantly promoted the re-epithelialization of chronic wounds and accelerated the healing of chronic wounds. In the process of chronic wounds healing, VAC + HYP-NPS played the antibacterial effect of chitosan, the angiogenic effect of VAC and the anti-inflammatory effect of HYP, and finally promoted the chronic wounds healing. Overall, the developed VAC + HYP-NPS have potential application in chronic wounds healing. In view of the complexity of the causes of chronic wounds, multi-target drug administration may be an effective way to treat chronic wounds.
Collapse
Affiliation(s)
- Bao Hou
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Yuanyuan Wen
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Xuerui Zhu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Mengting Qi
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Weiwei Cai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Bin Du
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Haijian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China
| | - Liying Qiu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, PR China.
| |
Collapse
|
19
|
Idowu OK, Oluyomi OO, Faniyan OO, Dosumu OO, Akinola OB. The synergistic ameliorative activity of peroxisome proliferator-activated receptor-alpha and gamma agonists, fenofibrate and pioglitazone, on hippocampal neurodegeneration in a rat model of insulin resistance. IBRAIN 2022; 8:251-263. [PMID: 37786742 PMCID: PMC10528802 DOI: 10.1002/ibra.12059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 10/04/2023]
Abstract
Insulin resistance (IR) is a risk factor for metabolic disorders and neurodegeneration. Peroxisome proliferator-activated receptor (PPAR) agonists have been proven to mitigate the neuronal pathology associated with IR. However, the synergetic efficacy of these agonists is yet to be fully described. Hence, we aimed to investigate the efficacy of PPARα/γ agonists (fenofibrate and pioglitazone) on a high-fat diet (HFD) and streptozotocin (STZ)-induced hippocampal neurodegeneration. Male Wistar rats (200 ± 25 mg/body weight [BW]) were divided into five groups. The experimental groups were fed on an HFD for 12 weeks coupled with 5 days of an STZ injection (30 mg/kg/BW, i.p) to induce IR. Fenofibrate (FEN; 100 mg/kg/BW, orally), pioglitazone (PIO; 20 mg/kg/BW, orally), and their combination were administered for 2 weeks postinduction. Behavioral tests were conducted, and blood was collected to determine insulin sensitivity after treatment. Animals were killed for assessment of oxidative stress, cellular morphology characterization, and astrocytic evaluation. HFD/STZ-induced IR increased malondialdehyde (MDA) levels and decreased glutathione (GSH) levels. Evidence of cellular alterations and overexpression of astrocytic protein was observed in the hippocampus. By contrast, monotherapy of FEN and PIO increased the GSH level (p < 0.05), decreased the MDA level (p < 0.05), and improved cellular morphology and astrocytic expression. Furthermore, the combined treatment led to improved therapeutic activities compared to monotherapies. In conclusion, FEN and PIO exerted a therapeutic synergistic effect on HFD/STZ-induced IR in the hippocampus.
Collapse
Affiliation(s)
| | | | - Oluwatomisin O. Faniyan
- Department of Physiology, School of Bioscience and Veterinary MedicineUniversity of CamerinoCamerinoItaly
| | | | | |
Collapse
|
20
|
Busato S, Ford HR, Abdelatty AM, Estill CT, Bionaz M. Peroxisome Proliferator-Activated Receptor Activation in Precision-Cut Bovine Liver Slices Reveals Novel Putative PPAR Targets in Periparturient Dairy Cows. Front Vet Sci 2022; 9:931264. [PMID: 35903133 PMCID: PMC9315222 DOI: 10.3389/fvets.2022.931264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolic challenges experienced by dairy cows during the transition between pregnancy and lactation (also known as peripartum), are of considerable interest from a nutrigenomic perspective. The mobilization of large amounts of non-esterified fatty acids (NEFA) leads to an increase in NEFA uptake in the liver, the excess of which can cause hepatic accumulation of lipids and ultimately fatty liver. Interestingly, peripartum NEFA activate the Peroxisome Proliferator-activated Receptor (PPAR), a transcriptional regulator with known nutrigenomic properties. The study of PPAR activation in the liver of periparturient dairy cows is thus crucial; however, current in vitro models of the bovine liver are inadequate, and the isolation of primary hepatocytes is time consuming, resource intensive, and prone to errors, with the resulting cells losing characteristic phenotypical traits within hours. The objective of the current study was to evaluate the use of precision-cut liver slices (PCLS) from liver biopsies as a model for PPAR activation in periparturient dairy cows. Three primiparous Jersey cows were enrolled in the experiment, and PCLS from each were prepared prepartum (−8.0 ± 3.6 DIM) and postpartum (+7.7± 1.2 DIM) and treated independently with a variety of PPAR agonists and antagonists: the PPARα agonist WY-14643 and antagonist GW-6471; the PPARδ agonist GW-50156 and antagonist GSK-3787; and the PPARγ agonist rosiglitazone and antagonist GW-9662. Gene expression was assayed through RT-qPCR and RNAseq, and intracellular triacylglycerol (TAG) concentration was measured. PCLS obtained from postpartum cows and treated with a PPARγ agonist displayed upregulation of ACADVL and LIPC while those treated with PPARδ agonist had increased expression of LIPC, PPARD, and PDK4. In PCLS from prepartum cows, transcription of LIPC was increased by all PPAR agonists and NEFA. TAG concentration tended to be larger in tissue slices treated with PPARδ agonist compared to CTR. Use of PPAR isotype-specific antagonists in PCLS cultivated in autologous blood serum failed to decrease expression of PPAR targets, except for PDK4, which was confirmed to be a PPARδ target. Transcriptome sequencing revealed considerable differences in response to PPAR agonists at a false discovery rate-adjusted p-value of 0.2, with the most notable effects exerted by the PPARδ and PPARγ agonists. Differentially expressed genes were mainly related to pathways involved with lipid metabolism and the immune response. Among differentially expressed genes, a subset of 91 genes were identified as novel putative PPAR targets in the bovine liver, by cross-referencing our results with a publicly available dataset of predicted PPAR target genes, and supplementing our findings with prior literature. Our results provide important insights on the use of PCLS as a model for assaying PPAR activation in the periparturient dairy cow.
Collapse
Affiliation(s)
- Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Hunter R. Ford
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Alzahraa M. Abdelatty
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- *Correspondence: Massimo Bionaz
| |
Collapse
|
21
|
Yin B, Wang YB, Li X, Hou XW. β‑aminoisobutyric acid ameliorates hypertensive vascular remodeling via activating the AMPK/SIRT1 pathway in VSMCs. Bioengineered 2022; 13:14382-14401. [PMID: 36694438 PMCID: PMC9995136 DOI: 10.1080/21655979.2022.2085583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) play a fundamental role in the pathogenesis of hypertension-related vascular remodeling. β-aminoisobutyric acid (BAIBA) is a nonprotein β-amino acid with multiple pharmacological actions. Recently, BAIBA has been shown to attenuate salt‑sensitive hypertension, but the role of BAIBA in hypertension-related vascular remodeling has yet to be fully clarified. This study examined the potential roles and underlying mechanisms of BAIBA in VSMC proliferation and migration induced by hypertension. Primary VSMCs were cultured from the aortas of Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Our results showed that BAIBA pretreatment obviously alleviated the phenotypic transformation, proliferation, and migration of SHR-derived VSMCs. Exogenous BAIBA significantly inhibited the release of inflammatory cytokines by diminishing phosphorylation and nuclear translocation of p65 NFκB, retarding IκBα phosphorylation and degradation, as well as erasing STAT3 phosphorylation in VSMCs. Supplementation of BAIBA triggered Nrf2 dissociation from Keap1 and inhibited oxidative stress in VSMCs from SHR. Mechanistically, activation of the AMPK/sirtuin 1 (SIRT1) axis was required for BAIBA to cube hypertension-induced VSMC proliferation, migration, oxidative damage and inflammatory response. Most importantly, exogenous BAIBA alleviated hypertension, ameliorated vascular remodeling and fibrosis, abated vascular oxidative burst and inflammation in SHR, an effect that was abolished by deficiency of AMPKα1 and SIRT1. BAIBA might serve as a novel therapeutic agent to prevent vascular remodeling in the context of hypertension.
Collapse
Affiliation(s)
- Bo Yin
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu-Bin Wang
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xiang Li
- Department of General Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Xu-Wei Hou
- Department of Human Anatomy, Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
22
|
Tanaka T, Talegawkar SA, Jin Y, Candia J, Tian Q, Moaddel R, Simonsick EM, Ferrucci L. Metabolomic Profile of Different Dietary Patterns and Their Association with Frailty Index in Community-Dwelling Older Men and Women. Nutrients 2022; 14:2237. [PMID: 35684039 PMCID: PMC9182888 DOI: 10.3390/nu14112237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022] Open
Abstract
Diet quality has been associated with slower rates of aging; however, the mechanisms underlying the role of a healthy diet in aging are not fully understood. To address this question, we aimed to identify plasma metabolomic biomarkers of dietary patterns and explored whether these metabolites mediate the relationship between diet and healthy aging, as assessed by the frailty index (FI) in 806 participants of the Baltimore Longitudinal Study of Aging. Adherence to different dietary patterns was evaluated using the Mediterranean diet score (MDS), Mediterranean-DASH Diet Intervention for Neurodegenerative Delay (MIND) score, and Alternate Healthy Eating Index-2010 (AHEI). Associations between diet, FI, and metabolites were assessed using linear regression models. Higher adherence to these dietary patterns was associated with lower FI. We found 236, 218, and 278 metabolites associated with the MDS, MIND, and AHEI, respectively, with 127 common metabolites, which included lipids, tri/di-glycerides, lyso/phosphatidylcholine, amino acids, bile acids, ceramides, cholesterol esters, fatty acids and acylcarnitines, indoles, and sphingomyelins. Metabolomic signatures of diet explained 28%, 37%, and 38% of the variance of the MDS, MIND, and AHEI, respectively. Signatures of MIND and AHEI mediated 55% and 61% of the association between each dietary pattern with FI, while the mediating effect of MDS signature was not statistically significant. The high number of metabolites associated with the different dietary patterns supports the notion of common mechanisms that underly the relationship between diet and frailty. The identification of multiple metabolite classes suggests that the effect of diet is complex and not mediated by any specific biomarkers. Furthermore, these metabolites may serve as biomarkers for poor diet quality to identify individuals for targeted dietary interventions.
Collapse
Affiliation(s)
- Toshiko Tanaka
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD 21224, USA; (J.C.); (Q.T.); (E.M.S.); (L.F.)
| | - Sameera A. Talegawkar
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA; (S.A.T.); (Y.J.)
| | - Yichen Jin
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA; (S.A.T.); (Y.J.)
| | - Julián Candia
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD 21224, USA; (J.C.); (Q.T.); (E.M.S.); (L.F.)
| | - Qu Tian
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD 21224, USA; (J.C.); (Q.T.); (E.M.S.); (L.F.)
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD 21224, USA;
| | - Eleanor M. Simonsick
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD 21224, USA; (J.C.); (Q.T.); (E.M.S.); (L.F.)
| | - Luigi Ferrucci
- Longitudinal Studies Section, National Institute on Aging, Baltimore, MD 21224, USA; (J.C.); (Q.T.); (E.M.S.); (L.F.)
| |
Collapse
|
23
|
Liu X, Pan Y, Shen Y, Liu H, Zhao X, Li J, Ma N. Protective Effects of Abrus cantoniensis Hance on the Fatty Liver Hemorrhagic Syndrome in Laying Hens Based on Liver Metabolomics and Gut Microbiota. Front Vet Sci 2022; 9:862006. [PMID: 35498747 PMCID: PMC9051509 DOI: 10.3389/fvets.2022.862006] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 11/22/2022] Open
Abstract
As a metabolic disease, fatty liver hemorrhagic syndrome (FLHS) has become a serious concern in laying hens worldwide. Abrus cantoniensis Hance (AC) is a commonly used plant in traditional medicine for liver disease treatment. Nevertheless, the effect and mechanism of the decoction of AC (ACD) on FLHS remain unclear. In this study, ultra-high performance liquid chromatography analysis was used to identify the main phytochemicals in ACD. FLHS model of laying hens was induced by a high-energy low-protein (HELP) diet, and ACD (0.5, 1, 2 g ACD/hen per day) was given to the hens in drinking water at the same time for 48 days. Biochemical blood indicators and histopathological analysis of the liver were detected and observed to evaluate the therapeutic effect of ACD. Moreover, the effects of ACD on liver metabolomics and gut microbiota in laying hens with FLHS were investigated. The results showed that four phytochemicals, including abrine, hypaphorine, vicenin-2, and schaftoside, were identified in ACD. ACD treatment ameliorated biochemical blood indicators in laying hens with FLHS by decreasing aspartate aminotransferase, alanine aminotransferase, triglycerides, low-density lipoprotein cholesterol, and total cholesterol, and increasing high-density lipoprotein cholesterol. In addition, lipid accumulation in the liver and pathological damages were relieved in ACD treatment groups. Moreover, distinct changes in liver metabolic profile after ACD treatment were observed, 17 endogenous liver metabolites mainly associated with the metabolism of arachidonic acid, histidine, tyrosine, and tryptophan were reversed by ACD. Gut microbiota analysis revealed that ACD treatment significantly increased bacterial richness (Chao 1, P < 0.05; Ace, P < 0.01), and upregulated the relative abundance of Bacteroidetes and downregulated Proteobacteria, improving the negative effects caused by HELP diet in laying hens. Taken together, ACD had a protective effect on FLHS by regulating blood lipids, reducing liver lipid accumulation, and improving the dysbiosis of liver metabolomics and gut microbiota.
Collapse
Affiliation(s)
- Xu Liu
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding, China
| | - Yinchuan Pan
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding, China
| | - Youming Shen
- Research Institute of Pomology, Chinese Academy of Agricultural Sciences, Xingcheng, China
| | - Hailong Liu
- Institute of Animal Husbandry and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding, China
| | - Jianyong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou, China
- Jianyong Li
| | - Ning Ma
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding, China
- *Correspondence: Ning Ma
| |
Collapse
|
24
|
Erol SA, Anuk AT, Tanaçan A, Semiz H, Keskin HL, Neşelioğlu S, Erel Ö, Moraloğlu Tekin Ö, Şahin D. An evaluation of maternal serum dynamic thiol-disulfide homeostasis and ischemia modified albumin changes in pregnant women with COVID-19. Turk J Obstet Gynecol 2022; 19:21-27. [PMID: 35343216 PMCID: PMC8966320 DOI: 10.4274/tjod.galenos.2022.72929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: It is thought that oxidative stress, free radicals, reactive oxygen species and reactive nitrogen species affect the pathophysiology of coronavirus disease-2019 (COVID-19). This study aimed to evaluate the oxidative status in pregnant patients with COVID-19 infection according to the changes seen in the levels of maternal serum thiol-disulfide and ischemia-modified albumin (IMA). Materials and Methods: A study group was formed of 40 pregnant women with confirmed COVID-19 infection (study group) and a control group of 40 healthy pregnant women with no risk factors determined. In this prospective, case-controlled study, analyses were made of the maternal serum native thiol, total thiol, disulfide, IMA, and disulfide/native thiol concentrations. Results: The maternal serum native thiol and total thiol concentrations in the study group were determined to be statistically significantly lower (p=0.007 and p=0.006, respectively), and the disulfide/native thiol ratio was higher but not to a level of statistical significance (p=0.473). There was no difference between the two groups regarding IMA levels (p=0.731). Conclusion: The thiol-disulfide balance was seen to shift in the oxidant direction in pregnancies with COVID-19, which might support the view that ischemic processes play a role in the etiopathogenesis of this novel disease.
Collapse
|
25
|
Fofana S, Ouédraogo M, Esposito RC, Ouedraogo WP, Delporte C, Van Antwerpen P, Mathieu V, Guissou IP. Systematic Review of Potential Anticancerous Activities of Erythrina senegalensis DC (Fabaceae). PLANTS (BASEL, SWITZERLAND) 2021; 11:plants11010019. [PMID: 35009024 PMCID: PMC8747466 DOI: 10.3390/plants11010019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 05/04/2023]
Abstract
The objective of this study was to carry out a systematic review of the substances isolated from the African medicinal plant Erythrina senegalensis, focusing on compounds harboring activities against cancer models detailed in depth herein at both in vitro and in vivo preclinical levels. The review was conducted through Pubmed and Google Scholar. Nineteen out of the forty-two secondary metabolites isolated to date from E. senegalensis displayed interesting in vitro and/or in vivo antitumor activities. They belonged to alkaloid (Erysodine), triterpenes (Erythrodiol, maniladiol, oleanolic acid), prenylated isoflavonoids (senegalensin, erysenegalensein E, erysenegalensein M, alpinumisoflavone, derrone, warangalone), flavonoids (erythrisenegalone, senegalensein, lupinifolin, carpachromene) and pterocarpans (erybraedine A, erybraedine C, phaseollin). Among the isoflavonoids called "erysenegalensein", only erysenealenseins E and M have been tested for their anticancerous properties and turned out to be cytotoxic. Although the stem bark is the most frequently used part of the plant, all pterocarpans were isolated from roots and all alkaloids from seeds. The mechanisms of action of its metabolites include apoptosis, pyroptosis, autophagy and mitophagy via the modulation of cytoplasmic proteins, miRNA and enzymes involved in critical pathways deregulated in cancer. Alpinumisoflavone and oleanolic acid were studied in a broad spectrum of cancer models both in vitro and in preclinical models in vivo with promising results. Other metabolites, including carpachromen, phaseollin, erybraedin A, erysenegalensein M and maniladiol need to be further investigated, as they display potent in vitro effects.
Collapse
Affiliation(s)
- Souleymane Fofana
- Laboratory of Drug Science, Higher Institute of Health Sciences (INSSA), Nazi BONI University, Bobo-Dioulasso P.O. Box 1091, Burkina Faso;
| | - Moussa Ouédraogo
- Laboratory of Drug Development (LADME), Training and Research Unit, Health Sciences, Joseph KI-ZERBO University, Ouagadougou P.O. Box 7021, Burkina Faso; (M.O.); (W.P.O.)
| | - Rafaèle Calvo Esposito
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- Protein Chemistry Unit, Department of General Chemistry I, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Windbedema Prisca Ouedraogo
- Laboratory of Drug Development (LADME), Training and Research Unit, Health Sciences, Joseph KI-ZERBO University, Ouagadougou P.O. Box 7021, Burkina Faso; (M.O.); (W.P.O.)
| | - Cédric Delporte
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Universite’ Libre de Bruxelles (ULB), 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Pierre Van Antwerpen
- RD3-Pharmacognosy, Bioanalysis and Drug Discovery Unit and Analytical Platform, Faculty of Pharmacy, Universite’ Libre de Bruxelles (ULB), 1050 Brussels, Belgium; (C.D.); (P.V.A.)
| | - Véronique Mathieu
- Department of Pharmacotherapy and Pharmaceuticals, Faculty of Pharmacy, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
- ULB Cancer Research Center, Université Libre de Bruxelles (ULB), 1050 Bruxelles, Belgium
- Correspondence: (V.M.); (I.P.G.); Tel.: +32-478-31-73-88 (V.M.)
| | - Innocent Pierre Guissou
- Faculty of Health Sciences, Saint Thomas d’Aquin University, Ouagadougou P.O. Box 10212, Burkina Faso
- Correspondence: (V.M.); (I.P.G.); Tel.: +32-478-31-73-88 (V.M.)
| |
Collapse
|
26
|
DAI YL, ZHOU DY, JIANG YF, ZHENG F, YUE H, You-Jin JEON. 6-Bromohypaphorine isolated from red sea cucumbers Apostichopus japonicus exhibits potent anticancer activity in A549 cancer cell line. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2021. [DOI: 10.1016/j.cjac.2021.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
27
|
Huang W, Lv Q, Xiao Y, Zhong Z, Hu B, Yan S, Yan Y, Zhang J, Shi T, Jiang L, Li W, Lu G. Triggering Receptor Expressed on Myeloid Cells 2 Protects Dopaminergic Neurons by Promoting Autophagy in the Inflammatory Pathogenesis of Parkinson's Disease. Front Neurosci 2021; 15:745815. [PMID: 34867158 PMCID: PMC8641649 DOI: 10.3389/fnins.2021.745815] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disorder with an inflammatory response as the core pathogenic mechanism. Previous human genetics findings support the view that the loss of TREM2 function will aggravate neurodegeneration, and TREM2 is one of the most highly expressed receptors in microglia. However, the role of TREM2 in the inflammatory mechanism of PD is not clear. In our study, it was found both in vivo and in vitro that the activation of microglia not only promoted the secretion of inflammatory factors but also decreased the level of TREM2 and inhibited the occurrence of autophagy. In contrast, an increase in the level of TREM2 decreased the expression of inflammatory factors and enhanced the level of autophagy through the p38 MAPK/mTOR pathway. Moreover, increased TREM2 expression significantly decreased the apoptosis of dopaminergic (DA) neurons and improved the motor ability of PD mice. In summary, TREM2 is an important link between the pathogenesis of PD and inflammation. Our study provides a new view for the mechanism of TREM2 in PD and reveals TREM2 as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Wei Huang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qiankun Lv
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfei Xiao
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Zhen Zhong
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Hu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Si Yan
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yufang Yan
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junjun Zhang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Shi
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lijuan Jiang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohui Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
28
|
Preparation of W/O Hypaphorine-Chitosan Nanoparticles and Its Application on Promoting Chronic Wound Healing via Alleviating Inflammation Block. NANOMATERIALS 2021; 11:nano11112830. [PMID: 34835594 PMCID: PMC8625710 DOI: 10.3390/nano11112830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/10/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022]
Abstract
Chronic wound repair is a common complication in patients with diabetes mellitus, which causes a heavy burden on social medical resources and the economy. Hypaphorine (HYP) has good anti-inflammatory effect, and chitosan (CS) is used in the treatment of wounds because of its good antibacterial effect. The purpose of this research was to investigate the role and mechanism of HYP-nano-microspheres in the treatment of wounds for diabetic rats. The morphology of HYP-NPS was observed by transmission electron microscopy (TEM). RAW 264.7 macrophages were used to assess the bio-compatibility of HYP-NPS. A full-thickness dermal wound in a diabetic rat model was performed to evaluate the wound healing function of HYP-NPS. The results revealed that HYP-NPS nanoparticles were spherical with an average diameter of approximately 50 nm. The cell experiments hinted that HYP-NPS had the potential as a trauma material. The wound test in diabetic rats indicated that HYP-NPS fostered the healing of chronic wounds. The mechanism was through down-regulating the expression of pro-inflammatory cytokines IL-1β and TNF-α in the skin of the wound, and accelerating the transition of chronic wound from inflammation to tissue regeneration. These results indicate that HYP-NPS has a good application prospect in the treatment of chronic wounds.
Collapse
|
29
|
Florian SU, Sepulveda B, Torres-Benítez A, Simirgiotis MJ, Fuentes-Estrada M, Areche C, García-Beltrán O. Erythrinoid and indol alkaloids isolated from the seeds of Erythrina rubrinervia Kunth: Chemotaxonomic significance. BIOCHEM SYST ECOL 2021. [DOI: 10.1016/j.bse.2021.104295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
30
|
Zhang M, Wu Q, Zhao R, Yao X, Du X, Liu Q, Lv G, Xiao S. Isobavachalcone ameliorates cognitive deficits, and Aβ and tau pathologies in triple-transgenic mice with Alzheimer's disease. Food Funct 2021; 12:7749-7761. [PMID: 34269361 DOI: 10.1039/d1fo01306h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects 50 million people worldwide. The current medicines have modest benefits in preventing or curing AD. Thus, it is urgent to discover drugs with the potential to change the progression of the disease. The primary clinical symptoms are memory loss and anxiety, while the critical pathological characteristics are Aβ plaques and hyperphosphorylated tau tangles. In this study, isobavachalcone (ISO), isolated from Psoralea corylifolia, was administered to 3×Tg-AD mice. It has been shown that this compound could significantly improve anxiety, memory and recognition deficits in the AD mice, attenuate the accumulation of Aβ oligomers, reduce the hyperphosphorylation of tau, and prevent the production of tau filaments. The metabolomic analysis implicates that the most probable pathways affected by ISO were bile secretion, tyrosine metabolism, and purine metabolism. In summary, ISO possesses the potential for further development as a drug candidate for AD.
Collapse
Affiliation(s)
- Mohan Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ding YH, Miao RX, Zhang Q. Hypaphorine exerts anti-inflammatory effects in sepsis induced acute lung injury via modulating DUSP1/p38/JNK pathway. Kaohsiung J Med Sci 2021; 37:883-893. [PMID: 34250720 DOI: 10.1002/kjm2.12418] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/02/2023] Open
Abstract
Sepsis is a systemic inflammatory response syndrome attributed to infection, while sepsis-induced acute lung injury (ALI) has high morbidity and mortality. Here, we aimed to explore the specific mechanism of hypaphorine's anti-inflammatory effects in ALI. Lipopolysaccharide (LPS) was adopted to construct ALI model both in vivo and in vitro. BEAS-2B cell viability and apoptosis was testified by the MTT assay and flow cytometry. Reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were performed to examine the expression of proinflammatory cytokines (IL-1β, IL-6, TNF-α, and IL-18), and Western blot was adopted to examine the expression of the apoptosis-related proteins (Bax, Bcl2, and Caspase3) and the DUSP1/p38/JNK signaling pathway. At the same time, lung injury score, lactate dehydrogenase (LDH) and myeloperoxidase (MPO) activity were monitored. The dry/wet weight method was used to examine lung edema, and the total protein content in BALF was determined to test pulmonary vascular permeability. As the data suggested, hypaphorine inhibited the LPS-mediated apoptosis of alveolar epithelial cells. What is more, hypaphorine attenuated the expression of inflammatory factors (IL-1β, IL-6, TNF-α, and IL-18) and inactivated the p38/JNK signaling pathway through upregulating DUSP1 in a dose-dependent manner. Meanwhile, DUSP1 knockdown weakened the anti-inflammatory effect of hypaphorine on LPS-mediated lung injury. Furthermore, hypaphorine also relieved LPS induced ALI in rats with anti-inflammatory effects. Taken together, hypaphorine prevented LPS-mediated ALI and proinflammatory response via inactivating the p38/JNK signaling pathway by upregulating DUSP1.
Collapse
Affiliation(s)
- Yu-Hua Ding
- Department of Pharmacy, Zaozhuang Hospital of Traditional Chinese Medicine, Zaozhuang, Shandong, China
| | - Run-Xin Miao
- Department of Emergency, Zaozhuang Hospital of Traditional Chinese Medicine, Zaozhuang, Shandong, China
| | - Qiang Zhang
- Department of Pharmacy, Zaozhuang Hospital of Traditional Chinese Medicine, Zaozhuang, Shandong, China
| |
Collapse
|
32
|
Xiao S, Wu Q, Yao X, Zhang J, Zhong W, Zhao J, Liu Q, Zhang M. Inhibitory Effects of Isobavachalcone on Tau Protein Aggregation, Tau Phosphorylation, and Oligomeric Tau-Induced Apoptosis. ACS Chem Neurosci 2021; 12:123-132. [PMID: 33320518 DOI: 10.1021/acschemneuro.0c00617] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases without any effective medicine treatments. The neurofibrillary tangles containing hyperphosphorylated tau protein are one important pathological characteristic. Thus, one practicable strategy for AD drug design is to discover compounds that could inhibit tau protein aggregation and/or phosphorylation. In this study, isobavachalcone, a natural plant-derived compound, has been shown to inhibit tau protein aggregation and disaggregate tau fibrils in vitro by directly interacting with tau protein at amino acids I278, V309, etc. It is able to reduce tau phosphorylation at four disease-related sites in vivo by regulating the critical kinase and protein phosphatase, GSK3β and PP2A. The compound also exhibits protection against tau oligomers-induced apoptosis through the mitochondria and ER mediated apoptotic pathways. In summary, isobavachalcone is a promising candidate for further evaluation as a potential preventive and therapeutic medicine for AD.
Collapse
Affiliation(s)
- Shifeng Xiao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
- Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
| | - Qiuping Wu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xuanbao Yao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jiahao Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Weicong Zhong
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Junyi Zhao
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Mohan Zhang
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518060, China
| |
Collapse
|
33
|
Che Man R, Sulaiman N, Ishak MF, Bt Hj Idrus R, Abdul Rahman MR, Yazid MD. The Effects of Pro-Inflammatory and Anti-Inflammatory Agents for the Suppression of Intimal Hyperplasia: An Evidence-Based Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217825. [PMID: 33114632 PMCID: PMC7672569 DOI: 10.3390/ijerph17217825] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Anti-atherogenic therapy is crucial in halting the progression of inflammation-induced intimal hyperplasia. The aim of this concise review was to methodically assess the recent findings of the different approaches, mainly on the recruitment of chemokines and/or cytokine and its effects in combating the intimal hyperplasia caused by various risk factors. Pubmed and Scopus databases were searched, followed by article selection based on pre-set inclusion and exclusion criteria. The combination of keywords used were monocyte chemoattractant protein-1 OR MCP-1 OR TNF-alpha OR TNF-α AND hyperplasia OR intimal hyperplasia OR neointimal hyperplasia AND in vitro. These keywords combination was incorporated in the study and had successfully identified 77 articles, with 22 articles were acquired from Pubmed, whereas 55 articles were obtained from Scopus. However, after title screening, only twelve articles meet the requirements of defined inclusion criteria. We classified the data into 4 different approaches, i.e., utilisation of natural product, genetic manipulation and protein inhibition, targeted drugs in clinical setting, and chemokine and cytokines induction. Most of the articles are working on genetic manipulation targeted on specific pathway to inhibit the pro-inflammatory factors expression. We also found that the utilisation of chemokine- and cytokine-related treatments are emerging throughout the years. However, there is no study utilising the combination of approaches that might give a better outcome in combating intimal hyperplasia. Hopefully, this concise review will provide an insight regarding the usage of different novel approaches in halting the progression of intimal hyperplasia, which serves as a key factor for the development of atherosclerosis in cardiovascular disease.
Collapse
Affiliation(s)
- Rohaina Che Man
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Mohamad Fikeri Ishak
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia
| | - Mohd Ramzisham Abdul Rahman
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras 56000, Kuala Lumpur, Malaysia; (R.C.M.); (N.S.); (M.F.I.); (R.B.H.I.)
- Correspondence: ; Tel.: +603-9145-6995
| |
Collapse
|
34
|
Federico S, Pozzetti L, Papa A, Carullo G, Gemma S, Butini S, Campiani G, Relitti N. Modulation of the Innate Immune Response by Targeting Toll-like Receptors: A Perspective on Their Agonists and Antagonists. J Med Chem 2020; 63:13466-13513. [PMID: 32845153 DOI: 10.1021/acs.jmedchem.0c01049] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are a class of proteins that recognize pathogen-associated molecular patterns (PAMPs) and damaged-associated molecular patterns (DAMPs), and they are involved in the regulation of innate immune system. These transmembrane receptors, localized at the cellular or endosomal membrane, trigger inflammatory processes through either myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling pathways. In the last decades, extensive research has been performed on TLR modulators and their therapeutic implication under several pathological conditions, spanning from infections to cancer, from metabolic disorders to neurodegeneration and autoimmune diseases. This Perspective will highlight the recent discoveries in this field, emphasizing the role of TLRs in different diseases and the therapeutic effect of their natural and synthetic modulators, and it will discuss insights for the future exploitation of TLR modulators in human health.
Collapse
Affiliation(s)
- Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Luca Pozzetti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
35
|
Zhou Y, Little PJ, Downey L, Afroz R, Wu Y, Ta HT, Xu S, Kamato D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol Transl Sci 2020; 3:457-471. [PMID: 32566912 PMCID: PMC7296543 DOI: 10.1021/acsptsci.9b00100] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are dominant components of the innate immune system. Activated by both pathogen-associated molecular patterns and damage-associated molecular patterns, TLRs underpin the pathology of numerous inflammation related diseases that include not only immune diseases, but also cardiovascular disease (CVD), diabetes, obesity, and cancers. Growing evidence has demonstrated that TLRs are involved in multiple cardiovascular pathophysiologies, such as atherosclerosis and hypertension. Specifically, a trial called the Canakinumab Anti-inflammatory Thrombosis Outcomes Study showed the use of an antibody that neutralizes interleukin-1β, reduces the recurrence of cardiovascular events, demonstrating inflammation as a therapeutic target and also the research value of targeting the TLR system in CVD. In this review, we provide an update of the interplay between TLR signaling, inflammatory mediators, and atherothrombosis, with an aim to identify new therapeutic targets for atherothrombotic CVD.
Collapse
Affiliation(s)
- Ying Zhou
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Peter J. Little
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| | - Liam Downey
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Rizwana Afroz
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Yuao Wu
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Hang T. Ta
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Suowen Xu
- Aab
Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Danielle Kamato
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| |
Collapse
|
36
|
Dehydroabietic acid alleviates high fat diet-induced insulin resistance and hepatic steatosis through dual activation of PPAR-γ and PPAR-α. Biomed Pharmacother 2020; 127:110155. [PMID: 32413669 DOI: 10.1016/j.biopha.2020.110155] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/01/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Dual-PPAR-α/γ agonist has the dual potentials to improve insulin resistance (IR) and hepatic steatosis associated with obesity. This study aimed to investigate whether dehydroabietic acid (DA), a naturally occurred compound, can bind to and activate both PPAR-γ and PPAR-α to ameliorate IR and hepatic steatosis in high-fat diet (HFD)-fed mice.. We found that DA formed stable hydrogen bonds with the ligand-binding domains of PPAR-γ and PPAR-α. DA treatment also promoted 3T3-L1 differentiation via PPAR-γ activation, and mitochondrial oxygen consumption in HL7702 cells via PPAR-α activation. In HFD-fed mice, DA treatment alleviated glucose intolerance and IR, and reduced hepatic steatosis, liver injury markers (ALT, AST), and lipid accumulation, and promoted mRNA expression of PPAR-γ and PPAR-α signaling elements involved in IR and lipid metabolism in vivo and in vitro, and inhibited mRNA expression of pro-inflammatory factors. Therefore, DA is a dual-PPAR-α/γ and PPAR-γ partial agonist, which can attenuate IR and hepatic steatosis induced by HFD-consumption in mice.
Collapse
|
37
|
Cheng WX, Zhong S, Meng XB, Zheng NY, Zhang P, Wang Y, Qin L, Wang XL. Cinnamaldehyde Inhibits Inflammation of Human Synoviocyte Cells Through Regulation of Jak/Stat Pathway and Ameliorates Collagen-Induced Arthritis in Rats. J Pharmacol Exp Ther 2020; 373:302-310. [PMID: 32029577 DOI: 10.1124/jpet.119.262907] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 02/03/2020] [Indexed: 08/19/2024] Open
Abstract
Cinnamaldehyde (Cin), a bioactive cinnamon essential oil from traditional Chinese medicine herb Cinnamomum cassia, has been reported to have multipharmacological activities including anti-inflammation. However, its role and molecular mechanism of anti-inflammatory activity in musculoskeletal tissues remains unclear. Here, we first investigated the effects and molecular mechanisms of Cin in human synoviocyte cells. Then in vivo therapeutic effect of Cin on collagen-induced arthritis (CIA) also studied. Cell Counting Kit CCK-8 assay was performed to evaluate the cell cytotoxicity. Proinflammatory cytokine expression was evaluated using quantitative polymerase chain reaction and ELISA. Protein expression was measured by western blotting. The in vivo effect of Cin (75 mg/kg per day) was evaluated in rats with CIA by gavage administration. Disease progression was assessed by clinical scoring, radiographic, and histologic examinations. Cin significantly inhibited interleukin (IL)-1β-induced IL-6, IL-8, and tumor necrosis factor-α release from human synoviocyte cells. The molecular analysis revealed that Cin impaired IL-6-induced activation of Janus kinase 2 (JAK2), signal transducer and activator of transcription 1 (STAT1), and STAT3 signaling pathway by inhibiting the phosphorylation of JAK2, STAT1, and STAT3, without affecting NF-κB pathway. Cin reduced collagen-induced swollen paw volume of arthritic rats. The anti-inflammation effects of Cin were associated with decreased severity of arthritis, joint swelling, and reduced bone erosion and destruction. Furthermore, serum IL-6 level was decreased when Cin administered therapeutically to CIA rats. Cin suppresses IL-1β-induced inflammation in synoviocytes through the JAK/STAT pathway and alleviated collagen-induced arthritis in rats. These data indicated that Cin might be a potential traditional Chinese medicine-derived, disease-modifying, antirheumatic herbal drug. SIGNIFICANCE STATEMENT: In this study, we found that cinnamaldehyde (Cin) suppressed proinflammatory cytokines secretion in rheumatology arthritis synoviocyte cells by Janus kinase/signal transducer and activator of transcription pathway. The in vivo results showed that Cin ameliorated collagen-induced arthritis in rats. These findings indicate that Cin is a potential traditional Chinese medicine-derived, disease-modifying, antirheumatic herbal drug.
Collapse
Affiliation(s)
- Wen-Xiang Cheng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Shan Zhong
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Xiang-Bo Meng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Nian-Ye Zheng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Peng Zhang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Yun Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| | - Xin-Luan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (W.-X.C., S.Z., X.-B.M., P.Z., L.Q., X.-L.W.); University of Chinese Academy of Sciences, Beijing, China (W.-X.C., P.Z., X.-L.W.); Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China (N.-Y.Z., L.Q., X.-L.W.); and Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China (S.Z., Y.W.)
| |
Collapse
|
38
|
Fu XQ, Liu B, Wang YP, Li JK, Zhu PL, Li T, Tse KW, Chou JY, Yin CL, Bai JX, Liu YX, Chen YJ, Yu ZL. Activation of STAT3 is a key event in TLR4 signaling-mediated melanoma progression. Cell Death Dis 2020; 11:246. [PMID: 32312954 PMCID: PMC7171093 DOI: 10.1038/s41419-020-2440-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/22/2022]
Abstract
Malignant melanoma is aggressive and has a high mortality rate. Toll-like receptor 4 (TLR4) has been linked to melanoma growth, angiogenesis and metastasis. However, signal transduction mediated by TLR4 for driving melanoma progression is not fully understood. Signal transducer and activator of transcription 3 (STAT3) has been identified as a major oncogene in melanoma progression. We found: that TLR4 expression positively correlates with activation/phosphorylation of STAT3 in human melanoma samples; that TLR4 ligands activate STAT3 through MYD88 and TRIF in melanoma cells; and that intratumoral activation of TLR4 increases STAT3 activation in the tumor and promotes tumor growth, angiogenesis, epithelial-mesenchymal transition (EMT) and the formation of an immunosuppressive tumor microenvironment in mice. Further, we found that the effects mediated by activating TLR4 are weakened by suppressing STAT3 function with a dominant negative STAT3 variant in melanoma. Collectively, our work identifies STAT3 activation as a key event in TLR4 signaling-mediated melanoma progression, shedding new light on the pathophysiology of melanoma.
Collapse
Affiliation(s)
- Xiu-Qiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
- Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Bin Liu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ya-Ping Wang
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jun-Kui Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Pei-Li Zhu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
- Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ting Li
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Kai-Wing Tse
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ji-Yao Chou
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Cheng-Le Yin
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Jing-Xuan Bai
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Yu-Xi Liu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Ying-Jie Chen
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Consun Chinese Medicines Research Centre for Renal Diseases, Hong Kong Baptist University, Hong Kong, China.
- Research and Development Centre for Natural Health Products, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.
- JaneClare Transdermal TCM Therapy Laboratory, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
39
|
Xie Z, Zhao J, Wang H, Jiang Y, Yang Q, Fu Y, Zeng H, Hölscher C, Xu J, Zhang Z. Magnolol alleviates Alzheimer's disease-like pathology in transgenic C. elegans by promoting microglia phagocytosis and the degradation of beta-amyloid through activation of PPAR-γ. Biomed Pharmacother 2020; 124:109886. [PMID: 32000045 DOI: 10.1016/j.biopha.2020.109886] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/25/2019] [Accepted: 12/29/2019] [Indexed: 12/28/2022] Open
Abstract
This study aims to investigate whether magnolol (MG), a natural neolignane compound, can prevent AD induced by beta-amyloid (Aβ) and the possible mechanisms involved. MG dose-dependently reduces Aβ deposition, toxicity and memory impairment caused by Aβ in transgenic C. elegans. More importantly, these effects are reversed by GW9662, a selective peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist. MG is more effective in enhancing PPAR-γ luciferase levels than honokiol (HK). Meanwhile, MG has the potential to bind with the ligand binding domain of PPAR-γ (PPAR-γ-LBD). As expected, MG inhibited the luciferase activity of NF-κB and its target genes of inflammatory cytokines, and this effect was blocked by GW9662. The luciferase activity of Nrf2-ARE expression can be activated by MG and decreased Aβ-induced reactive oxygen species (ROS). The target gene LXR of PPAR-γ is activated by MG, which upregulates ApoE and promotes microglia phagocytosis and the degradation of Aβ, and these effects were also reversed by GW9662. In summary, MG can attenuate Aβ-induced AD and the underlying mechanism is the reduction of inflammation and promotion of phagocytosis and degradation of Aβ, which is dependent on PPAR-γ.
Collapse
Affiliation(s)
- Zhishen Xie
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jianping Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Hui Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yali Jiang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qiaoling Yang
- Department of Pharmacy, Children's Hospital of Shanghai, Children's Hospital Affiliate to Shanghai Jiao Tong University, Shanghai 200040, China
| | - Yu Fu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Huahui Zeng
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Christian Hölscher
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
40
|
Chen X, Wang R, Meng W, Zhang X. Exploration of the Molecular Mechanism of FUZI (Aconiti Lateralis Radix Praeparata) in Allergic Rhinitis Treatment Based on Network Pharmacology. Med Sci Monit 2020; 26:e920872. [PMID: 32114589 PMCID: PMC7065509 DOI: 10.12659/msm.920872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
FUZI (Aconiti Lateralis Radix Praeparata) is a traditional Chinese medicine herb used extensively for nourishing yang (regarded as the positive, male universal force), which is critical in treatment of allergic rhinitis. In this paper, FUZI was explored based on network pharmacology. The active components of FUZI were screened out, its protein targets were assessed, and the protein interaction network map was built with the differential protein of allergic rhinitis, as an attempt to determine the critical targets of FUZI for treating allergic rhinitis. Subsequently, DAVID was employed to explore the biological function and pathway enrichment to determine the biological pathway of FUZI for treating allergic rhinitis. As suggested by the results, FUZI is likely to affect the inhibition of inflammation and the regulation of immunity, probably reducing the incidence of allergic rhinitis, or alleviating nasal discomfort attributed to allergic inflammation. The targets and pathways of FUZI for treating allergic rhinitis assessed by network pharmacology provided a direction for our subsequent studies and may be a novel therapeutic target.
Collapse
Affiliation(s)
- Xiangjing Chen
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Renzhong Wang
- Department of Otorhinolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Wei Meng
- Department of Otorhinolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| | - Xin Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China (mainland)
| |
Collapse
|
41
|
Amorim J, Borges MDC, Fabro AT, Contini SHT, Valdevite M, Pereira AMS, Carmona F. The ethanolic extract from Erythrina mulungu Benth. flowers attenuates allergic airway inflammation and hyperresponsiveness in a murine model of asthma. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:111467. [PMID: 30102994 DOI: 10.1016/j.jep.2018.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erythrina mulungu Benth. ("mulungu", Fabaceae) is a Brazilian native species with ethnopharmacological use for respiratory diseases. However, the effects of E. mulungu on the respiratory were never studied. AIMS OF THE STUDY To evaluate the effects of an ethanolic extract from flowers of E. mulungu in ovalbumin (OVA)-induced asthma in mice, and to study the mechanisms involved. MATERIALS AND METHODS OVA-sensitized mice were intraperitoneally (i.p.) treated with four doses (200, 400, 600, and 800 mg/kg) of the E. mulungu extract or dexamethasone (DEXA, 2 mg/kg) during seven consecutive days and simultaneously challenged with intranasal OVA. Bronchial hyperresponsiveness was evaluated in vivo, 24 h after the last OVA challenge. Bronchoalveolar lavage (BAL) was collected for counting the number of total and differential inflammatory cells. Blood was collected for measurement of anti-OVA IgE levels. Levels of cytokines interleukin (IL)- 4, IL-5, IL-10, IL-13, and interferon (INF)-γ were measured in pulmonary homogenate by ELISA. The recruitment of inflammatory cells to the lung tissue was determined using hematoxylin and eosin staining (H&E). The extract's chromatographic profile was evaluated by ultra-performance liquid chromatography-mass spectrometry (UPLC-MS). RESULTS The treatment with E. mulungu extract significantly reduced bronchial hyperresponsiveness, significantly reduced the number of leukocytes, eosinophils, and lymphocytes in BAL, and significantly decreased the levels of IL-4 and IL-5, while increased levels of IL-13 and INF-γ. In addition, E. mulungu significantly decreased the cellular inflammatory infiltration in the lung tissue. Erysotrine, erysotrine-N-oxide, and hypaphorine were the major constituents identified in the extract. CONCLUSION Collectively, these results confirm the potential of E. mulungu for asthma treatment, through modulation of inflammatory response, supporting its ethnopharmacological use for respiratory diseases.
Collapse
Affiliation(s)
- Jowanka Amorim
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes S/N, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Marcos de Carvalho Borges
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes S/N, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Alexandre Todorovic Fabro
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes S/N, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Silvia Helena Taleb Contini
- Department of Biotechnology in Medicinal Plants, Ribeirão Preto University, Av. Costábile Romano 2201, 14096-900 Ribeirão Preto, SP, Brazil
| | - Mayara Valdevite
- Department of Biotechnology in Medicinal Plants, Ribeirão Preto University, Av. Costábile Romano 2201, 14096-900 Ribeirão Preto, SP, Brazil
| | - Ana Maria Soares Pereira
- Department of Biotechnology in Medicinal Plants, Ribeirão Preto University, Av. Costábile Romano 2201, 14096-900 Ribeirão Preto, SP, Brazil
| | - Fabio Carmona
- Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes S/N, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
42
|
Caruso G, Fresta CG, Fidilio A, O'Donnell F, Musso N, Lazzarino G, Grasso M, Amorini AM, Tascedda F, Bucolo C, Drago F, Tavazzi B, Lazzarino G, Lunte SM, Caraci F. Carnosine Decreases PMA-Induced Oxidative Stress and Inflammation in Murine Macrophages. Antioxidants (Basel) 2019; 8:E281. [PMID: 31390749 PMCID: PMC6720685 DOI: 10.3390/antiox8080281] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 02/06/2023] Open
Abstract
Carnosine is an endogenous dipeptide composed of β-alanine and L-histidine. This naturally occurring molecule is present at high concentrations in several mammalian excitable tissues such as muscles and brain, while it can be found at low concentrations in a few invertebrates. Carnosine has been shown to be involved in different cellular defense mechanisms including the inhibition of protein cross-linking, reactive oxygen and nitrogen species detoxification as well as the counteraction of inflammation. As a part of the immune response, macrophages are the primary cell type that is activated. These cells play a crucial role in many diseases associated with oxidative stress and inflammation, including atherosclerosis, diabetes, and neurodegenerative diseases. In the present study, carnosine was first tested for its ability to counteract oxidative stress. In our experimental model, represented by RAW 264.7 macrophages challenged with phorbol 12-myristate 13-acetate (PMA) and superoxide dismutase (SOD) inhibitors, carnosine was able to decrease the intracellular concentration of superoxide anions (O2-•) as well as the expression of Nox1 and Nox2 enzyme genes. This carnosine antioxidant activity was accompanied by the attenuation of the PMA-induced Akt phosphorylation, the down-regulation of TNF-α and IL-6 mRNAs, and the up-regulation of the expression of the anti-inflammatory mediators IL-4, IL-10, and TGF-β1. Additionally, when carnosine was used at the highest dose (20 mM), there was a generalized amelioration of the macrophage energy state, evaluated through the increase both in the total nucleoside triphosphate concentrations and the sum of the pool of intracellular nicotinic coenzymes. Finally, carnosine was able to decrease the oxidized (NADP+)/reduced (NADPH) ratio of nicotinamide adenine dinucleotide phosphate in a concentration dependent manner, indicating a strong inhibitory effect of this molecule towards the main source of reactive oxygen species in macrophages. Our data suggest a multimodal mechanism of action of carnosine underlying its beneficial effects on macrophage cells under oxidative stress and inflammation conditions.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Department of Laboratories, Oasi Research Institute-IRCCS, 94018 Troina, Italy.
| | - Claudia G Fresta
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
| | - Annamaria Fidilio
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Fergal O'Donnell
- School of Biotechnology, Dublin City University, D09W6Y4 Dublin, Ireland
| | - Nicolò Musso
- Bio-Nanotech Research and Innovation Tower (BRIT), University of Catania, 95125 Catania, Italy
| | - Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Margherita Grasso
- Department of Laboratories, Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Angela M Amorini
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy.
| | - Susan M Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
| | - Filippo Caraci
- Department of Laboratories, Oasi Research Institute-IRCCS, 94018 Troina, Italy
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| |
Collapse
|
43
|
The effect of fenofibrate, a PPARα activator on toll-like receptor-4 signal transduction in melanoma both in vitro and in vivo. Clin Transl Oncol 2019; 22:486-494. [PMID: 31175545 DOI: 10.1007/s12094-019-02150-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/29/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND The anti-cancer effect of peroxisome proliferator-activated receptor (PPAR) α ligands on growth and metastatic potential of melanoma cells has been shown previously. However, the mechanism underlying these effects remains to be elucidated. Here, we investigated the effects of fenofibrate (PPAR ligand) on Toll-like receptor-4 (TLR-4) signaling in mice melanoma. METHODS Mice melanoma cells (B16F10) were treated with fenofibrate or LPS or LPS + fenofibrate or pre-treated with CLI-095 (a TLR4 inhibitor), followed by fenofibrate. In in vivo model, C57BL/6 mice were subcutaneously injected with B16F10 cells (with/without LPS pre-treatment), and fenofibrate was administrated after development of palpable tumors. Cell proliferation, the expression level of Tlr4, Myd88, Nf-κb1 genes, TLR-4 protein expression, TNF-α levels, and tumor volume were measured. RESULT Our results indicated that fenofibrate significantly inhibited the Tlr-4, Myd-88, and Nf-kb1 mRNA expression and TNF-α concentration in B16F10 LPS-stimulated cells. In addition, blocking TLR-4 signaling increased the anti-inflammatory potential of fenofibrate. Also fenofibrate can reduce LPS-induced tumor volume, Tlr-4, Myd-88, Nf-kb1 mRNA, and TLR-4 protein expression in tumor tissue and also TNF-α level in tumor tissue lysate. CONCLUSION Our data indicate that fenofibrate may exert its anti-melanoma effects via interaction with TLR4-dependent signaling pathway (TLR-4/MyD-88/ NF-kB).
Collapse
|
44
|
Wang JS, Xiao WW, Zhong YS, Li XD, Du SX, Xie P, Zheng GZ, Han JM. Galectin-3 deficiency protects lipopolysaccharide-induced chondrocytes injury via regulation of TLR4 and PPAR-γ-mediated NF-κB signaling pathway. J Cell Biochem 2019; 120:10195-10204. [PMID: 30565728 DOI: 10.1002/jcb.28304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/28/2018] [Indexed: 02/05/2023]
Abstract
The aim of the present study was to identify the functional role of galectin-3 (Gal-3) in lipopolysaccharide (LPS)-induced injury in ATDC5 cells and to explore the probable molecular mechanisms. Here, we identified that LPS is sufficient to enhance the expression of Gal-3 in ATDC5 cells. In addition, repression of Gal-3 obviously impeded LPS-stimulated inflammation damage as exemplified by a reduction in the release of inflammatory mediators interleukin (IL)-1β, IL-6, and tumor necrosis factor-α, as well as the production of nitric oxide and prostaglandin E2 (PGE2) concomitant with the downregulation of matrix metalloproteinases (MMP)-13 and MMP-3 expression in ATDC5 cells after LPS administration. Moreover, ablation of Gal-3 dramatically augmented cell ability and attenuated cell apoptosis accompanied by an increase in the expression of antiapoptotic protein Bcl-2 and a decrease in the expression of proapoptotic protein Bax and caspase-3 in ATDC5 cells subjected with LPS. Importantly, we observed that forced expression of TLR4 or blocked PPAR-γ with the antagonist GW9662 effectively abolished Gal-3 inhibition-mediated anti-inflammatory and antiapoptosis effects triggered by LPS. Mechanistically, depletion of Gal-3 prevents the NF-κB signaling pathway. Taken together, these findings indicated that the absence of Gal-3 exerted chondroprotective properties dependent on TLR4 and PPAR-γ-mediated NF-κB signaling, indicating that Gal-3 functions as a protector in the development and progression of osteoarthritis.
Collapse
Affiliation(s)
- Jian-Sheng Wang
- Department of Orthopaedics Ward II, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Wei-Wei Xiao
- Department of Clinical Laboratory, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Yong-Sheng Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xue-Dong Li
- Department of Orthopaedics, the Third Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Shi-Xin Du
- Department of Orthopaedics, the Third Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Peng Xie
- Department of Orthopaedics, the Third Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Gui-Zhou Zheng
- Department of Orthopaedics, the Third Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Jing-Ming Han
- Department of Orthopaedics Ward II, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
45
|
Rao Z, Cao H, Shi B, Liu X, Luo J, Zeng N. Inhibitory Effect of Jing-Fang Powder n-Butanol Extract and Its Isolated Fraction D on Lipopolysaccharide-Induced Inflammation in RAW264.7 Cells. J Pharmacol Exp Ther 2019; 370:62-71. [DOI: 10.1124/jpet.118.255893] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 04/16/2019] [Indexed: 01/08/2023] Open
|
46
|
Li KX, Du Q, Wang HP, Sun HJ. Death-associated protein kinase 3 deficiency alleviates vascular calcification via AMPK-mediated inhibition of endoplasmic reticulum stress. Eur J Pharmacol 2019; 852:90-98. [PMID: 30851272 DOI: 10.1016/j.ejphar.2019.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 12/11/2022]
Abstract
Vascular calcification (VC) is a critical feature of chronic kidney disease (CKD), diabetes, hypertension, and atherosclerosis. Death-associated protein kinase 3 (DAPK3) is involved in vascular remodeling in hypertension. However, it remains to be clarified whether DAPK3 controls vascular smooth muscle cell (VSMC) phenotypic transition into an osteogenic cell phenotype, which is an important process for VC. In vivo VC was induced in rats by vitamin D3 and nicotine. VSMCs were incubated with calcifying media containing β-glycerophosphate and Ca2+ to induce VC in vitro. Herein, we demonstrated increased expression of DAPK3 in the aortas of VC rats and VSMCs cultured in calcifying media. Knockdown of DAPK3 significantly inhibited calcifying media-induced VSMC mineralization and retarded the phenotypic transformation of VSMCs into osteogenic cells. Silencing of DAPK3 suppressed endoplasmic reticulum stress (ERS) related protein expressions, but upregulated the phosphorylation level of AMP-activated protein kinase (AMPK) in calcified VSMCs. Moreover, pretreatment with AMPK inhibitor Compound C abolished DAPK3 shRNA-mediated inhibition of ERS in VSMCs. In vivo, DAPK inhibitor significantly prevented calcium deposition in the aortas of VC rats. The present results revealed that DAPK3 modulated VSMC calcification through AMPK-mediated ERS signaling.
Collapse
Affiliation(s)
- Ke-Xue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Qiong Du
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, PR China
| | - Hui-Ping Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
47
|
Mirza AZ, Althagafi II, Shamshad H. Role of PPAR receptor in different diseases and their ligands: Physiological importance and clinical implications. Eur J Med Chem 2019; 166:502-513. [DOI: 10.1016/j.ejmech.2019.01.067] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 12/15/2022]
|
48
|
Dana N, Vaseghi G, Haghjooy Javanmard S. Crosstalk between Peroxisome Proliferator-Activated Receptors and Toll-Like Receptors: A Systematic Review. Adv Pharm Bull 2019; 9:12-21. [PMID: 31011554 PMCID: PMC6468223 DOI: 10.15171/apb.2019.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/08/2019] [Accepted: 01/12/2019] [Indexed: 12/11/2022] Open
Abstract
As one of the four major families of pattern recognition receptors (PRRs), toll like receptors (TLRs)
are crucial and important components of the innate immune system. Peroxisome proliferatoractivated
receptors (PPARs) with three isoforms are transcription factors classified as a subfamily
of nuclear receptor proteins, and are of significant regulatory activity in cellular differentiation,
development, metabolism, and tumorigenesis. It is well established that PPARs agonists display
anti-inflammatory effects through inhibition of the nuclear factor-kappa B (NF-κB) pathway, a
key regulator of immune and inflammatory responses, in a sense that TLRs signaling pathways
are mainly toward activation of NF-κB. Through a systematic review of previous studies, we
aimed to address and clarify the reciprocal interaction between TLRs and PPARs in hope to find
alternative therapeutic approaches for inflammatory diseases. Among the available scientific
database, 31 articles were selected for this review. A comprehensive review of this database
confirms the presence of a cross-talk between PPARs and TLRs, indicating that not only
PPARs stimulation may affect the expression level of TLRs via several mechanisms leading to
modulating TLRs activities, but also TLRs have the potential to moderate the expression of PPARs.
We, therefore, conclude that, as a key regulator of the innate immune system, the interaction
between PPARs and TLRs is a potential therapeutic target in disease treatment.
Collapse
Affiliation(s)
- Nasim Dana
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical sciences, Isfahan, Iran.,Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
49
|
Yi J, Gao ZF. MicroRNA-9-5p promotes angiogenesis but inhibits apoptosis and inflammation of high glucose-induced injury in human umbilical vascular endothelial cells by targeting CXCR4. Int J Biol Macromol 2019; 130:1-9. [PMID: 30716366 DOI: 10.1016/j.ijbiomac.2019.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023]
Abstract
High glucose (HG) has the potential to cause vascular endothelial cell injury, while microRNAs (miRNAs) play a key role in treating endothelial cell injury. CXC chemokine receptor-4 (CXCR4) is reported to be expressed in vascular endothelial cells. Hence, this study investigated role of miR-9-5p in the angiogenesis and apoptosis of HG-induced human umbilical vascular endothelial cells (HUVECs) injury. Dual luciferase reporter gene assay verified that miR-9-5p targeted CXCR4. RT-qPCR and western blot analysis revealed that miR-9-5p was down-regulated, meanwhile CXCR4 was up-regulated in the HG-induced HUVECs. HUVECs were cultured in 30 mmol/L HG in vitro, and then transfected with miR-9-5p mimic or CXCR4 siRNA to identify the effect of miR-9-5p on cell activity, angiogenesis, apoptosis, and inflammation of HG-induced HUVECs. The results suggested that overexpression of miR-9-5p or silencing of CXCR4 in HG-induced HUVECs increased cell proliferation and tubule length, while decreasing the apoptosis rate and the expression of inflammatory factors. Furthermore, miR-9-5p inhibited the phosphorylation of extracellular regulated protein kinases (ERK), protein kinase B (AKT), and Mammalian Target of Rapamycin (mTOR) proteins via downregulation of CXCR4. Therefore, overexpression of miR-9-5p suppressed the mitogen-activated protein kinase (MAPK)/ERK and the PI3K/AKT/mTOR pathway by inhibiting CXCR4, thereby reducing HG-induced injury in HUVECs.
Collapse
Affiliation(s)
- Jun Yi
- Department of Anesthesiology, Beijing Jishuitan Hospital, Beijing 100035, PR China
| | - Zhi-Feng Gao
- Department of Anesthesiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, PR China.
| |
Collapse
|
50
|
Zeng ZS, Lin J, Xu CB, Cao L, Chen C, Li J. Minimally modified low-density lipoprotein upregulates the ET B and α 1 receptors in mouse mesenteric arteries in vivo by activating the PI3K/Akt pathway. J Pharm Pharmacol 2019; 71:937-944. [PMID: 30663067 DOI: 10.1111/jphp.13069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The current study aimed to explore whether minimally modified low-density lipoprotein (mmLDL) via tail vein injection upregulates the ETB and α1 receptors in mouse mesenteric arteries by activating the PI3K/Akt pathway. METHODS The contraction curves of the mesenteric arteries caused by sarafotoxin 6c (S6c, ETB receptor agonist) and phenylephrine (PE, α1 receptor agonist) were measured by a myograph system. Serum oxLDL was detected using enzyme-linked immunosorbent assays. The levels of the ETB receptor, the α1 receptor, PI3K, p-PI3K and p-Akt were detected using real-time polymerase chain reaction and Western blot analyses. KEY FINDINGS Minimally modified low-density lipoprotein noticeably enhanced the contraction effect curves of S6c and PE, with significantly increased Emax values (P < 0.01), compared to those of the control group. This treatment significantly increased the mRNA expression and protein levels of the ETB and α1 receptors and the protein levels of p-PI3K and p-Akt in the vessel wall (P < 0.01). LY294002 inhibited the effect of mmLDL. CONCLUSIONS An increase in mmLDL activated the PI3K/Akt pathway, which upregulated the expression of the ETB and α1 receptors and enhanced the ETB and α1- receptor-mediated contractile function.
Collapse
Affiliation(s)
- Zhong-San Zeng
- Institute of Pharmacy and Pharmacology, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Jie Lin
- Institute of Pharmacy and Pharmacology, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Cang-Bao Xu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Lei Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chen Chen
- Institute of Pharmacy and Pharmacology, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Jie Li
- Institute of Pharmacy and Pharmacology, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| |
Collapse
|