1
|
Lai S, Ye Y, Ding Q, Hu X, Fu A, Wu L, Cao W, Liu Q, Dou X, Qi X. Thonningianin A ameliorates acetaminophen-induced liver injury by activating GPX4 and modulating endoplasmic reticulum stress. Front Pharmacol 2025; 16:1531277. [PMID: 40051561 PMCID: PMC11882853 DOI: 10.3389/fphar.2025.1531277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Acetaminophen (APAP) is widely used as an analgesic and antipyretic. However overdose APAP can lead to acute liver injury (ALI), representing a significant challenge for public health due to limited treatment options. Current research highlights the need for safer and more effective therapies for APAP-induced liver injury, especially those that target oxidative and endoplasmic reticulum (ER) stress pathways. This study investigates the protective effects of Thonningianin A (TA), a flavonoid compound derived from Penthorum chinense Pursh, in mitigating APAP-induced hepatotoxicity. Methods The experimental design involved administering TA at doses of 20 mg/kg and 40 mg/kg to C57BL/6 mice prior to inducing hepatotoxicity with APAP. Results and discussion TA treatment significantly lowered plasma ALT and AST levels, inhibited the production of inflammatory cytokines, and reduced oxidative stress markers in liver tissues. Furthermore, TA modulated apoptosis-related proteins by increasing BCL-2 expression while decreasing CHOP and BAX levels. It alleviated endoplasmic reticulum (ER) stress by downregulating GRP78, p-PERK, and ATF4. Notably, liver-specific GPX4 knockdown, achieved through AAV-8-mediated shRNA delivery, abolished the hepatoprotective effects of TA, underscoring GPX4's essential role in mediating TA-induced hepatoprotection. These findings suggest TA as a promising therapeutic agent in managing APAP-induced liver injury, with its unique action on both oxidative and ER stress pathways contributing to its hepatoprotective efficacy.
Collapse
Affiliation(s)
- Shanglei Lai
- Department of Medical Research Center, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Yingyan Ye
- Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaokai Hu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ai Fu
- Institute of Hepatology and Epidemiology, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lan Wu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenjing Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qingsheng Liu
- Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurosurgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
2
|
Li YM, Yan MM, Luo T, Zhu W, Jiang JG. Comparative hepatoprotective effects of flavonoids-rich fractions from flowers and leaves of Penthorum chinense Pursh in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118960. [PMID: 39426574 DOI: 10.1016/j.jep.2024.118960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Penthorum chinense Pursh is a traditional Miao ethnomedicine rich in bioactive components, widely recognized for its hepatoprotective properties. However, the hepatoprotective effects of its flowers and leaves have not been individually elucidated. AIMS OF THE STUDY The objective of this study was to isolate and purify flavonoids-rich fractions from the flowers (PFF) and leaves (PLF) of P. chinense, and to assess their potential protective effects against oxidative, alcohol-induced, and free fatty acid (FFA) induced injury in hepatic cells. MATERIALS AND METHODS The P. chinense flowers and leaves flavonoids-rich fractions were extracted by the method optimized by response surface methodology, and the extracts were subsequently purified using petroleum ether and microporous column. The physical characteristics and component composition of PFF and PLF were analyzed by FT-IR and UPLC-MS/MS. The hepatoprotective activities of PFF and PLF were evaluated by the alcohol, H2O2, and FFA-induced hepatocyte injury cellular model in vitro. The protective effects of PFF and PLF on the hepatic cells were evaluated by assessing cell apoptosis rate, enzymes activities, mitochondrial membrane potential, and mRNA expression in relevant signaling pathways. RESULTS The results revealed that PFF was mainly composed of pinocembrin, quercitrin and quercetin, while PLF was predominantly composed of quercetin, pinocembrin, and kaempferol and their derivatives. PFF and PLF exhibited distinct effects on increasing the cell proliferation rate, regulating the MDA, GOT and GPT levels, and modulating the mRNA expression in apoptosis and antioxidant pathways in alcohol damaged LO2 cells. PFF exhibited superior efficacy in reducing cell apoptosis in alcohol-damage cells compared to PLF. Both PFF and PLF alleviated mitochondrial stress in H2O2-induced LO2 cells. Additionally, the PFF and PLF attenuated lipid accumulation and activated mRNA expressions in PPARα/ACOX1/CPT-1 lipid metabolism pathways, as well as Nrf2/ARE oxidative stress pathways. CONCLUSION This study compared the hepatoprotective activities of flavonoids-rich fractions purified from the flowers and leaves of P. chinense. The results contribute to the enhanced development and utilization of various parts of P. chinense aimed at medical and health food applications.
Collapse
Affiliation(s)
- Yi-Meng Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; Dermatology Hospital of Southern Medical University, Guangzhou, 510091, China; The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Mao-Mao Yan
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Ting Luo
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Wei Zhu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, 510120, China.
| | - Jian-Guo Jiang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
3
|
Lin Q, Zhu F, Shiye A, Liu R, Wang X, Ye Z, Ding Y, Sun X, Ma Y. Antimicrobial Effects of Thonningianin a (TA)-Loaded Chitosan Nanoparticles Encapsulated by a PF-127 hydrogel in Diabetic Wound Healing. Int J Nanomedicine 2024; 19:12835-12850. [PMID: 39651354 PMCID: PMC11624675 DOI: 10.2147/ijn.s488115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/12/2024] [Indexed: 12/11/2024] Open
Abstract
Background and Purpose Diabetic wounds are serious chronic complications of diabetes and can lead to amputation and death. Although considerable progress has been made in drugs and materials for treating it, it's still an urgent clinical problem as the materials and drugs have potential therapeutic drawbacks, such as low delivery efficiency and poor tissue permeability. To promote diabetic wound healing, a composite of thonningianin A (TA)-loaded chitosan nanoparticles (CNPS) encapsulated by a Pluronic F-127 (PF-127) hydrogel (TA-CNPS-PF) was developed in this study. Methods TA-CNPS was prepared by ionic gelation method and TA-CNPS was thoroughly dispersed into PF-127 hydrogel to prepare TA-CNPS-PF. The particle size, hydrogel structure, encapsulation ratio, release ratio, antimicrobial properties of TA-CNPS-PF were determined and the effect of TA-CNPS-PF on diabetic wounds was assessed. The effect of TA on macrophage polarization was also examined in vitro. Results The particle size was approximately 100 nm of TA-CNPS-PF and the hydrogel had a homogeneous three-dimensional reticulation structure. The encapsulation efficiency of TA in the CNPS were 99.3% and the release ratio of TA-CNPS-PF was approximately 86% and has antimicrobial properties. TA-CNPS-PF promoted diabetic wound healing significantly. Histopathology confirmed that TA-CNPS-PF promoted complete re-epithelialization and adequate collagen deposition. TA promoted the polarization of M1 macrophages into M2 macrophages via light microscopy, immunocytometry and flow cytometry. TA-CNPS-PF also promoted an increase in the number of M2 macrophages in diabetic wounds. Conclusion TA promotes diabetic wound healing by promoting the polarization of M1 macrophages into M2 macrophages and TA-CNPS-PF has good antimicrobial activity and a good drug release ratio in this study, which provides a new direction for the treatment of diabetic wounds and is expected to be highly advantageous in clinical diabetes wound therapy.
Collapse
Affiliation(s)
- Qian Lin
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Fucheng Zhu
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Aji Shiye
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Runyu Liu
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Hepatobiliary Pancreatic Vascular Surgery, the Second Affiliated Hospital of Chengdu Medical College, Chengdu, 610057, People’s Republic of China
| | - Xiaolan Wang
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Zi Ye
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yinhuan Ding
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Sichuan, 646000, People’s Republic of China
| | - Xiaolei Sun
- Department of General Surgery (Vascular Surgery), the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Interventional Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Laboratory of Nucleic Acids in Medicine for National High-Level Talent, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, People’s Republic of China
- School of Cardiovascular Medicine and Sciences, King’s College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King’s College London, London, SE5 9NU, UK
| | - Yarong Ma
- Department of Ophthalmology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
4
|
Wang XX, Ji X, Lin J, Wong IN, Lo HH, Wang J, Qu L, Wong VKW, Chung SK, Law BYK. GPCR-mediated natural products and compounds: Potential therapeutic targets for the treatment of neurological diseases. Pharmacol Res 2024; 208:107395. [PMID: 39241934 DOI: 10.1016/j.phrs.2024.107395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/01/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
G protein-coupled receptors (GPCRs), widely expressed in the human central nervous system (CNS), perform numerous physiological functions and play a significant role in the pathogenesis of diseases. Consequently, identifying key therapeutic GPCRs targets for CNS-related diseases is garnering immense interest in research labs and pharmaceutical companies. However, using GPCRs drugs for treating neurodegenerative diseases has limitations, including side effects and uncertain effective time frame. Recognizing the rich history of herbal treatments for neurological disorders like stroke, Alzheimer's disease (AD), and Parkinson's disease (PD), modern pharmacological research is now focusing on the understanding of the efficacy of traditional Chinese medicinal herbs and compounds in modulating GPCRs and treatment of neurodegenerative conditions. This paper will offer a comprehensive, critical review of how certain natural products and compounds target GPCRs to treat neurological diseases. Conducting an in-depth study of herbal remedies and their efficacies against CNS-related disorders through GPCRs targeting will augment our strategies for treating neurological disorders. This will not only broaden our understanding of effective therapeutic methodologies but also identify the root causes of altered GPCRs signaling in the context of pathophysiological mechanisms in neurological diseases. Moreover, it would be informative for the creation of safer and more effective GPCR-mediated drugs, thereby establishing a foundation for future treatment of various neurological diseases.
Collapse
Affiliation(s)
- Xing Xia Wang
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China; Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiang Ji
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China
| | - Jing Lin
- Department of Endocrinology, Luzhou Hospital of Traditional Chinese Medicine, Luzhou, Sichuan, China
| | - Io Nam Wong
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR China
| | - Hang Hong Lo
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China
| | - Jian Wang
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Liqun Qu
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China
| | - Vincent Kam Wai Wong
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China
| | - Sookja Kim Chung
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China; Faculty of Medicine, Macau University of Science and Technology, Macau SAR China.
| | - Betty Yuen Kwan Law
- Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR China.
| |
Collapse
|
5
|
Yu L, Wang H, Yao Q, Li K, Qu L, Tang B, Zeng W, Qiao G, Tang Y, Hu G, Hu G, Wong VKW, Wang Q, Qin D, Wu J, Zhou X, Sun X, Law BYK, Wu A. Thonningianin A from Penthorum chinense Pursh as a targeted inhibitor of Alzheimer's disease-related β-amyloid and Tau proteins. Phytother Res 2024; 38:4815-4831. [PMID: 39225174 DOI: 10.1002/ptr.8060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 09/04/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by complex pathogenesis mechanisms. Among these, β-amyloid plaques and hyperphosphorylated Tau protein tangles have been identified as significant contributors to neuronal damage. This study investigates thonningianin A (TA) from Penthorum chinense Pursh (PCP) as a potential inhibitor targeting these pivotal proteins in AD progression. The inhibitory potential of PCP and TA on Aβ fibrillization was initially investigated. Subsequently, ultra-high performance liquid chromatography-quadrupole time-of-flight tandem mass spectrometry and biolayer interferometry were employed to determine TA's affinity for both Aβ and Tau. The inhibitory effects of TA on the levels and cytotoxicity of AD-related proteins were then assessed. In 3xTg-AD mice, the therapeutic potential of TA was evaluated. Additionally, the molecular interactions between TA and either Aβ or Tau were explored using molecular docking. We found that PCP-total ethanol extract and TA significantly inhibited Aβ fibrillization. Additionally, TA demonstrated strong affinity to Aβ and Tau, reduced levels of amyloid precursor protein and Tau, and alleviated mitochondrial distress in PC-12 cells. In 3xTg-AD mice, TA improved cognition, reduced Aβ and Tau pathology, and strengthened neurons. Moreover, molecular analyses revealed efficient binding of TA to Aβ and Tau. In conclusion, TA, derived from PCP, shows significant neuroprotection against AD proteins, highlighting its potential as an anti-AD drug candidate.
Collapse
Affiliation(s)
- Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
- Department of Chemistry, School of Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Huimiao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Qianfang Yao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Keru Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Liqun Qu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Bin Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Wu Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Gan Qiao
- Nucleic Acid Medicine of Luzhou Key Laboratory, Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Guishan Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Guangqiang Hu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Qiong Wang
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiaolei Sun
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Zhang S, Zhang S, Bai X, Wang Y, Liu Y, Liu W. Thonningianin A ameliorated renal interstitial fibrosis in diabetic nephropathy mice by modulating gut microbiota dysbiosis and repressing inflammation. Front Pharmacol 2024; 15:1389654. [PMID: 39193336 PMCID: PMC11347433 DOI: 10.3389/fphar.2024.1389654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Objectives This study was conducted to examine the potential health benefits of thonningianin A (TA) on renal injury and interstitial fibrosis in diabetic nephropathy (DN) mice. Methods In this study, a DN mice model was established using male C57BL/6 mice injected with streptozotocin (STZ, 50 mg/kg) intraperitoneally and treated with TA for 12 weeks. Firstly, the therapeutic and anti-fibrotic effects of TA on DN were evaluated. Secondly, the effect of TA on renal inflammation was evaluated and Western blot was used to detect the changes of NLRP3/ASC/Caspase-1 pathway-related protein expressions in kidney. Furthermore, the effect of TA on impairments in the intestinal mucosa barrier was evaluated and the changes of lipopolysaccharide (LPS) levels in feces and serum were detected by ELISA. Finally, 16S rRNA sequencing was used to detect alteration of gut microbiota diversity and abundance in mice after TA treatment. Results The results showed that TA markedly mitigated blood glucose (Glu), decreased 24-h urinary total protein (24hUTP), improved renal dysfunction and kidney index (KI) in DN mice. Furthermore, TA significantly alleviated renal injury and interstitial fibrosis, repressing renal inflammation. Western blot results showed that the NLRP3/ASC/Caspase-1 signaling pathway-related proteins decreased after TA treatment. In addition, TA also ameliorated impairments in the intestinal mucosa barrier and restored the expressions of intestinal tight junction proteins (Claudin-1, Occludin and ZO-1). Subsequently, it reduced LPS levels of DN mice in fecal and serum. Furthermore, 16S rRNA high-throughput sequencing showed that TA modulated gut microbiota dysbiosis and decreased the abundance of Gram-negative bacteria (Proteobacteria and Escherichia-Shigella). Conclusion This study suggested that TA might exert a beneficial effect on renal interstitial fibrosis in DN mice by modulating gut microbiota dysbiosis, ameliorating impairments in the intestinal mucosa barrier, reducing the production and release of LPS, inhibiting the activation of NLRP3/ASC/Caspase-1 signaling pathway, and repressing renal inflammatory.
Collapse
Affiliation(s)
- Shujiao Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shuaixing Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xuehui Bai
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yaoxian Wang
- Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuning Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Renal Research Institution of Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
7
|
Yong YY, Yan L, Wang BD, Fan DS, Guo MS, Yu L, Wu JM, Qin DL, Law BYK, Wong VKW, Yu CL, Zhou XG, Wu AG. Penthorum chinense Pursh inhibits ferroptosis in cellular and Caenorhabditis elegans models of Alzheimer's disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155463. [PMID: 38452694 DOI: 10.1016/j.phymed.2024.155463] [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: 06/23/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Ferroptosis, a unique type of cell death triggered by iron-dependent lipid peroxidation, plays a critical role in the pathogenesis of Alzheimer's disease (AD), a debilitating condition marked by memory loss and cognitive impairment due to the accumulation of beta-amyloid (Aβ) and hyperphosphorylated Tau protein. Increasing evidence suggests that inhibitors of ferroptosis could be groundbreaking in the treatment of AD. METHOD In this study, we established in vitro ferroptosis using erastin-, RSL-3-, hemin-, and iFSP1-induced PC-12 cells. Using MTT along with Hoechst/PI staining, we assessed cell viability and death. To determine various aspects of ferroptosis, we employed fluorescence probes, including DCFDA, JC-1, C11 BODIPY, Mito-Tracker, and PGSK, to measure ROS production, mitochondrial membrane potential, lipid peroxidation, mitochondrial morphology, and intracellular iron levels. Additionally, Western blotting, biolayer interferometry technology, and shRNA were utilized to investigate the underlying molecular mechanisms. Furthermore, p-CAX APP Swe/Ind- and pRK5-EGFP-Tau P301L overexpressing PC-12 cells, along with Caenorhabditis elegans (C. elegans) strains CL4176, CL2331, and BR5270, were employed to examine ferroptosis in AD models. RESULTS Here, we conducted a screening of our natural medicine libraries and identified the ethanol extract of Penthorum chinense Pursh (PEE), particularly its ethyl acetate fraction (PEF), displayed inhibitory effects on ferroptosis in cells. Specifically, PEF inhibited the generation of ROS, lipid peroxidation, and intracellular iron levels. Furthermore, PEF demonstrated protective effects against H2O2-induced cell death, ROS production, and mitochondrial damage. Mechanistic investigations unveiled PEF's modulation of intracellular iron accumulation, GPX4 expression and activity, and FSP1 expression. In p-CAX APP Swe/Ind and pRK5-EGFP-Tau P301L overexpressing PC-12 cells, PEF significantly reduced cell death, as well as ROS and lipid peroxidase production. Moreover, PEF ameliorated paralysis and slowing rate in Aβ and Tau transgenic C. elegans models, while inhibiting ferroptosis, as evidenced by decreased DHE intensity, lipid peroxidation levels, iron accumulation, and expression of SOD-3 and gst-4. CONCLUSION Our findings highlight the suppressive effects of PEF on ferroptosis in AD cellular and C. elegans models. This study helps us better understand how ferroptosis affects AD and emphasizes the potential of PCP as a candidate for AD intervention.
Collapse
Affiliation(s)
- Yuan-Yuan Yong
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Bin-Ding Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Dong-Sheng Fan
- Department of Pharmacy, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Gui Yang, 550000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 99078, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau 99078, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
8
|
Zhu F, Wang B, Qin D, Su X, Yu L, Wu J, Law BY, Guo M, Yu C, Zhou X, Wu A. Carpesii fructus extract exhibits neuroprotective effects in cellular and Caenorhabditis elegans models of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14515. [PMID: 37905594 PMCID: PMC11017466 DOI: 10.1111/cns.14515] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/05/2023] [Accepted: 10/14/2023] [Indexed: 11/02/2023] Open
Abstract
OBJECTIVE Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra. Despite extensive research, no definitive cure or effective disease-modifying treatment for PD exists to date. Therefore, the identification of novel therapeutic agents with neuroprotective properties is of utmost importance. Here, we aimed to investigate the potential neuroprotective effects of Carpesii fructus extract (CFE) in both cellular and Caenorhabditis elegans (C. elegans) models of PD. METHODS The neuroprotective effect of CFE in H2O2- or 6-OHDA-induced PC-12 cells and α-synuclein-overexpressing PC-12 cells were investigated by determining the cell viability, mitochondrial damage, reactive oxygen species (ROS) production, apoptosis, and α-synuclein expression. In NL5901, BZ555, and N2 worms, the expression of α-synuclein, motive ability, the viability of dopaminergic neurons, lifespan, and aging-related phenotypes were investigated. The signaling pathway was detected by Western blotting and validated by employing small inhibitors and RNAi bacteria. RESULTS In cellular models of PD, CFE significantly attenuated H2O2- or 6-OHDA-induced toxicity, as evidenced by increased cell viability and reduced apoptosis rate. In addition, CFE treatment suppressed ROS generation and restored mitochondrial membrane potential, highlighting its potential as a mitochondrial protective agent. Furthermore, CFE reduced the expression of α-synuclein in wide type (WT)-, A53T-, A30P-, or E46K-α-synuclein-overexpressing PC-12 cells. Our further findings reveal that CFE administration reduced α-synuclein expression and improved its induced locomotor deficits in NL5901 worms, protected dopaminergic neurons against 6-OHDA-induced degeneration in BZ555 worms, extended lifespan, delayed aging-related phenotypes, and enhanced the ability of stress resistance in N2 worms. Mechanistic studies suggest that the neuroprotective effects of CFE may involve the modulation of the MAPK signaling pathway, including ERK, JNK, and p38, whereas the interference of these pathways attenuated the neuroprotective effect of CFE in vitro and in vivo. CONCLUSION Overall, our study highlights the potential therapeutic value of CFE as a neuroprotective agent in the context of PD. Furthermore, elucidation of the active compounds of CFE will provide valuable insights for the development of novel therapeutic strategies for PD.
Collapse
Affiliation(s)
- Feng‐Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Bin‐Ding Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Da‐Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Xiao‐Hui Su
- Institute of Chinese Materia Medica, China Academy of Chinese Medical SciencesBeijingChina
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Jian‐Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Betty Yuen‐Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and TechnologyTaipaChina
| | - Min‐Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Chong‐Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - Xiao‐Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| | - An‐Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of EducationSchool of Pharmacy, Southwest Medical UniversityLuzhouChina
| |
Collapse
|
9
|
Wang Z, Zhou J, Pan J, Cheng W, Fang J, Lv Q, Lin X, Cheng W, Zhang L, Cheng K. Insights into the Superrosids phylogeny and flavonoid synthesis from the telomere-to-telomere gap-free genome assembly of Penthorum chinense Pursh. HORTICULTURE RESEARCH 2024; 11:uhad274. [PMID: 38344651 PMCID: PMC10857932 DOI: 10.1093/hr/uhad274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/11/2023] [Indexed: 10/28/2024]
Abstract
The completion of the first telomere-to-telomere (T2T) genome assembly of Penthorum chinense Pursh (PC), a prominent medicinal plant in China, represents a significant achievement. This assembly spans a length of 257.5 Mb and consists of nine chromosomes. PC's notably smaller genome size in Saxifragales, compared to that of Paeonia ostii, can be attributed to the low abundance of transposable elements. By utilizing single-copy genes from 30 species, including 28 other Superrosids species, we successfully resolved a previously debated Superrosids phylogeny. Our findings unveiled Saxifragales as the sister group to the core rosids, with both being the sister group to Vitales. Utilizing previously characterized cytochrome P450 (CYP) genes, we predicted the compound classes that most CYP genes of PC are involved in synthesizing, providing insight into PC's potential metabolic diversity. Metabolomic and transcriptomic data revealed that the richest sources of the three most noteworthy medicinal components in PC are young leaves and flowers. We also observed higher activity of upstream genes in the flavonoid synthesis pathway in these plant parts. Additionally, through weighted gene co-expression network analysis, we identified gene regulatory networks associated with the three medicinal components. Overall, these findings deepen our understanding of PC, opening new avenues for further research and exploration.
Collapse
Affiliation(s)
- Zhoutao Wang
- Genomics and Genetic Engineering Laboratory of Ornamental Plants, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 311300, China
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
- Product Development Department, Zhejiang Shaowei Yuanzhi Science and Technology Development Co., Ltd, Lishui 323000, China
| | - Junmei Zhou
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Junjie Pan
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Wei Cheng
- Genomics and Genetic Engineering Laboratory of Ornamental Plants, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 311300, China
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Jie Fang
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Qundan Lv
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Xiaodan Lin
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, 311300, China
| | - Wenliang Cheng
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
| | - Liangsheng Zhang
- Genomics and Genetic Engineering Laboratory of Ornamental Plants, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 311300, China
| | - Kejun Cheng
- Postdoctoral Research Enter, Zhejiang Kangning Pharmaceutical Co., Ltd, Lishui 323000, China
- Product Development Department, Zhejiang Shaowei Yuanzhi Science and Technology Development Co., Ltd, Lishui 323000, China
- Zhejiang Provincial Key Laboratory of Resources Protection and Innovation of Traditional Chinese Medicine, Zhejiang A&F University, Hangzhou, 311300, China
| |
Collapse
|
10
|
Shen J, Zhang C, Liu Y, Zhao M, Wang Q, Li P, Liu R, Wai Wong VK, Zhang C, Sun X. L-type calcium ion channel-mediated activation of autophagy in vascular smooth muscle cells via thonningianin A (TA) alleviates vascular calcification in type 2 diabetes mellitus. Eur J Pharmacol 2023; 959:176084. [PMID: 37806540 DOI: 10.1016/j.ejphar.2023.176084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/10/2023]
Abstract
Vascular calcification (VC) is associated with increased morbidity and mortality, especially among people with type 2 diabetes mellitus (T2DM). The pathogenesis of vascular calcification is incompletely understood, and until now, there have been no effective therapeutics for vascular calcification. The L-type calcium ion channel in the cell membrane is vital for Ca2+ influx. The effect of L-type calcium ion channels on autophagy remains to be elucidated. Here, the natural compound thonningianin A (TA) was found to ameliorate vascular calcification in T2DM via the activation of L-type calcium ion channels. The results showed that TA had a concentration-dependent ability to decrease the transcriptional and translational expression of the calcification-related proteins runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2) and osteopontin (OPN) (P < 0.01) via ATG7-dependent autophagy in β-glycerophosphate (β-GP)- and high glucose (HG)-stimulated primary mouse aortic smooth muscle cells (MASMCs) and alleviate aortic vascular calcification in VitD3-stimulated T2DM mice. However, nifedipine, an inhibitor of L-type calcium ion channels, reversed TA-induced autophagy and Ca2+ influx in MASMCs. Molecular docking analysis revealed that TA was located in the hydrophobic pocket of Cav1.2 α1C and was mainly composed of the residues Ile, Phe, Ala and Met, which confirmed the efficacy of TA in targeting the L-type calcium channel of Cav1.2 on the cell membrane. Moreover, in an in vivo model of vascular calcification in T2DM mice, nifedipine reversed the protective effects of TA on aortic calcification and the expression of the calcification-related proteins RUNX2, BMP2 and OPN (P < 0.01). Collectively, the present results reveal that the activation of cell membrane L-type calcium ion channels can induce autophagy and ameliorate vascular calcification in T2DM. Thonningianin A (TA) can target and act as a potent activator of L-type calcium ion channels. Thus, this research revealed a novel mechanism for autophagy induction via L-type calcium ion channels and provided a potential therapeutic for vascular calcification in T2DM.
Collapse
Affiliation(s)
- Jialing Shen
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Vascular Surgery, The First People's Hospital of Yibin, Yibin, 644000, China
| | - Cheng Zhang
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, PR China
| | - Qianqian Wang
- Medical College, Dalian University, Dalian, 116622, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Runyu Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chunxiang Zhang
- Laboratory of Nucleic Acids in Medicine for National High-level Talents, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Xiaolei Sun
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Laboratory of Nucleic Acids in Medicine for National High-level Talents, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China; Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China; School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London, SE5 9NU, United Kingdom.
| |
Collapse
|
11
|
Yang K, Zeng L, Zeng J, Deng Y, Wang S, Xu H, He Q, Yuan M, Luo Y, Ge A, Ge J. Research progress in the molecular mechanism of ferroptosis in Parkinson's disease and regulation by natural plant products. Ageing Res Rev 2023; 91:102063. [PMID: 37673132 DOI: 10.1016/j.arr.2023.102063] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 09/08/2023]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder of the central nervous system after Alzheimer's disease. The current understanding of PD focuses mainly on the loss of dopamine neurons in the substantia nigra region of the midbrain, which is attributed to factors such as oxidative stress, alpha-synuclein aggregation, neuroinflammation, and mitochondrial dysfunction. These factors together contribute to the PD phenotype. Recent studies on PD pathology have introduced a new form of cell death known as ferroptosis. Pathological changes closely linked with ferroptosis have been seen in the brain tissues of PD patients, including alterations in iron metabolism, lipid peroxidation, and increased levels of reactive oxygen species. Preclinical research has demonstrated the neuroprotective qualities of certain iron chelators, antioxidants, Fer-1, and conditioners in Parkinson's disease. Natural plant products have shown significant potential in balancing ferroptosis-related factors and adjusting their expression levels. Therefore, it is vital to understand the mechanisms by which natural plant products inhibit ferroptosis and relieve PD symptoms. This review provides a comprehensive look at ferroptosis, its role in PD pathology, and the mechanisms underlying the therapeutic effects of natural plant products focused on ferroptosis. The insights from this review can serve as useful references for future research on novel ferroptosis inhibitors and lead compounds for PD treatment.
Collapse
Affiliation(s)
- Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Academy of Chinese Medicine, Changsha, Hunan, China.
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Deng
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hao Xu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qi He
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Mengxia Yuan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou University Medical College, Shantou, China
| | - Yanfang Luo
- The Central Hospital of Shaoyang, Shaoyang, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Academy of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
12
|
Lu M, Ruan J, Yu R, Zhang Y, Zhao W, Yang D, Wang W, Zhang Y, Wang T. Neolignan derivatives from Penthorum chinense with antitumor activity in human colorectal cancer cells by regulating Wnt/β-catenin signaling pathway. PHYTOCHEMISTRY 2023; 214:113827. [PMID: 37595774 DOI: 10.1016/j.phytochem.2023.113827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
In vitro cytotoxicity-guided isolation based on a MTT assay was conducted for Penthorum chinense Pursh. (Penthoraceae). In the active components (EtOAc extract of P. chinense), eight undescribed neolignans, penthoneolignans A-H (1-8), and two known analogs (9 and 10) were obtained and identified. Their absolute configurations were determined by experimental and computational comparison of electronic circular dichroism spectra analysis. The MTT experiment results of the obtained neolignans on HT29 and LoVo cells indicated previously undescribed neolignans, penthoneolignans A (1) and F (6), showed better cytotoxicity than the positive drug 5-fluorouracil. Then, functional technologies such as the 5-ethyny1-2'-deoxyridine, wound healing, Transwell, and Western blot assays indicated that they could significantly inhibit the proliferation of HT29 and Lovo cells, promote apoptosis by up-regulating Bax, and down-regulating B-cell CLL/lymphoma 2 and poly ADP-ribose polymerase. Furthermore, a Western blot assay combining the Dsh homolog 2 agonist IWP-L6 and the β-catenin agonist MG132 suggested their mechanism of action was closely related to the inhibition of the Wnt/β-catenin signaling pathway. In conclusion, previously undescribed neolignans, penthoneolignans A (1) and F (6), may intervene in the development and progression of colorectal cancer by inhibiting the Wnt/β-catenin signaling pathway and have the potential to be drug candidates.
Collapse
Affiliation(s)
- Mengqi Lu
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Jingya Ruan
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Rui Yu
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Ying Zhang
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Wei Zhao
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Dingshan Yang
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Wanxia Wang
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China
| | - Yi Zhang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China; Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China.
| | - Tao Wang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China; Institute of TCM, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, 301617, Tianjin, China.
| |
Collapse
|
13
|
An Updated Review on Efficiency of Penthorum chinense Pursh in Traditional Uses, Toxicology, and Clinical Trials. BIOMED RESEARCH INTERNATIONAL 2023; 2023:4254051. [PMID: 36852294 PMCID: PMC9966574 DOI: 10.1155/2023/4254051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 02/20/2023]
Abstract
Traditional Chinese medicines (TCM) play an important role in the control and treatment of several animal diseases. Penthorum chinense Pursh (PCP) is a famous plant for its use in traditional medication practice and therapeutic effects in numerous pathological conditions. In China, PCP is utilized for both food and medication due to numerous bioactivities. PCP is widely administered in prevention and treatment of traumatic injury, edema, and liver diseases with functions of reducing swelling, support diuresis, blood stasis, and mitigation symptoms of excessive alcohol intake. Recently, PCP highlighted for research trials in various fields including pharmacology, pharmacognosy, cosmeceuticals, nutraceuticals, and pharmaceuticals due to medicinal significance with less toxicity and an effective ethnomedicine in veterinary practice. PCP contains diverse important ingredients such as flavonoids, organic acids, coumarins, lignans, polyphenols, and sterols that are important bioactive constituents of PCP exerting the therapeutic benefits and organ-protecting effects. In veterinary, PCP extract, compound, and phytochemicals/biomolecules significantly reversed the liver and kidney injuries, via antioxidation, oxidative stress, apoptosis, mitochondrial signaling pathways, and related genes. PCP water extract and compounds also proved in animal and humans' clinical trial for their hepatoprotective, antiaging, nephroprotective, anti-inflammatory, antidiabetic, antibacterial, antiapoptotic, immune regulation, and antioxidative stress pathways. This updated review spotlighted the current information on efficiency and application of PCP by compiling and reviewing recent publications on animal research. In addition, this review discussed the toxicology, traditional use, comparative, and clinical application of PCP in veterinary practices to authenticate and find out new perspectives on the research and development of this herbal medicine.
Collapse
|
14
|
Protective Effect of Phloretin against Hydrogen Peroxide-Induced Oxidative Damage by Enhancing Autophagic Flux in DF-1 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8359118. [PMID: 36620085 PMCID: PMC9822743 DOI: 10.1155/2022/8359118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022]
Abstract
Phloretin (PHL) is a dihydrochalcone flavonoid isolated from the peel and root bark of apples, strawberries, and other plants with antioxidative characteristic. In this study, we aimed to investigate the protective effect and the potential mechanism of PHL on hydrogen peroxide (H2O2)-induced oxidative damage in DF-1 cells. The results showed that PHL exhibited no cytotoxic effect on DF-1 cells at concentration below 20 μM. PHL markedly increased H2O2-reduced cell viability, decreased H2O2-induced apoptosis, as evidenced by reduced apoptosis rate, the upregulation of gene and protein level of Bcl-2, and the downregulation of gene and protein level of Bax and Cleaved caspase3. In addition, PHL reduced H2O2-induced reactive oxygen species (ROS) production and restored antioxidant enzymes activities as well as mitochondrial membrane potential in a dose-dependent manner. Moreover, PHL prior to H2O2 further increased LC3-II level, promoted p62 turnover and improved lysosomal function. Importantly, autophagy inhibitor chloroquine (CQ) reversed the protective effect of PHL, and increased H2O2-induced apoptosis. Furthermore, PHL inhibited the phosphorylation levels of ERK, p38, and JNK. Collectively, these results indicate that PHL could attenuate H2O2-induced oxidative injury and apoptosis by maintaining lysosomal function and promoting autophagic flux, and MAPKs pathway may be involved in this process. Our study provides evidence that PHL could as a new strategy to against oxidative damage in poultry industry.
Collapse
|
15
|
Zhou XG, Qiu WQ, Yu L, Pan R, Teng JF, Sang ZP, Law BYK, Zhao Y, Zhang L, Yan L, Tang Y, Sun XL, Wong VKW, Yu CL, Wu JM, Qin DL, Wu AG. Targeting microglial autophagic degradation of the NLRP3 inflammasome for identification of thonningianin A in Alzheimer's disease. Inflamm Regen 2022; 42:25. [PMID: 35918778 PMCID: PMC9347127 DOI: 10.1186/s41232-022-00209-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/05/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND NLRP3 inflammasome-mediated neuroinflammation plays a critical role in the pathogenesis and development of Alzheimer's disease (AD). Microglial autophagic degradation not only decreases the deposits of extracellular Aβ fibrils but also inhibits the activation of NRLP3 inflammasome. Here, we aimed to identify the potent autophagy enhancers from Penthorum chinense Pursh (PCP) that alleviate the pathology of AD via inhibiting the NLRP3 inflammasome. METHODS At first, autophagic activity-guided isolation was performed to identify the autophagy enhancers in PCP. Secondly, the autophagy effect was monitored by detecting LC3 protein expression using Western blotting and the average number of GFP-LC3 puncta per microglial cell using confocal microscopy. Then, the activation of NLRP3 inflammasome was measured by detecting the protein expression and transfected fluorescence intensity of NLRP3, ASC, and caspase-1, as well as the secretion of proinflammatory cytokines. Finally, the behavioral performance was evaluated by measuring the paralysis in C. elegans, and the cognitive function was tested by Morris water maze (MWM) in APP/PS1 mice. RESULTS Four ellagitannin flavonoids, including pinocembrin-7-O-[4″,6″-hexahydroxydiphenoyl]-glucoside (PHG), pinocembrin-7-O-[3″-O-galloyl-4″,6″-hexahydroxydiphenoyl]-glucoside (PGHG), thonningianin A (TA), and thonningianin B (TB), were identified to be autophagy enhancers in PCP. Among these, TA exhibited the strongest autophagy induction effect, and the mechanistic study demonstrated that TA activated autophagy via the AMPK/ULK1 and Raf/MEK/ERK signaling pathways. In addition, TA effectively promoted the autophagic degradation of NLRP3 inflammasome in Aβ(1-42)-induced microglial cells and ameliorated neuronal damage via autophagy induction. In vivo, TA activated autophagy and improved behavioral symptoms in C. elegans. Furthermore, TA might penetrate the blood-brain barrier and could improve cognitive function and ameliorate the Aβ pathology and the NLRP3 inflammasome-mediated neuroinflammation via the AMPK/ULK1 and Raf/MEK/ERK signaling pathways in APP/PS1 mice. CONCLUSION We identified TA as a potent microglial autophagy enhancer in PCP that promotes the autophagic degradation of the NLRP3 inflammasome to alleviate the pathology of AD via the AMPK/ULK1 and Raf/MEK/ERK signaling pathways, which provides novel insights for TA in the treatment of AD.
Collapse
Affiliation(s)
- Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wen-Qiao Qiu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Department of Neurosurgery Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Rong Pan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jin-Feng Teng
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Zhi-Pei Sang
- School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Ya Zhao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Li Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Xiao-Lei Sun
- Vascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Da-Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
16
|
Liu D, Cheng Y, Mei X, Xie Y, Tang Z, Liu J, Cao X. Mechanisms of acrolein induces toxicity in human umbilical vein endothelial cells: Oxidative stress, DNA damage response, and apoptosis. ENVIRONMENTAL TOXICOLOGY 2022; 37:708-719. [PMID: 34908224 DOI: 10.1002/tox.23436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/03/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
Acrolein is a ubiquitous environmental pollutant that produced by the incomplete combustion of cigarette smoke, forest fires, petroleum fuels, plastic materials, and cooking fumes. Inhalation is a common form of people exposure to acrolein, increasing evidence demonstrates that acrolein impairs the cardiovascular system by targeting vascular endothelial cells. However, the molecular mechanism of the cytotoxicity of acrolein exposure on vascular endothelial cells remains unclear. This work focused on the toxicity of acrolein on human umbilical vein endothelial cells (HUVECs). The molecular mechanism was studied based on oxidative stress, DNA damage response (DDR), and mitochondrial apoptosis pathways. After HUVECs were treated with 12.5, 25, and 50 μM acrolein for 24 h, cell viability, cell colony formation, mitochondrial membrane potential, and adenosine triphosphate content significantly reduced, and acrolein increased intracellular reactive oxygen species, apoptosis rate, and 8-hydroxy-2 deoxyguanosine (8-OHdG) level. Furthermore, p38MAPK and c-Jun N-terminal kinase signaling pathways were activated in response to oxidative stress. Moreover, acrolein induced G0/G1phase arrest, promoted the expression of γ-H2AX, activated the DDR signaling pathway (Ataxia-Telangiectasia-Mutated [ATM] and Rad-3-related/Chk1 and ATM/Chk2), and triggered the consequent cell cycle checkpoints. Finally, the protein expression of Bax/Bcl-2 and cleaved Caspase-3 was up-regulated, suggesting apoptosis was induced by triggering the mitochondrial apoptosis pathway. All these results indicated that acrolein induced HUVECs cytotoxicity by regulating oxidative stress, DNA damage, and apoptosis. This study provides a novel perspective on the mechanism of acrolein-induced cardiovascular toxicity, it will be helpful for the prevention of acrolein-induced cardiovascular disease.
Collapse
Affiliation(s)
- Dan Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Ye Cheng
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Xueying Mei
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Yanzhen Xie
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Zhipeng Tang
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Jianli Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| | - Xiangyu Cao
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, China
| |
Collapse
|
17
|
Shen X, Li Z, Guo Z, Wang Y, Li T, Li G. Nonselective Cell Necrosis Mediated by the Total Flavones of Penthorum Chinensis Pursh and Thonningianin-A in Human Hepatic and Hepatoma Cells. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221086903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Penthorum chinensis Pursh (PCP), family Penthoraceae, has been used for hundreds of years in China. With the launch of PCP tablets, clinical applications focused on liver fibrosis and hepatocarcinoma. The purpose of this research was to explore the selectivity and toxicity of the active pharmacodynamic ingredients of PCP in vitro. The total flavones of PCP (TFPCE) and thonningianin-A (Th-A), a major flavone in TFPCE, were investigated on the cell death patterns in human hepatoma cells (HepG2) and human hepatic cells (LO2), followed by a concentration detection of LDH in the supernatants. Apoptosis and necrosis detection kits were used to validate the patterns of cell death caused by TFPCE and Th-A. Finally, the cytotoxicity of both TFPCE and Th-A were reproduced in the colorectal adenocarcinoma cells (NCI-H716). The results indicated that TFPCE inhibits the cell viability of HepG2 cells at a concentration lower than 25 μg/mL. Alternatively, the cell viability of LO2 cells dramatically decreased in the treatment of TFPCE at 25 μg/mL. The effects of Th-A on the cell viability of HepG2 cells and LO2 cells were consistent with TFPCE. LDH detection indicated that TFPCE and Th-A increased the LDH concentration of the supernatants in a dose-dependent way, indicating the pattern of cell necrosis. Fluorescence staining verified the necrosis cell death caused by TFPCE and Th-A. A dose-dependent tendency was obtained in NCI-H716 cells, indicating that the cell viability of NCI-H716 cells was significantly suppressed with the treatment of TFPCE and Th-A. Our results bring the potential toxicity of PCP to the forefront of public attention. Therefore, the clinical application of P chinensis is required to focus more on its cytotoxic effect.
Collapse
Affiliation(s)
- Xin Shen
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zekun Li
- Shijiazhuang Yiling Pharmaceutical, Shijiazhuang, China
- The Pennsylvania State University, University Park, PA, USA
| | - Zhifang Guo
- Shijiazhuang Yiling Pharmaceutical, Shijiazhuang, China
| | - Yanan Wang
- Shijiazhuang Yiling Pharmaceutical, Shijiazhuang, China
| | - Tongtong Li
- College of Integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Guohui Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Tao W, Yue X, Ye R, Nabi F, Shang Y, Zhu Z, Ahmed BZ, Liu J. Hepatoprotective Effect of the Penthorum Chinense Pursh Extract against the CCl 4-Induced Acute Liver Injury via NF-κB and p38-MAPK PATHWAYS in Dogs. Animals (Basel) 2022; 12:ani12050569. [PMID: 35268138 PMCID: PMC8909057 DOI: 10.3390/ani12050569] [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: 12/29/2021] [Revised: 02/03/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
Acute liver injury (ALI), manifested by acute hepatocellular damages and necrosis, is a life-threatening clinical syndrome and Penthorum Chinense Pursh (PCP) is a well-known folk medicine practiced for liver-related diseases. This study aimed to investigate the ameliorative effects of PCP extract (PCPE) on carbon tetrachloride (CCl4) induced ALI in dogs via mitogen-activated protein kinase (MAPK) and Nuclear factor κB (NF-κB) signaling pathway. Healthy dogs were induced by CCl4 and treated with different dosage regimes of PCPE for 7 days. CCl4 produced acute liver injury and induced both oxidative stress and an inflammatory response in dogs. The PCPE significantly ameliorated and improved vacuolar inflammatory lesions in liver tissues during ALI, enhanced activity of superoxide dismutase, and restored glutathione peroxidase, further significantly reducing the indices of malondialdehyde and nitric oxide in serum. Inflammatory factors (IL-1β, IL-6, and TNF-α) were declined and anti-inflammatory factors (IL-10) were increased by the application of PCPE. PCPE treatment, down-regulated the MEKK4, MKK3, p38MAPK, MSK1, and NF-κB, and upregulated the IkB mRNA levels (p < 0.01) in ALI affected dogs. In conclusion, PCPE repaired acute liver injury by improving antioxidant enzymes and by reducing oxidation products. Furthermore, the PCPE inhibited the MAPK/NF-κB signaling pathway, which resulted in anti-inflammatory and antioxidant effects on ALI-induced dogs. In the future, PCPE could be a useful ethnomedicine in veterinary clinical practices for the treatment of liver injuries or failures.
Collapse
Affiliation(s)
- Weilai Tao
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
| | - Xin Yue
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
| | - Ruiling Ye
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
| | - Fazul Nabi
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
| | - Yangfei Shang
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
| | - Zhaorong Zhu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
- Chinese Veterinary Herbal Drugs Innovation Research Lab, University Veterinary Science Engineering Research Center in Chongqing, Chongqing 402460, China
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing 402460, China
| | - Bhutto Zohaib Ahmed
- Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water, and Marine Sciences, Uthal 90150, Pakistan;
| | - Juan Liu
- College of Veterinary Medicine, Southwest University, Chongqing 402460, China; (W.T.); (X.Y.); (R.Y.); (F.N.); (Y.S.); (Z.Z.)
- Chinese Veterinary Herbal Drugs Innovation Research Lab, University Veterinary Science Engineering Research Center in Chongqing, Chongqing 402460, China
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing 402460, China
- Correspondence:
| |
Collapse
|
19
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
20
|
Tang X, Zhang Y, Liu X, Li X, Zhao H, Cui H, Shi Y, Chen Y, Xu H, Meng Z, Zhao L, Chen H, Wang Z, Zhu M, Lin Y, Yang B, Zhang Y. Aloe-emodin derivative produces anti-atherosclerosis effect by reinforcing AMBRA1-mediated endothelial autophagy. Eur J Pharmacol 2022; 916:174641. [PMID: 34800465 DOI: 10.1016/j.ejphar.2021.174641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/15/2021] [Accepted: 11/12/2021] [Indexed: 11/29/2022]
Abstract
Atherosclerosis is an inflammatory disease of high lethality associated with endothelial dysfunction. Due to the pathophysiological complexity and our incomplete understanding of the mechanisms for the development and progression of atherosclerosis, effective means for the prevention and treatment of atherosclerosis still need further exploration. This study was designed to investigate the potential effects and underlying mechanisms of aloe-emodin derivative (AED) on atherosclerosis. High fat diet (HFD) treated ApoE-/- mice were used as an animal model of atherosclerosis. Intragastric administration of aloe-emodin (AE) or AED for 12 weeks markedly reduced the atherosclerotic plaque in aorta with decreased plaque area, lipid accumulation, macrophage infiltration, collagen content and metabolic abnormalities. By comparison, AED produced more potent anti-atherosclerosis effects than AE at the same dose. AED enhanced production of autophagy flux in cultured human aortic endothelial cells (HAECs). Moreover, AED increased the expression of activating molecule in Beclin1-regulated autophagy 1 (AMBRA1), a key protein involved in autophagosome formation. Furthermore, knockdown of AMBRA1 blocked the promotion effect of AED on autophagy in HAECs. Taken together, AED facilitates endothelial autophagy via AMBRA1 during the progression of atherosclerosis, suggesting the potential application of this compound for atherosclerosis treatment.
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yue Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xiaohan Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hongrui Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hao Cui
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yang Shi
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yongchao Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Honglin Xu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Ziyu Meng
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Limin Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hui Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Zhixia Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Mengying Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuan Lin
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Baofeng Yang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China; Department of Pharmacology and Therapeutics, Melbourne School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences University of Melbourne, Melbourne, Australia.
| | - Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China.
| |
Collapse
|
21
|
Nabi F, Tao W, Ye R, Li Z, Lu Q, Shang Y, Hu Y, Fang J, Bhutto ZA, Liu J. Penthorum Chinense Pursh Extract Alleviates Aflatoxin B1-Induced Liver Injury and Oxidative Stress Through Mitochondrial Pathways in Broilers. Front Vet Sci 2022; 9:822259. [PMID: 35187148 PMCID: PMC8847786 DOI: 10.3389/fvets.2022.822259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022] Open
Abstract
Aflatoxin is an important toxicant of the fungal origin and poses a threat to the poultry industry. This study was designed to reveal the underlying mechanism and protective methods against aflatoxin B1 (AFB1)-induced liver injury, oxidative stress, and apoptosis using a Traditional Chinese medicine, Penthorum chinense Pursh extract (PCPE), in broilers. A total of 164 (day-old) broilers were equally allocated to the control, AFB1 (3 mg/kg feed), positive drug (Yin-Chen-Hao Tang extract, 10 ml/kg feed), PCPE (2 g PCPE/kg), and PCPE low, medium, and high dose groups (1 g, 2 g, 3 g PCPE/kg feed, respectively). AFB1 significantly decreased the growth performance and serum immunoglobulin level, altered normal serum biochemical parameters and antioxidant activities, and induced histopathological lesions in the liver as compared to control group. Additionally, AFB1 significantly up-regulated the mRNA expression levels of apoptosis-related genes such as Bax, Bak, caspase-9, caspase-3, and p53, whereas it down-regulated the expression levels of BCL2 in the liver of broilers. The supplementation of different doses of PCPE to AFB1-affected birds significantly eased AFB1 negative effects by improving growth performance, immunoglobulin level, and oxidative capacity, and reversed oxidative stress and pathological lesions in liver. Furthermore, supplementation of PCPE to the AFB1 group reversed apoptosis by significantly down-regulating the mRNA expression levels of Bax, Bak, caspase-9, caspase-3, and p53 and up-regulating the expression levels of BCL2 in the liver of broilers. Based on these results, we conclude that supplementation of PCPE is protective and safe against oxidative stress, is anti-apoptotic, and reverses the liver damage caused by AFB1 in broilers.
Collapse
Affiliation(s)
- Fazul Nabi
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
- Department of Poultry Science, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Weilai Tao
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Ruiling Ye
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenzhen Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qin Lu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yangfei Shang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Hu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jiali Fang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zohaib Ahmed Bhutto
- Department of Poultry Science, Faculty of Veterinary and Animal Science, Lasbela University of Agriculture, Water and Marine Sciences, Uthal, Pakistan
| | - Juan Liu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing, China
- Chinese Veterinary Herbal Drugs Innovation Research Laboratory, University Veterinary Science Engineering Research Center in Chongqing, Chongqing, China
- Immunology Research Center of Medical Research Institute, Southwest University, Chongqing, China
- *Correspondence: Juan Liu
| |
Collapse
|
22
|
Liu Q, Li C, Zhao P, Li J, Deng Z. Quantification of thonningianin a in rat plasma by liquid chromatography tandem mass spectrometry and its application to a pharmacokinetic study. PHARMACEUTICAL BIOLOGY 2021; 59:525-531. [PMID: 33915063 PMCID: PMC8871622 DOI: 10.1080/13880209.2021.1913188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
CONTEXT Thonningianin A is an ellagitannin substance and displays multiple pharmacological activities. OBJECTIVE This study investigated the pharmacokinetic characteristics of thonningianin A after oral administration in rats using a fully validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. MATERIALS AND METHODS A sensitive and selective LC-MS/MS assay was developed for quantifying thonningianin A. Eighteen Wistar rats were randomly divided into three groups (n = 6), which were given at a single dose of 10, 20, or 40 mg/kg thonningianin A by gavage. Blood samples (200 µL) were collected from the orbit vein at designated time points and analyzed using the LC-MS/MS method to measure the levels of thonningianin A. RESULTS Thonningianin A and internal standard (IS) were eluted at 1.5 and ∼3.0 min, respectively. The selected reaction mode transitions monitored were m/z 873.2 > 300.3 and 819.3 > 610.6 for thonningianin A and the IS, respectively. The calibration range was 10-1200 ng/mL. The intra- and the inter-day accuracy and precision met the acceptance criteria. No carryover and matrix effect were observed. The plasma concentrations of thonningianin A increased rapidly after oral administration of three dosages and reached the mean peak concentrations (Cmax) within 0.61-0.83 h. Meanwhile, AUC0-t/AUC0-∞ of the three dosage groups was more than 89.0% (10 mg/kg), 95.7% (20 mg/kg), and 97.0% (40 mg/kg). DISCUSSION AND CONCLUSIONS The present method is the first report in terms of the simple precipitation procedure, high sensitivity, and high-throughput efficiency. This validated assay was successfully applied to determine the pharmacokinetic behaviours of thonningianin A in rats. This study should be helpful for providing references for understanding the action mechanism and further application of Penthorum chinense.
Collapse
Affiliation(s)
- Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Chunmin Li
- Department of Pharmacy, Jinan Maternity and Child Care Hospital, Jinan, PR China
| | - Pan Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Jing Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| | - Zhipeng Deng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, PR China
| |
Collapse
|
23
|
Tao W, Li Z, Nabi F, Hu Y, Hu Z, Liu J. Penthorum chinense Pursh Compound Ameliorates AFB1-Induced Oxidative Stress and Apoptosis via Modulation of Mitochondrial Pathways in Broiler Chicken Kidneys. Front Vet Sci 2021; 8:750937. [PMID: 34692815 PMCID: PMC8531719 DOI: 10.3389/fvets.2021.750937] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Aflatoxin B1 (AFB1) is a carcinogenic mycotoxin widely present in foods and animal feeds; it represents a great risk to human and animal health. The aim of this study was to investigate the protective effects of Penthorum chinense Pursh compound (PCPC) against AFB1-induced damage, oxidative stress, and apoptosis via mitochondrial pathways in kidney tissues of broilers. One-day-old chickens (n = 180) were randomly allocated to six groups: control, AFB1 (2.8 mg AFB1/kg feed), positive drug (10 mLYCHT/kg feed), and PCPC high, medium, and low-dose groups (15, 10, and 5 ml PCPC/kg feed, respectively). AFB1 treatment reduced weight gain and induced oxidative stress and kidney damage in broiler tissues; however, PCPC supplementation effectively enhanced broiler performance, ameliorated AFB1-induced oxidative stress, and inhibited apoptosis in the kidneys of broilers. The mRNA expression levels of mitochondria-related apoptosis genes (Bax, Bak, cytochrome c, caspase-9, and caspase-3) were significantly increased, whereas BCL2 expression level decreased in the AFB1 group. Supplementation of PCPC to the AFB1 group significantly reversed the changes in mRNA expression levels of these apoptosis-associated genes compared to those in the AFB1 group. The mRNA levels of NRF2 and HMOX1 in the kidneys of the AFB1 group were significantly reduced compared to those in the control group, whereas PCPC significantly increased the NRF2 and HMOX1 mRNA levels. AFB1 decreased the levels of Beclin1, LC3-I, and LC3-II and increased P53 levels in the kidney compared to those in the control, whereas PCPC significantly reversed these changes to normal levels of autophagy-related genes compared to those in the AFB1 group. In conclusion, our findings demonstrated that PCPC ameliorated AFB1-induced oxidative stress by regulating the expression of apoptosis-related genes and mitochondrial pathways. Our results suggest that PCPC represents a natural and safe agent for preventing AFB1-induced injury and damage in broiler tissues.
Collapse
Affiliation(s)
- Weilai Tao
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenzhen Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Fazul Nabi
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yu Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zeyu Hu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Juan Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China.,Chinese Veterinary Herbal Drugs Innovation Research Lab, University Veterinary Science Engineering Research Center in Chongqing, Chongqing, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| |
Collapse
|
24
|
Kaymak E, Akin AT, Öztürk E, Karabulut D, Kuloğlu N, Yakan B. Thymoquinone has a neuroprotective effect against inflammation, oxidative stress, and endoplasmic reticulum stress in the brain cortex, medulla, and hippocampus due to doxorubicin. J Biochem Mol Toxicol 2021; 35:e22888. [PMID: 34392583 DOI: 10.1002/jbt.22888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/22/2021] [Accepted: 08/06/2021] [Indexed: 01/02/2023]
Abstract
Although doxorubicin (DOX) is used in many cancer treatments, it causes neurotoxicity. In this study, the effect of thymoquinone (THQ), a powerful antioxidant, on DOX-induced neurotoxicity was evaluated. In total, 40 rats were used and 5 groups were formed. Group I: control group (n = 8); Group II: olive oil group (n = 8); Group III: the THQ group (n = 8); THQ 10 mg/kg per day was given intraperitoneally (i.p.) throughout the experiment; group IV: DOX group (n = 8); On Day 7 of the experiment, a single dose of 15 mg/kg intraperitoneally DOX injected; group V: DOX + THQ group (n = 8); Throughout the experiment, 10 mg/kg THQ per day and intraperitoneally 15 mg/kg DOX on Day 7 were injected. Immunohistochemically, tumor necrosis factor-α (TNF-α), interleukin-17 (IL-17), hypoxia-inducible factor 1α (HIF1-α), glucose regulatory protein 78 (GRP78), and the gene inducible by growth arrest and DNA damage 153 (GADD153) proteins were evaluated in the brain cortex, medulla, and hippocampus regions. Total oxidant status (TOS) levels and total antioxidant status (TAS) in the brain tissue were measured. TNF-α, IL-17, HIF1-α, GRP78, and GADD153 immunoreactivities significantly increased in the DOX group in the study. THQ significantly reduced these values. THQ increased the TAS level significantly and decreased the TOS level significantly compared to the DOX group. THQ may play a role as a neuroprotective agent in DOX-induced neurotoxicity in the cortex, medulla, and hippocampus regions of the brain.
Collapse
Affiliation(s)
- Emin Kaymak
- Department of Histology and Embryology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | | | - Emel Öztürk
- Histology-Embryology Department, Harran University, Harran, Turkey
| | - Derya Karabulut
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Nurhan Kuloğlu
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Birkan Yakan
- Department of Histology and Embryology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
25
|
Liu X, Xu Y, Cheng S, Zhou X, Zhou F, He P, Hu F, Zhang L, Chen Y, Jia Y. Geniposide Combined With Notoginsenoside R1 Attenuates Inflammation and Apoptosis in Atherosclerosis via the AMPK/mTOR/Nrf2 Signaling Pathway. Front Pharmacol 2021; 12:687394. [PMID: 34305600 PMCID: PMC8293676 DOI: 10.3389/fphar.2021.687394] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/24/2021] [Indexed: 01/21/2023] Open
Abstract
Inflammation and apoptosis of vascular endothelial cells play a key role in the occurrence and development of atherosclerosis (AS), and the AMPK/mTOR/Nrf2 signaling pathway plays an important role in alleviating the symptoms of AS. Geniposide combined with notoginsenoside R1 (GN combination) is a patented supplement for the prevention and treatment of AS. It has been proven to improve blood lipid levels and inhibit the formation of AS plaques; however, it is still unclear whether GN combination can inhibit inflammation and apoptosis in AS by regulating the AMPK/mTOR/Nrf2 signaling pathway and its downstream signals. Our results confirmed that the GN combination could improve blood lipid levels and plaque formation in ApoE−/− mice fed with a high-fat diet (HFD), inhibit the secretion of serum inflammatory factors and oxidative stress factors. It also decreased the expression of pyrin domain containing protein 3 (NLRP3) inflammasome-related protein and Bax/Bcl2/caspase-3 pathway-related proteins. At the same time, the GN combination could also inhibit the H2O2-induced inflammatory response and apoptosis of human umbilical vein endothelial cells (HUVECs), which is mainly related to the activation of the AMPK/mTOR pathway by GN combination, which in turn induces the activation of Nrf2/HO-1 signal. In addition, the above phenomenon could be significantly reversed by dorsomorphin. Therefore, our experiments proved for the first time that the GN combination can effectively inhibit AS inflammation and apoptosis by activating the AMPK/mTOR/Nrf2 signaling pathway to inhibit the NLRP3 inflammasome and Bax/Bcl2/caspase-3 pathway.
Collapse
Affiliation(s)
- Xiaoyu Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuling Xu
- College of Health, Fujian Medical University, Fuzhou, China
| | - Saibo Cheng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xinghong Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Fenghua Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Peikun He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Fang Hu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lifang Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuhua Jia
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Huang D, Wang X, Zhu Y, Gong J, Liang J, Song Y, Zhang Y, Liu L, Wei C. Bazi Bushen Capsule Alleviates Post-Menopausal Atherosclerosis via GPER1-Dependent Anti-Inflammatory and Anti-Apoptotic Effects. Front Pharmacol 2021; 12:658998. [PMID: 34248622 PMCID: PMC8267998 DOI: 10.3389/fphar.2021.658998] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/07/2021] [Indexed: 01/16/2023] Open
Abstract
Bazi Bushen capsule (BZBS), as a Chinese medicine used to relieve fatigue, has been proven effective for the treatment of atherogenesis through antilipid effects. To investigate the potential mechanism of BZBS in the anti-atherosclerotic effect, Ovx/ApoE-/- mice were applied to investigate the anti-atherosclerotic efficiency and potential mechanism of BZBS. Therapeutic effect was evaluated based on the number of CD68+ and CD3+ cells, the level of intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), and the ratio of cleaved caspase-3/caspase-3, as well as increasing ratio of Bcl2/Bax. Human umbilical vein endothelial cells (HUVECs) were chosen to evaluate the role of GPER1. Treatment with BZBS reduced lipid deposition by reducing the numbers of CD68+ and CD3+ cells, the level of ICAM-1 and VCAM-1, and the ratio of cleaved caspase-3/caspase-3, and increasing the ratio of Bcl2/Bax as compared with the control group. In si-GPER1-treated HUVECs, the anti-apoptotic effect of BZBS was decreased. This study revealed that BZBS exhibited a clear effect against atherogenesis via GPER1-dependent anti-inflammatory and anti-apoptotic mechanisms. We believe that this manuscript is informative and useful for researchers pursuing the related alleviation of post-menopausal AS via anti-inflammatory and anti-apoptotic mechanisms.
Collapse
Affiliation(s)
- Dan Huang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Xindong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yunhong Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Juexiao Gong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Junqing Liang
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China
| | - Yanfei Song
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China
| | - Yiyan Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Linsheng Liu
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cong Wei
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
27
|
Raj SD, Fann DY, Wong E, Kennedy BK. Natural products as geroprotectors: An autophagy perspective. Med Res Rev 2021; 41:3118-3155. [PMID: 33973253 DOI: 10.1002/med.21815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022]
Abstract
Over the past decade, significant attention has been given to repurposing Food and Drug Administration approved drugs to treat age-related diseases. In contrast, less consideration has been given to natural bioactive compounds. Consequently, there have been limited attempts to translate these compounds. Autophagy is a fundamental biological pathway linked to aging, and numerous strategies to enhance autophagy have been shown to extend lifespan. Interestingly, there are a number of natural products that are reported to modulate autophagy, and here we describe a number of them that activate autophagy through diverse molecular and cellular mechanisms. Among these, Urolithin A, Spermidine, Resveratrol, Fatty Acids and Phospholipids, Trehalose and Lithium are featured in detail. Finally, we outline possible strategies to optimise and increase the translatability of natural products, with the overall aim of delaying the ageing process and improving human healthspan.
Collapse
Affiliation(s)
- Stephen D Raj
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore
| | - David Y Fann
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore
| | - Esther Wong
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brian K Kennedy
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre For Healthy Longevity, National University Health System, National University of Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Agency for Science, Technology and Research (A*STAR), Singapore Institute for Clinical Sciences, Singapore
| |
Collapse
|
28
|
Wang W, Huang L, Thomas ER, Hu Y, Zeng F, Li X. Notoginsenoside R1 Protects Against the Acrylamide-Induced Neurotoxicity via Upregulating Trx-1-Mediated ITGAV Expression: Involvement of Autophagy. Front Pharmacol 2020; 11:559046. [PMID: 32982756 PMCID: PMC7493052 DOI: 10.3389/fphar.2020.559046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/17/2020] [Indexed: 01/07/2023] Open
Abstract
Acrylamide (ACR) is a common chemical used in various industries and it said to have chronic neurotoxic effects. It is produced during tobacco smoking and is also generated in high-starch foods during heat processing. Notoginsenoside R1 (NR1) is a traditional Chinese medicine, which is used to improve the blood circulation and clotting. The objective of this study was to investigate the mechanism of ACR-triggered neurotoxicity and to identify the protective role of NR1 by upregulating thioredoxin-1 (Trx-1). Our results have shown that NR1 could block the spatial and cognitive impairment caused by ACR administration. Bioinformatics analysis revealed that Trx-1 regulated autophagy via Integrin alpha V (ITGAV). NR1 could resist the ACR-induced neurotoxicity by upregulating thioredoxin-1 in PC12 cells and mice. The autophagy-related proteins like autophagy-related gene (ATG) 4B, Cathepsin D, LC3 II, lysosomal-associated membrane protein 2a (LAMP2a), and ITGAV were restored to normal levels by NR1 treatment in both PC12 cells and mice. Besides, we also found that overexpression of Trx-1 resisted ACR-induced autophagy in PC12 cells and downregulation of Trx-1 triggered autophagy induced by ACR in PC12 cells. Therefore, it could be concluded that Trx-1 was involved in the autophagy pathway. Besides, we also found that ITGAV was an intermediate node linking Trx-1 and the autophagy pathway.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China.,Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Lu Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | | | - Yingying Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| |
Collapse
|
29
|
Chloride channel 7 protects from redox status impairment-induced renal tubular epithelial cell apoptosis by activating autophagy. Life Sci 2020; 261:118484. [PMID: 32976885 DOI: 10.1016/j.lfs.2020.118484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/08/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
Abstract
AIM Chloride channel 7 (CLC-7), broadly expressed in kidney tissues, affects the lysosome degradation pathway. And redox status impairment contributes to cell apoptosis and activates autophagy flux. This study mainly investigates the role and molecular mechanism of CLC-7 in redox status impairment-induced autophagic flux and apoptosis. MAIN METHODS When NRK52E cells, rat renal tubular epithelial cells, were exposed to H2O2 treatment, apoptosis, autophagy flux, and CLC-7 expression were detected. Further investigation was done to observe the change of apoptosis and autophagy flux in renal cells under overexpression or knocking down of CLC-7. The lysosomes acidity, lysosome enzyme Cathepsin D activity and phosphorylation of Ampk/mTOR were also examined when CLC-7 was overexpressed or knocked down. KEY FINDINGS Redox status impairment induced apoptosis and autophagy flux in NRK52E cells and upregulated CLC-7. Overexpression of CLC-7 increased lysosome acidity and Cathepsin D activity. In cells with CLC-7 overexpression, we observed a significant increase of autophagy flux and decline of apoptosis, as well as an apparent increase of p-Ampk and decrease of p-mTOR. On the contrary, cells with knocking down CLC-7 led to opposite results. SIGNIFICANCES CLC-7 is essential to maintain and enhance acidity and enzyme activity in lysosome. Through activating autophagy flux, it exerts survival against renal tubular epithelial cell apoptosis induced by redox status impairment. Its function to modulate Ampk/mTOR pathway is the possible reason why CLC-7 can trigger autophagy flux.
Collapse
|
30
|
An Overview of the Mechanism of Penthorum chinense Pursh on Alcoholic Fatty Liver. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4875764. [PMID: 33014105 PMCID: PMC7519454 DOI: 10.1155/2020/4875764] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 08/13/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
Alcohol liver disease (ALD) caused by excessive alcohol consumption is a progressive disease, and alcohol fatty liver disease is the primary stage. Currently, there is no approved drug for its treatment. Abstinence is the best way to heal, but patients' compliance is poor. Unlike other chronic diseases, alcohol fatty liver disease is not caused by nutritional deficiencies; it is caused by the molecular action of ingested alcohol and its metabolites. More and more studies have shown the potential of Penthorum chinense Pursh (PCP) in the clinical use of alcohol fatty liver treatment. The purpose of this paper is to reveal from the essence of PCP treatment of alcohol liver mechanism mainly by the ethanol dehydrogenase (ADH) and microsomal ethanol oxidation system-dependent cytochrome P4502E1 (CYP2E1) to exert antilipogenesis, antioxidant, anti-inflammatory, antiapoptotic, and autophagy effects, with special emphasis on its mechanisms related to SIRT1/AMPK, KEAP-1/Nrf2, and TLR4/NF-κB. Overall, data from the literature shows that PCP appears to be a promising hepatoprotective traditional Chinese medicine (TCM).
Collapse
|
31
|
Castejón-Vega B, Giampieri F, Alvarez-Suarez JM. Nutraceutical Compounds Targeting Inflammasomes in Human Diseases. Int J Mol Sci 2020; 21:E4829. [PMID: 32650482 PMCID: PMC7402342 DOI: 10.3390/ijms21144829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
The macromolecular complex known as "inflammasome" is defined as an intracellular multi-protein complex composed of a sensor receptor (PRR), an adaptor protein and an effector enzyme (caspase-1), which oligomerize when they sense danger, such as how the NLR family, AIM-2 and RIG-1 receptors protect the body against danger via cytokine secretion. Within the NLR members, NLRP3 is the most widely known and studied inflammasome and has been linked to many diseases. Nowadays, people's interest in their lifestyles and nutritional habits is increasing, mainly due to the large number of diseases that seem to be related to both. The term "nutraceutical" has recently emerged as a hybrid term between "nutrition" and "pharmacological" and it refers to a wide range of bioactive compounds contained in food with relevant effects on human health. The relationship between these compounds and diseases based on inflammatory processes has been widely exposed and the compounds stand out as an alternative to the pathological consequences that inflammatory processes may have, beyond their defense and repair action. Against this backdrop, here we review the results of studies using several nutraceutical compounds in common diseases associated with the inflammation and activation of the NLRP3 inflammasomes complex. In general, it was found that there is a wide range of nutraceuticals with effects through different molecular pathways that affect the activation of the inflammasome complex, with positive effects mainly in cardiovascular, neurological diseases, cancer and type 2 diabetes.
Collapse
Affiliation(s)
- Beatriz Castejón-Vega
- Research Laboratory, Oral Medicine Department, University of Sevilla, 41009 Sevilla, Spain;
| | - Francesca Giampieri
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain;
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche (DISCO)-Sez, Biochimica, Facoltà di Medicina, Università Politecnica delle Marche, 60131 Ancona, Italy
- College of Food Science and Technology, Northwest University, Xi’an 710069, China
| | - José M. Alvarez-Suarez
- Facultad de Ingeniería y Ciencias Aplicadas (FICA), AgroScience & Food Research Group, Universidad de Las Américas, 170125 Quito, Ecuador
- King Fahd Medical Research Center, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| |
Collapse
|