1
|
Tang J, Liu Y, Wu Y, Li S, Zhang D, Wang H, Wang W, Song X, Li Y. Saponins as potential novel NLRP3 inflammasome inhibitors for inflammatory disorders. Arch Pharm Res 2024; 47:757-792. [PMID: 39549164 DOI: 10.1007/s12272-024-01517-x] [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: 06/29/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024]
Abstract
Nucleotide-binding domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) is a downstream protein from the pattern recognition receptor family that forms the NLRP3 inflammasome. The NLRP3 inflammasome releases caspase-1, IL-1β, and IL-18, contributing to inflammatory responses associated with diabetes mellitus, arthritis, and ischemia-reperfusion injury. Recent studies suggest that specific saponin monomers and extracts from traditional Chinese medicines can inhibit inflammatory responses and related pathways, including the production of inflammatory factors. MCC950 is one of the most influential and specific NLRP3 inhibitors. Comparative molecular docking studies have identified 22 of the 37 saponin components as more robust binders to NLRP3 than MCC950. Dioscin, polyphyllin H, and saikosaponin-a have the highest binding affinities and potential NLRP3 inhibitors, offering a theoretical basis for developing novel anti-inflammatory therapies.
Collapse
Affiliation(s)
- Jiamei Tang
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yaxiao Liu
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Ying Wu
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Shixing Li
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Dongdong Zhang
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Haifang Wang
- Shaanxi Province Key Laboratory of Integrated Traditional Chinese and Western Medicine for the Prevention and Treatment of Cardiovascular Diseases, Xianyang, 712046, China
| | - Wei Wang
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xiaomei Song
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| | - Yuze Li
- Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
2
|
Song H, Han MG, Lee R, Park HJ. Neonatal exposure to high D-galactose affects germ cell development in neonatal testes organ culture. Sci Rep 2024; 14:24029. [PMID: 39402149 PMCID: PMC11473950 DOI: 10.1038/s41598-024-74895-3] [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: 05/23/2024] [Accepted: 09/30/2024] [Indexed: 10/17/2024] Open
Abstract
Excess exogenous supplementation of D-galactose (D-gal), a monosaccharide and reducing sugar, generates reactive oxygen species (ROS), leading to cell damage and death. ROS accumulation is critical in aging. Therefore, D-gal-induced aging mouse models are used in aging studies. Herein, we evaluated D-gal's effect on neonatal testis development using an in vitro organ culture method. Mouse testicular fragments (MTFs) derived from neonatal testes (postnatal day 5) were cultured with 500 mM D-gal for 5 days. D-gal-treated MTFs showed a significantly increased and decreased expression of undifferentiated and differentiated germ cell markers, respectively, with a substantial reduction in meiotic cells. In D-gal-exposed MTFs, expression levels of Sertoli cell markers (Sox9 and Wt1) increased, while those of StAR and 17β-HSD3, whose expressions are abundant in D-Gal treated adult Leydig cells, decreased. Additionally, the enzyme 3 β-HSD1, essential for steroidogenesis in Leydig cells, was significantly reduced in D-gal-exposed MTFs compared to that in controls.D-gal significantly increased the expression of Bad, Bax, and cleaved caspase-3 and -8. Via oxidative stress in MTF. Overall, D-gal negatively regulates germ cell and Leydig cell development in neonatal testes through pro-apoptotic mechanisms and ROS.
Collapse
Affiliation(s)
- Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, KIT, Konkuk University, Seoul, 05029, Republic of Korea
| | - Min-Gi Han
- Department of Stem Cell and Regenerative Biotechnology, KIT, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ran Lee
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si, 26339, Republic of Korea.
| | - Hyun-Jung Park
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si, 26339, Republic of Korea.
| |
Collapse
|
3
|
Wang H, Liu Y, Cui M, Guo Z, Zhao Y, Yang J, Wu C. Pseudoginsenoside-F11 reduces cognitive impairment and white matter injury in vascular dementia by alleviating autophagy-lysosomal pathway deficiency. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155883. [PMID: 39059268 DOI: 10.1016/j.phymed.2024.155883] [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: 02/25/2024] [Revised: 05/26/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Vascular dementia (VaD) resulting from chronic cerebral hypoperfusion (CCH) induces cognitive impairment and white matter injury (WMI). We previously found that CCH induces dysfunction of the autophagy-lysosomal pathway (ALP) in white matter (WM) of rats. Enhancing oligodendrocyte autophagy to counteract ALP deficiency is beneficial for cognitive recovery. Pseudogenoside-F11 (PF11), a saponin extracted from Panax quinquefolium l., provides neuroprotective benefits in many animal models of cerebral ischemia and dementia. PURPOSE To investigate how PF11 affects cognitive deterioration in rats with VaD induced by two vessel occlusion (2VO), and to determine if PF11 regulates ALP dysfunction in WM. METHODS CCH-related VaD was induced in rats using the 2VO method. PF11 (6, 12, 24 mg/kg, intragastric administration) was given continuously for 4 weeks postoperatively. Behavioral tests related to cognitive function were performed on the 28th day following 2VO. Transmission electron microscopy, immunofluorescence, western blotting and Luxol fast blue staining were used to assess the WMI and the mechanism of action of PF11 in 2VO-induced VaD. RESULTS PF11 (12 mg/kg) ameliorated 2VO-induced cognitive impairment. PF11 also alleviated WMI on the 28th day following 2VO, as characterized by reduction of neuronal axonal demyelination and axonal loss. Furthermore, PF11 prevented mature oligodendrocytes death by attenuating ALP deficiency in WM on the 14th day following 2VO, as manifested by enhancement of mechanistic target of rapamycin-mediated autophagy and lysosomal function, thereby reducing the aberrant accumulation of autophagy substrates and increasing the level of autophagosomes in WM. In addition, PF11 also prevented microglia and astrocytes from activating in WM on the 28th day following 2VO. CONCLUSION PF11 significantly ameliorates cognitive impairment and WMI, and the mechanism is at least partly related to lessening ALP dysfunction in WM by enhancing autophagy and reducing lysosomal defects in oligodendrocytes.
Collapse
Affiliation(s)
- Huiyang Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Minghui Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Zhenkun Guo
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Yang Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China.
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Box 31, 103 Wenhua Road, Shenyang, 110016, PR China.
| |
Collapse
|
4
|
Hamzeh O, Rabiei F, Shakeri M, Parsian H, Saadat P, Rostami-Mansoor S. Mitochondrial dysfunction and inflammasome activation in neurodegenerative diseases: Mechanisms and therapeutic implications. Mitochondrion 2023; 73:S1567-7249(23)00087-9. [PMID: 39492438 DOI: 10.1016/j.mito.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/02/2023] [Accepted: 10/28/2023] [Indexed: 11/05/2024]
Abstract
Impaired mitochondrial function is crucial to the pathogenesis of several neurodegenerative diseases. It causes the release of mitochondrial DNA (mtDNA), mitochondrial reactive oxygen species (mtROS), ATP, and cardiolipin, which activate the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome. NLRP3 inflammasome is an important innate immune system element contributing to neuroinflammation and neurodegeneration. Therefore, targeting the NLRP3 inflammasome has become an interesting therapeutic approach for treating neurodegenerative diseases. This review describes the role of mitochondrial abnormalities and over-activated inflammasomes in the progression of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple sclerosis (MS), Amyotrophic lateral sclerosis (ALS), and Friedrich ataxia (FRDA). We also discuss the therapeutic strategies focusing on signaling pathways associated with inflammasome activation, which potentially alleviate neurodegenerative symptoms and impede disease progression.
Collapse
Affiliation(s)
- Olia Hamzeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Fatemeh Rabiei
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mahdi Shakeri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Parsian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Payam Saadat
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sahar Rostami-Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
5
|
Zheng X, Lin Y, Huang L, Lin X. Effect of lidocaine on cognitively impaired rats: Anti-inflammatory and antioxidant mechanisms in combination with CRMP2 antiphosphorylation. Immun Inflamm Dis 2023; 11:e1040. [PMID: 37904712 PMCID: PMC10566448 DOI: 10.1002/iid3.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/11/2023] [Accepted: 09/20/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Studies have shown that lidocaine has antioxidative stress, anti-inflammatory, and nerve-protective effects. The current study investigated the effects of lidocaine on cognitive function in rats with cognitive dysfunction. METHODS A total of 48 rats were randomly assigned to four groups of 12 rats each: control group; L (lidocaine) + D (d-galactose) group, d-galactose group (D group); and D + L group. We assessed cognitive function using a Morris water maze (MWM) and pathologic changes of hippocampal sections. An enzyme-linked immunosorbent assay (ELIZA) was used to detect serum malondialdehyde (MDA) and superoxide dismutase (SOD) levels in rats, and protein immunoblotting (western blot) was used to detect brain tissue proteins (collapsing response mediator protein-2 [CRMP2], phosphorylated-collapsing response mediator protein-2 [P-CRMP2], and β-amyloid protein [Aβ]). RESULTS The MWM showed that the d-gal group (284.09 ± 20.46, 5.20 ± 0.793) performed worse than the L + D (265.37 ± 22.34, 4.170 ± 0.577; p = .000) and D + L groups (254.72 ± 27.87, 3.750; p = .000) in escape latency and number of platform crossings, respectively. The L + D group (44.94 ± 2.92 pg/mL, 6.22 ± 0.50 pg/mL, and 460.02 ± 8.26 nmol/mL) and D + L group (46.88 ± 2.63 pg/mL, 5.90 ± 0.38 pg/mL, and 465.6 ± 16.07 nmol/mL) had significantly lower serum inflammatory levels of interleukin-6, tumor necrosis factor-α, and MDA than the d-gal group (57.79 ± 3.96 pg/mL, 11.25 ± 1.70 pg/mL, and 564.9 ± 15.90 nmol/mL), respectively. The L + D group (3.17 ± 0.41 μg/mL) and D + L group (3.08 ± 0.09 μg/mL) had significantly higher serum inflammatory levels of SOD than the d-gal group (2.20 ± 0.13 μg/mL) (all p = .000). The levels of CRMP2, P-CRMP2, and Aβ in the brain tissue homogenates of the L + D group (0.87 ± 0.04, 0.57 ± 0.0, and 0.16 ± 0.02) and the D + L group (0.82 ± 0.05, 0.58 ± 0.09, and 0.15 ± 0.02) were significantly different than the d-gal group (0.67 ± 0.03, 0.96 ± 0.040, and 0.29 ± 0.05). CONCLUSIONS Lidocaine was shown to reduce cognitive impairment in rats with cognitive dysfunction through anti-inflammatory and antioxidative stress mechanisms in combination with CRMP2 antiphosphorylation.
Collapse
Affiliation(s)
- Xiaohong Zheng
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Yuerong Lin
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Linshen Huang
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| | - Xianzhong Lin
- Department of Anesthesiology, First Affiliated HospitalFujian Medical UniversityFuzhouChina
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated HospitalFujian Medical UniversityFuzhouChina
| |
Collapse
|
6
|
Shin SW, Cho IH. Panax ginseng as a potential therapeutic for neurological disorders associated with COVID-19; Toward targeting inflammasome. J Ginseng Res 2023; 47:23-32. [PMID: 36213093 PMCID: PMC9529349 DOI: 10.1016/j.jgr.2022.09.004] [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: 06/22/2022] [Revised: 08/15/2022] [Accepted: 09/27/2022] [Indexed: 01/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious respiratory disease caused by a severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). SARS-CoV-2 infection may cause clinical manifestations of multiple organ damage, including various neurological syndromes. There are currently two oral antiviral drugs-Paxlovid and molnupiravir-that are recognized to treat COVID-19, but there are still no drugs that can specifically fight the challenges of SARS-CoV-2 variants. Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) inflammasome is a multimolecular complex that can sense heterogeneous pathogen-associated molecular patterns associated with neurological disorders. The NLRP3 activation stimulates the production of caspase-1-mediated interleukin (IL)-1β, IL-18, and other cytokines in immune cells. Panax (P.) ginseng is a medicinal plant that has traditionally been widely used to boost immunity and treat various pathological conditions in the nervous system due to its safety and anti-inflammatory/oxidant/viral activities. Several recent reports have indicated that P. ginseng and its active ingredients may regulate NLRP3 inflammasome activation in the nervous system. Therefore, this review article discusses the current knowledge regarding the pathogenesis of neurological disorders related to COVID-19 and NLRP3 inflammasome activation and the possibility of using P. ginseng in a strategy targeting this pathway to treat neurological disorders.
Collapse
Affiliation(s)
- Seo Won Shin
- Department of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ik Hyun Cho
- Department of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea,Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea,Corresponding author. D.V.M. & Ph.D. Department of Convergence Medical Science and Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
7
|
Guo C, Kong X, Fan Y, Zhang R. Aerobic Treadmill Exercise Upregulates Epidermal Growth Factor Levels and Improves Learning and Memory in d-galactose-Induced Aging in a Mouse Model. Am J Alzheimers Dis Other Demen 2023; 38:15333175231211082. [PMID: 37977137 PMCID: PMC10657526 DOI: 10.1177/15333175231211082] [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] [Indexed: 11/19/2023]
Abstract
Previous studies have demonstrated that exercise improves cognitive function in Alzheimer's disease mice but the exact mechanism needs further studies. This research aimed to study the effects of aerobic treadmill exercise on epidermal growth factor (EGF) levels and learning and memory in d-galactose-induced aging in a mouse model. Forty male Kunming mice were analyzed in this study and randomly divided into 4 groups: control (C group), aerobic exercise (AE group), d-galactose (D-gal group), and d-galactose + aerobic exercise (D-gal + AE group). The C and AE groups received a daily mid-scapular subcutaneous injection of .9% saline for 40 days. Mice in the D-gal and D-gal + AE groups were subcutaneously injected with d-galactose (1.25 mg/kg) once daily for 40 days. The mice in the AE group and D-gal + AE group completed 40 days of aerobic treadmill exercise. Learning and memory were evaluated by step-down tests. Specifically, 24 h after the behavioral test, blood was collected and brain tissue was extracted, and superoxide dismutase (SOD) and acetylcholinesterase activities were detected. The neurons in the CA1 and CA3 regions of the hippocampus were counted by Nissl staining. The number of EGF-positive cells was observed by immunohistochemical methods. In the learning test, the reaction time in the D-gal group increased significantly (P < .05), while the error numbers in the D-gal group tended to decrease compared with AE, D-gal + AE, and C groups. In the memory test, the latency of mice in the D-gal group was lower, while the error in this group was higher than in the other groups (P < .05). The activities of SOD and acetylcholinesterase were lower in the D-gal group than in the other groups (P < .05). The number of EGF-positive cells and neurons in the hippocampal CA1 and CA3 regions in the D-gal + AE group was higher compared to those in the D-gal group (P < .05), and lower in groups with mice that were not injected with d-galactose. Aerobic treadmill exercise inhibited SOD activity, increased EGF-positive cells, and decreased neuronal death and apoptosis, thereby improving learning and memory in the mouse model of d-galactose-induced aging.
Collapse
Affiliation(s)
- Cheng Guo
- Capital University of Physical Education and Sports, Beijing, China
| | - Xiaoyang Kong
- Capital University of Physical Education and Sports, Beijing, China
| | - Yongzhao Fan
- Department of Physical Education, Henan Normal University, Xinxiang, China
| | | |
Collapse
|
8
|
Feng H, Xue M, Deng H, Cheng S, Hu Y, Zhou C. Ginsenoside and Its Therapeutic Potential for Cognitive Impairment. Biomolecules 2022; 12:1310. [PMID: 36139149 PMCID: PMC9496100 DOI: 10.3390/biom12091310] [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: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cognitive impairment (CI) is one of the major clinical features of many neurodegenerative diseases. It can be aging-related or even appear in non-central nerve system (CNS) diseases. CI has a wide spectrum that ranges from the cognitive complaint with normal screening tests to mild CI and, at its end, dementia. Ginsenosides, agents extracted from a key Chinese herbal medicine (ginseng), show great promise as a new therapeutic option for treating CI. This review covered both clinical trials and preclinical studies to summarize the possible mechanisms of how ginsenosides affect CI in different diseases. It shows that ginsenosides can modulate signaling pathways associated with oxidative stress, apoptosis, inflammation, synaptic plasticity, and neurogenesis. The involved signaling pathways mainly include the PI3K/Akt, CREB/BDNF, Keap1/Nrf2 signaling, and NF-κB/NLRP3 inflammasome pathways. We hope to provide a theoretical basis for the treatment of CI for related diseases by ginsenosides.
Collapse
Affiliation(s)
- Hui Feng
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Mei Xue
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Hao Deng
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300073, China
| | - Shiqi Cheng
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330008, China
| | - Yue Hu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210024, China
| |
Collapse
|
9
|
Suzen S, Tucci P, Profumo E, Buttari B, Saso L. A Pivotal Role of Nrf2 in Neurodegenerative Disorders: A New Way for Therapeutic Strategies. Pharmaceuticals (Basel) 2022; 15:ph15060692. [PMID: 35745610 PMCID: PMC9227112 DOI: 10.3390/ph15060692] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/22/2022] [Accepted: 05/28/2022] [Indexed: 02/04/2023] Open
Abstract
Clinical and preclinical research indicates that neurodegenerative diseases are characterized by excess levels of oxidative stress (OS) biomarkers and by lower levels of antioxidant protection in the brain and peripheral tissues. Dysregulations in the oxidant/antioxidant balance are known to be a major factor in the pathogenesis of neurodegenerative diseases and involve mitochondrial dysfunction, protein misfolding, and neuroinflammation, all events that lead to the proteostatic collapse of neuronal cells and their loss. Nuclear factor-E2-related factor 2 (Nrf2) is a short-lived protein that works as a transcription factor and is related to the expression of many cytoprotective genes involved in xenobiotic metabolism and antioxidant responses. A major emerging function of Nrf2 from studies over the past decade is its role in resistance to OS. Nrf2 is a key regulator of OS defense and research supports a protective and defending role of Nrf2 against neurodegenerative conditions. This review describes the influence of Nrf2 on OS and in what way Nrf2 regulates antioxidant defense for neurodegenerative conditions. Furthermore, we evaluate recent research and evidence for a beneficial and potential role of specific Nrf2 activator compounds as therapeutic agents.
Collapse
Affiliation(s)
- Sibel Suzen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Tandogan, 06100 Ankara, Turkey
- Correspondence: ; Tel.: +90-533-391-5844
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy;
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (E.P.); (B.B.)
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (E.P.); (B.B.)
| | - Luciano Saso
- Department of Physiology and Pharmacology ‘‘Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
10
|
Jung EM, Lee GS. Korean Red Ginseng, a regulator of NLRP3 inflammasome, in the COVID-19 pandemic. J Ginseng Res 2022; 46:331-336. [PMID: 35194373 PMCID: PMC8851744 DOI: 10.1016/j.jgr.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) exhibits various symptoms, ranging from asymptomatic to severe pneumonia or death. The major features of patients in severe COVID-19 are the dysregulation of cytokine secretion, pneumonia, and acute lung injury. Consequently, it leads to acute respiratory distress syndrome, disseminated intravascular coagulation, multiple organ failure, and death. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of COVID-19, influences nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3 (NLRP3), the sensor of inflammasomes, directly or indirectly, culminating in the assembly of NLRP3 inflammasome and activation of inflammatory caspases, which induce the inflammatory disruption in severe COVID-19. Accordingly, the target therapeutics for inflammasome has attracted attention as a treatment for COVID-19. Korean Red Ginseng (KRG) inhibits several inflammatory responses, including the NLRP3 inflammasome signaling. This review discusses the role of KRG in the treatment and prevention of COVID-19 based on its anti-NLRP3 inflammasome efficacy.
Collapse
Affiliation(s)
- Eui-Man Jung
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
11
|
Yi YS. Potential benefits of ginseng against COVID-19 by targeting inflammasomes. J Ginseng Res 2022; 46:722-730. [PMID: 35399195 PMCID: PMC8979607 DOI: 10.1016/j.jgr.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogenic virus that causes coronavirus disease 2019 (COVID-19), with major symptoms including hyper-inflammation and cytokine storm, which consequently impairs the respiratory system and multiple organs, or even cause death. SARS-CoV-2 activates inflammasomes and inflammasome-mediated inflammatory signaling pathways, which are key determinants of hyperinflammation and cytokine storm in COVID-19 patients. Additionally, SARS-CoV-2 inhibits inflammasome activation to evade the host's antiviral immunity. Therefore, regulating inflammasome initiation has received increasing attention as a preventive measure in COVID-19 patients. Ginseng and its major active constituents, ginsenosides and saponins, improve the immune system and exert anti-inflammatory effects by targeting inflammasome stimulation. Therefore, this review discussed the potential preventive and therapeutic roles of ginseng in COVID-19 based on its regulatory role in inflammasome initiation and the host's antiviral immunity.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, 16227, Republic of Korea.
| |
Collapse
|
12
|
Tastan B, Arioz BI, Genc S. Targeting NLRP3 Inflammasome With Nrf2 Inducers in Central Nervous System Disorders. Front Immunol 2022; 13:865772. [PMID: 35418995 PMCID: PMC8995746 DOI: 10.3389/fimmu.2022.865772] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/04/2022] [Indexed: 12/15/2022] Open
Abstract
The NLRP3 inflammasome is an intracellular multiprotein complex that plays an essential role in the innate immune system by identifying and eliminating a plethora of endogenous and exogenous threats to the host. Upon activation of the NLRP3 complex, pro-inflammatory cytokines are processed and released. Furthermore, activation of the NLRP3 inflammasome complex can induce pyroptotic cell death, thereby propagating the inflammatory response. The aberrant activity and detrimental effects of NLRP3 inflammasome activation have been associated with cardiovascular, neurodegenerative, metabolic, and inflammatory diseases. Therefore, clinical strategies targeting the inhibition of the self-propelled NLRP3 inflammasome activation are required. The transcription factor Nrf2 regulates cellular stress response, controlling the redox equilibrium, metabolic programming, and inflammation. The Nrf2 pathway participates in anti-oxidative, cytoprotective, and anti-inflammatory activities. This prominent regulator, through pharmacologic activation, could provide a therapeutic strategy for the diseases to the etiology and pathogenesis of which NLRP3 inflammasome contributes. In this review, current knowledge on NLRP3 inflammasome activation and Nrf2 pathways is presented; the relationship between NLRP3 inflammasome signaling and Nrf2 pathway, as well as the pre/clinical use of Nrf2 activators against NLRP3 inflammasome activation in disorders of the central nervous system, are thoroughly described. Cumulative evidence points out therapeutic use of Nrf2 activators against NLRP3 inflammasome activation or diseases that NLRP3 inflammasome contributes to would be advantageous to prevent inflammatory conditions; however, the side effects of these molecules should be kept in mind before applying them to clinical practice.
Collapse
Affiliation(s)
- Bora Tastan
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Burak I. Arioz
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Sermin Genc
- Genc Laboratory, Izmir Biomedicine and Genome Center, Izmir, Turkey,Department of Neuroscience, Health Sciences Institute, Dokuz Eylul University, Izmir, Turkey,*Correspondence: Sermin Genc,
| |
Collapse
|
13
|
Zhang M, Wu Y, Gao R, Chen X, Chen R, Chen Z. Glucagon-like peptide-1 analogs mitigate neuroinflammation in Alzheimer's disease by suppressing NLRP2 activation in astrocytes. Mol Cell Endocrinol 2022; 542:111529. [PMID: 34906628 DOI: 10.1016/j.mce.2021.111529] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 01/15/2023]
Abstract
Neuroinflammation is closely linked to the pathogenesis of Alzheimer's disease (AD). Glucagon-like peptide-1 (GLP-1) analogs exhibit anti-inflammatory and neuroprotective effects; hence, we investigated whether they reduce cognitive impairment and protect astrocytes from oxidative stress. We found that 5 × FAD transgenic mice treated with the synthetic GLP-1 receptor agonist exenatide had improved cognitive function per the Morris water maze test. Immunohistochemistry, western blotting, and ELISAs used to detect inflammatory factors revealed reduced neuroinflammation in extracted piriform cortexes of exenatide-treated mice as well as lower amyloid β1-42-induced oxidative stress and inflammation in astrocytes treated with exendin-4 (the natural analog of exenatide). Adenovirus-mediated overexpression of nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 2 (NLRP2) revealed that exenatide/exendin-4 function may be attributed to NLRP2 inflammasome inhibition. Collectively, our results indicate that GLP-1 analogs improve cognitive dysfunction in vivo and protect astrocytes in vitro, potentially via the downregulation of the NLRP2 inflammasome.
Collapse
Affiliation(s)
- Mengjun Zhang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Yubin Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Ruonan Gao
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xinwei Chen
- Graduate School of Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Ruiyu Chen
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Zhou Chen
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
14
|
Zeng Y, Chen Y, Zhang S, Ren H, Xia J, Liu M, Shan B, Ren Y. Natural Products in Modulating Methamphetamine-Induced Neuronal Apoptosis. Front Pharmacol 2022; 12:805991. [PMID: 35058785 PMCID: PMC8764133 DOI: 10.3389/fphar.2021.805991] [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: 10/31/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (METH), an amphetamine-type psychostimulant, is highly abused worldwide. Chronic abuse of METH causes neurodegenerative changes in central dopaminergic neurons with numerous neuropsychiatric consequences. Neuronal apoptosis plays a critical role in METH-induced neurotoxicity and may provide promising pharmacological targets for preventing and treating METH addiction. In recent years, accumulating evidence has revealed that natural products may possess significant potentials to inhibit METH-evoked neuronal apoptosis. In this review, we summarized and analyzed the improvement effect of natural products on METH-induced neuronal apoptosis and their potential molecular mechanisms on modulating dopamine release, oxidative stress, mitochondrial-dependent apoptotic pathway, endoplasmic reticulum stress-mediated apoptotic pathway, and neuroinflammation. Hopefully, this review may highlight the potential value of natural products in modulating METH-caused neuronal apoptosis and provide useful information for future research and developments of novel and efficacious pharmacotherapies in this field.
Collapse
Affiliation(s)
- Yiwei Zeng
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunhui Chen
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Su Zhang
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jialin Xia
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengnan Liu
- Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, China
| | - Baozhi Shan
- School of Humanities, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yulan Ren
- College of Acupuncture-moxibustion and Tuina, College of Basic Medicine, College of Nursing, College of Chinese Classics, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Mizuno Y, Abolhassani N, Mazzei G, Saito T, Saido TC, Yamasaki R, Kira JI, Nakabeppu Y. Deficiency of MTH1 and/or OGG1 increases the accumulation of 8-oxoguanine in the brain of the App NL-G-F/NL-G-F knock-in mouse model of Alzheimer's disease, accompanied by accelerated microgliosis and reduced anxiety-like behavior. Neurosci Res 2021; 177:118-134. [PMID: 34838904 DOI: 10.1016/j.neures.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 11/12/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022]
Abstract
Oxidative stress is a major risk factor for Alzheimer's disease (AD). Among various oxidized molecules, the marked accumulation of an oxidized form of guanine, 8-oxo-7,8-dihydroguanine (8-oxoG), is observed in the AD brain. 8-oxo-2'-deoxyguanosine triphosphatase (MTH1) and 8-oxoG DNA glycosylase (OGG1) minimize the 8-oxoG accumulation in DNA, and their expression is decreased in the AD brain. MTH1 and/or OGG1 may suppress the pathogenesis of AD; however, their exact roles remain unclear. We evaluated the roles of MTH1 and OGG1 during the pathogenesis of AD using AppNL-G-F/NL-G-F knock-in mice (a preclinical AD model). Six-month-old female AppNL-G-F/NL-G-F mice with MTH1 and/or OGG1 deficiency exhibited reduced anxiety-related behavior, but their cognitive and locomotive functions were unchanged; the alteration was less evident in 12-month-old mice. MTH1 and/or OGG1 deficiency accelerated the 8-oxoG accumulation and microgliosis in the amygdala and cortex of six-month-old mice; the alteration was less evident in 12-month-old mice. Astrocytes and neurons were not influenced. We showed that MTH1 and OGG1 are essential for minimizing oxidative DNA damage in the AppNL-G-F/NL-G-F brain, and the effects are age-dependent. MTH1 and/or OGG1 deficiency reduced anxiety-related behavior in AppNL-G-F/NL-G-F mice with a significant acceleration of the 8-oxoG burden and microgliosis, especially in the cortex and amygdala.
Collapse
Affiliation(s)
- Yuri Mizuno
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nona Abolhassani
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Guianfranco Mazzei
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan; Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Translational Neuroscience Center, Graduate School of Medicine, School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Okawa, Fukuoka, 831-8501, Japan
| | - Yusaku Nakabeppu
- Division of Neurofunctional Genomics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
16
|
Yi YS. New mechanisms of ginseng saponin-mediated anti-inflammatory action via targeting canonical inflammasome signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114292. [PMID: 34089812 DOI: 10.1016/j.jep.2021.114292] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is an ethnopharmacological herbal plant in Asian countries, particularly in Korea, China, and Japan. Ginseng saponins, including ginsenosides, are major active components in ginseng and have been demonstrated to have numerous pharmacological effects on various human diseases. AIM OF THE REVIEW Many previous studies investigating the anti-inflammatory effect of ginseng saponins have mostly focused on the 'priming' step rather than the 'triggering' step. This review aims to discuss the studies investigating an inhibitory role of ginseng saponins in inflammasome activation of the triggering step. MATERIALS AND METHODS The literature was explored using the search strings, such as "ginseng saponins and inflammasomes" and "ginsenosides and inflammasomes" in several resources, such as PubMed, Google Scholar, and Scopus databases. RESULTS Various ginseng saponins of Panax ginseng, Panax japonicas, and Panax quinquefolius alleviated inflammatory responses and diseases by inhibiting the nucleotide-binding oligomerization domain-like receptor (NLR) P3 (NLRP3) inflammasome activation. Also, ginseng saponin, Rg1 of Panax ginseng alleviated neuroinflammation and diseases by inhibiting NLRP1 inflammasome activation. Finally, ginseng saponins, Rh1 and Rg3 in Korea red ginseng (KRG) of Panax ginseng ameliorated sepsis by inhibiting absent in melanoma 2 (AIM2) inflammasome activation. CONCLUSION The studies discussed in this review provide insight into the new paradigm of the ginseng saponins as the promising anti-inflammatory agents that could be ethnopharmacologically used to prevent and treat inflammatory and inflammation-induced disorders via targeting inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| |
Collapse
|
17
|
Pseudoginsenoside-F11 attenuates cognitive dysfunction and tau phosphorylation in sporadic Alzheimer's disease rat model. Acta Pharmacol Sin 2021; 42:1401-1408. [PMID: 33277592 PMCID: PMC8379201 DOI: 10.1038/s41401-020-00562-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
We previously reported that pseudoginsenoside-F11 (PF11), an ocotillol-type saponin, significantly ameliorated Alzheimer's disease (AD)-associated cognitive defects in APP/PS1 and SAMP8 mice by inhibiting Aβ aggregation and tau hyperphosphorylation, suggesting a potential therapeutic effect of PF11 in the treatment of AD. In the present study we further evaluated the therapeutic effects of PF11 on relieving cognitive impairment in a rat model of sporadic AD (SAD). SAD was induced in rats by bilateral icv infusion of streptozotocin (STZ, 3 mg/kg). The rats were treated with PF11 (2, 4, 8 mg·kg-1·d-1, ig) or a positive control drug donepezil (5 mg·kg-1·d-1, ig) for 4 weeks. Their cognitive function was assessed in the nest building, Y-maze, and Morris water maze tests. We showed that STZ icv infusion significantly affected the cognitive function, tau phosphorylation, and insulin signaling pathway in the hippocampus. Furthermore, STZ icv infusion resulted in significant upregulation of the calpain I/cyclin-dependent protein kinase 5 (CDK5) signaling pathway in the hippocampus. Oral administration of PF11 dose-dependently ameliorated STZ-induced learning and memory defects. In addition, PF11 treatment markedly reduced the neuronal loss, protected the synapse structure, and modulated STZ-induced expression of tau phosphorylation by regulating the insulin signaling pathway and calpain I/CDK5 signaling pathway in the hippocampus. Donepezil treatment exerted similar beneficial effects in STZ-infused rats as the high dose of PF11 did. This study highlights the excellent therapeutic potential of PF11 in managing AD.
Collapse
|
18
|
Gao Y, Liu Y, Yang X, Zhang T, Hou Y, Wang P, Liu Y, Yuan L, Zhang H, Wu C, Yang J. Pseudoginsenoside-F11 ameliorates thromboembolic stroke injury in rats by reducing thromboinflammation. Neurochem Int 2021; 149:105108. [PMID: 34175350 DOI: 10.1016/j.neuint.2021.105108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/30/2021] [Accepted: 06/18/2021] [Indexed: 10/21/2022]
Abstract
Pseudoginsenoside-F11 (PF11), an ocotillol-type ginsenoside, has been reported to exert neuroprotective effects on ischemic stroke induced by permanent and transient middle cerebral artery occlusion in experimental animals. The aim of the present study was to investigate the effect of PF11 on thromboembolic stroke in rats and its possible mechanisms on thromboinflammation. PF11 (4, 12, 36 mg/kg) was injected intravenously (i.v.) once a day for 3 consecutive days to male Wistar rats followed by embolic middle cerebral artery occlusion (eMCAO). The results showed that PF11 significantly reduced the cerebral infarction volume, brain edema and neurological deficits induced by eMCAO. Meanwhile, the thromboinflammation in the ischemic hemisphere was observed at 24 h after eMCAO, as indicated by the increased number of microvascular thrombus and inflammatory response. Moreover, eMCAO resulted in the up-regulation of platelet glycoprotein Ibα (GPIbα) and VI (GPVI), as well as the activation of contact-kinin pathway. Notably, PF11 significantly reversed all these changes. Furthermore, PF11 prevented the eMCAO-induced loss of tight junction proteins and up-regulation of matrix metalloproteinase-9 (MMP-9), thus leading to the alleviation of blood-brain barrier (BBB) damage. In conclusion, the present study revealed that thromboinflammation was induced in the ischemic hemisphere of rats after eMCAO and PF11 exerted marked protective effects against thromboembolic stroke by attenuating thromboinflammation and preventing BBB damage. This research further identifies the potential therapeutic role of PF11 for ischemic stroke.
Collapse
Affiliation(s)
- Yongfeng Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Institute of Pharmacology, Shandong First Medical University, Shandong Academy of Medical Science, Tan'an, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Xue Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Tianyu Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Ying Hou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Pengwei Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yinglu Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Linlin Yuan
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Haotian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China.
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
19
|
Chu J, Wang J, Cui L, Liu S, An N, Han J, Che X, Wu C, Yang J. Pseudoginsenoside-F11 ameliorates okadiac acid-induced learning and memory impairment in rats via modulating protein phosphatase 2A. Mech Ageing Dev 2021; 197:111496. [PMID: 33957218 DOI: 10.1016/j.mad.2021.111496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/15/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
We have reported that pseudoginsenoside-F11 (PF11) can significantly improve the cognitive impairments in several Alzheimer's disease (AD) models, but the mechanism has not been fully elucidated. In the present study, the effects of PF11 on AD, in particular the underlying mechanisms related with protein phosphatase 2A (PP2A), were investigated in a rat model induced by okadaic acid (OA), a selective inhibitor of PP2A. The results showed that PF11 treatment dose-dependently improved the learning and memory impairments in OA-induced AD rats. PF11 could significantly inhibit OA-induced tau hyperphosphorylation, suppress the activation of glial cells, alleviate neuroinflammation, thus rescue the neuronal and synaptic damage. Further investigation revealed that PF11 could regulate the protein expression of methyl modifying enzymes (leucine carboxyl methyltransferase-1 and protein phosphatase methylesterase-1) in the brain, thus increase methyl-PP2A protein expression and indirectly increase the activity of PP2A. Molecular docking analysis, structural alignment and in vitro results showed that PF11 was similar in the shape and electrostatic field feature to a known activator of PP2A, and could directly bind and activate PP2A. In conclusion, the present data indicate that PF11 can ameliorate OA-induced learning and memory impairment in rats via modulating PP2A.
Collapse
Affiliation(s)
- Jinxiu Chu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China; Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, College of Elementary Medicine, North China University of Science and Technology, Tangshan 063210, PR China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Lijuan Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shuai Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Nina An
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jian Han
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
20
|
Zhou F, Zhang X, Jiang L, Li S, Chen Y, Wu J. Pseudoginsenoside F11 Enhances the Viability of Random-Pattern Skin Flaps by Promoting TFEB Nuclear Translocation Through AMPK-mTOR Signal Pathway. Front Pharmacol 2021; 12:667524. [PMID: 33995096 PMCID: PMC8116945 DOI: 10.3389/fphar.2021.667524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
Random-pattern skin flap is widely used in tissue reconstruction. However, necrosis occurring in the distal part of the flap limits its clinical application to some extent. Activation of autophagy has been considered as an effective approach to enhance the survival of skin flaps. Pseudoginsenoside F11 (PF11), an ocotillol-type saponin, is an important component of Panax quinquefolium which has been shown to confer protection against cerebral ischemia and alleviate oxidative stress. However, it is currently unknown whether PF11 induces autophagy to improve the survival of skin flaps. In this study, we investigated the effects of PF11 on blood flow and tissue edema. The results of histological examination and western blotting showed that PF11 enhanced angiogenesis, alleviated apoptosis and oxidative stress, thereby improving the survival of the flap. Further experiments showed that PF11 promoted nuclear translocation of TFEB and by regulating the phosphorylation of AMPK. In summary, this study demonstrates that PF11 activates autophagy through the AMPK-TFEB signal pathway in skin flaps and it could be a promising strategy for enhancing flap viability.
Collapse
Affiliation(s)
- Feiya Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Xian Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Liangfu Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Shi Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Yiheng Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Jianbin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
21
|
Han H, Liu Z, Yin J, Gao J, He L, Wang C, Hou R, He X, Wang G, Li T, Yin Y. D-Galactose Induces Chronic Oxidative Stress and Alters Gut Microbiota in Weaned Piglets. Front Physiol 2021; 12:634283. [PMID: 33897450 PMCID: PMC8060641 DOI: 10.3389/fphys.2021.634283] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/12/2021] [Indexed: 01/17/2023] Open
Abstract
Oxidative stress commonly occurs in pig production, which can severely damage the intestinal function of weaned piglets. This study was conducted to investigate the effects of D-galactose with different levels used to induce chronic oxidative stress on growth performance, intestinal morphology and gut microbiota in weaned piglets. The results showed that addition of 10 and 20 g/kg BW D-galactose reduced average daily gain and average daily feed intake from the first to the third week. 10 g/kg BW D-galactose increased the concentration of serum MDA at the second and third week. 10 g/kg BW D-galactose significantly influenced the jejunal and ileal expressions of GPx1, CAT1, and MnSOD. The results of 16S rRNA sequencing showed that compared with the control, 10 and 20 g/kg BW D-galactose significantly decreased the relative abundance of Tenericutes, Erysipelotrichia, Erysipelotrichales, and Erysipelotrichaceae, while increased the relative abundance of Negativicutes, Selenomonnadales, and Veillonellaceae. The results indicated that treatment with 10 g/kg BW/day D-galactose for 3 weeks could induce chronic oxidative stress, reduce the growth performance and alter gut microbiota in weaned piglets.
Collapse
Affiliation(s)
- Hui Han
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zemin Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jing Gao
- Research Institute of Oil Tea Camellia, Hunan Academy of Forestry, Changsha, China.,National Engineering Research Center for Oil Tea Camellia, Changsha, China
| | - Liuqin He
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Chenyu Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ruoxin Hou
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xingguo He
- Changsha Lvye Bio-Technology Co., Ltd., Changsha, China
| | - Guoqiang Wang
- Changsha Lvye Bio-Technology Co., Ltd., Changsha, China
| | - Tiejun Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China.,Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
22
|
Dwivedi S, Kushalan S, Paithankar JG, D'Souza LC, Hegde S, Sharma A. Environmental toxicants, oxidative stress and health adversities: interventions of phytochemicals. J Pharm Pharmacol 2021; 74:516-536. [PMID: 33822130 DOI: 10.1093/jpp/rgab044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Oxidative stress is the most common factor mediating environmental chemical-induced health adversities. Recently, an exponential rise in the use of phytochemicals as an alternative therapeutics against oxidative stress-mediated diseases has been documented. Due to their free radical quenching property, plant-derived natural products have gained substantial attention as a therapeutic agent in environmental toxicology. The present review aimed to describe the therapeutic role of phytochemicals in mitigating environmental toxicant-mediated sub-cellular and organ toxicities via controlling cellular antioxidant response. METHODS The present review has covered the recently related studies, mainly focussing on the free radical scavenging role of phytochemicals in environmental toxicology. KEY FINDINGS In vitro and in vivo studies have reported that supplementation of antioxidant-rich compounds can ameliorate the toxicant-induced oxidative stress, thereby improving the health conditions. Improving the cellular antioxidant pool has been considered as a mode of action of phytochemicals. However, the other cellular targets of phytochemicals remain uncertain. CONCLUSIONS Knowing the therapeutic value of phytochemicals to mitigate the chemical-induced toxicity is an initial stage; mechanistic understanding needs to decipher for development as therapeutics. Moreover, examining the efficacy of phytochemicals against mixer toxicity and identifying the bioactive molecule are major challenges in the field.
Collapse
Affiliation(s)
- Shiwangi Dwivedi
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Sharanya Kushalan
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Jagdish Gopal Paithankar
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Smitha Hegde
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| |
Collapse
|
23
|
Cao Y, Wang K, Xu S, Kong L, Bi Y, Li X. Recent Advances in the Semisynthesis, Modifications and Biological Activities of Ocotillol-Type Triterpenoids. Molecules 2020; 25:E5562. [PMID: 33260848 PMCID: PMC7730845 DOI: 10.3390/molecules25235562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
Ginseng is one of the most widely consumed herbs in the world and plays an important role in counteracting fatigue and alleviating stress. The main active substances of ginseng are its ginsenosides. Ocotillol-type triterpenoid is a remarkably effective ginsenoside from Vietnamese ginseng that has received attention because of its potential antibacterial, anticancer and anti-inflammatory properties, among others. The semisynthesis, modification and biological activities of ocotillol-type compounds have been extensively studied in recent years. The aim of this review is to summarize semisynthesis, modification and pharmacological activities of ocotillol-type compounds. The structure-activity relationship studies of these compounds were reported. This summary should prove useful information for drug exploration of ocotillol-type derivatives.
Collapse
Affiliation(s)
| | | | | | | | - Yi Bi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; (Y.C.); (K.W.); (S.X.); (L.K.); (X.L.)
| | | |
Collapse
|
24
|
Wu R, Wang H, Lv X, Shen X, Ye G. Rapid action of mechanism investigation of Yixin Ningshen tablet in treating depression by combinatorial use of systems biology and bioinformatics tools. JOURNAL OF ETHNOPHARMACOLOGY 2020; 257:112827. [PMID: 32276008 DOI: 10.1016/j.jep.2020.112827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/17/2020] [Accepted: 03/31/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yixin Ningshen tablet is a CFDA-approved TCM formula for treating depression clinically. However, little is known about its active compounds and related potential target proteins, so far, no researches have been performed to investigate its mechanism of action for the treatment of depression. AIM OF THE STUDY Here we develop an original bioinformatics pipeline composed of text mining tools, database querying and systems biology combinatorial analysis, which is applied to rapidly explore the mechanism of action of Yixin Ningshen tablet in treating depression. MATERIALS AND METHODS Text mining and database query were applied to identify active compounds in Yixin Ningshen tablet for the treatment of depression. Then SwissTargetPrediction was used to predict their potential target proteins. PubMed was retrieved to summarize known depression related systems biology results. Ingenuity Pathway Analysis (IPA) tools and STRING were applied to construct a compound-target protein-gene protein-differential protein-differential metabolite network with the integration of compound-target interaction and systems biology results, as well as enrich the target proteins related pathways. ChEMBL and CDOCKER were used to validate the compound-target interactions. RESULTS 62 active compounds and their 286 potential target proteins were identified in Yixin Ningshen tablet for the treatment of depression. The construction of compound-target protein-gene protein-differential protein-differential metabolite network shrinked the number of potential target proteins from 286 to 133. Pathway enrichment analysis of target proteins indicated that Neuroactive ligand-receptor interaction, Calcium signaling pathway, Serotonergic synapse, cAMP signaling pathway and Gap junction were the common primary pathways regulated by both Yixin Ningshen Tablet and anti-depressant drugs, and MAPK, Relaxin, AGE-RAGE, Estrogen, HIF-1, Jak-STAT signaling pathway, Endocrine resistance, Arachidonic acid metabolism and Regulation of actin cytoskeleton were the specifically main pathways regulated by Yixin Ningshen tablet for the treatment of depression. Further validations based on references and molecular docking results demonstrated that Yixin Ningshen tablet could primarily target MAPT, CHRM1 and DRD1, thus regulating serotonergic neurons, cholinergic transmission, norepinephrine and dopamine reuptake for the treatment of depression. CONCLUSIONS This study displays the power of extensive mining of public data and bioinformatical repositories to provide answers for a specific pharmacological question. It furthermore demonstrates how the usage of such a combinatorial approach is advantageous for the biologist in terms of experimentation time and costs.
Collapse
Affiliation(s)
- Ruoming Wu
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, 201203, China.
| | - Huijun Wang
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, 201203, China; The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xing Lv
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, 201203, China.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, 310000, China.
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd., Shanghai, 201203, China.
| |
Collapse
|
25
|
Wang W, Yang L, Liu T, Ma Y, Huang S, He M, Wang J, Wen A, Ding Y. Corilagin ameliorates sleep deprivation-induced memory impairments by inhibiting NOX2 and activating Nrf2. Brain Res Bull 2020; 160:141-149. [DOI: 10.1016/j.brainresbull.2020.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/07/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
|
26
|
Wu XL, Deng MZ, Gao ZJ, Dang YY, Li YC, Li CW. Neferine alleviates memory and cognitive dysfunction in diabetic mice through modulation of the NLRP3 inflammasome pathway and alleviation of endoplasmic-reticulum stress. Int Immunopharmacol 2020; 84:106559. [DOI: 10.1016/j.intimp.2020.106559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/13/2020] [Accepted: 04/30/2020] [Indexed: 12/26/2022]
|
27
|
Protective Effect of Ocotillol, the Derivate of Ocotillol-Type Saponins in Panax Genus, against Acetic Acid-Induced Gastric Ulcer in Rats Based on Untargeted Metabolomics. Int J Mol Sci 2020; 21:ijms21072577. [PMID: 32276345 PMCID: PMC7177626 DOI: 10.3390/ijms21072577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023] Open
Abstract
Gastric ulcer (GU), a prevalent digestive disease, has a high incidence and is seriously harmful to human health. Finding a natural drug with a gastroprotective effect is needed. Ocotillol, the derivate of ocotillol-type saponins in the Panax genus, possesses good anti-inflammatory activity. The study aimed to investigate the gastroprotective effect of ocotillol on acetic acid-induced GU rats. The serum levels of endothelin-1 (ET-1) and nitric oxide (NO), the gastric mucosa levels of epidermal growth factor, superoxide dismutase and NO were assessed. Hematoxylin and eosin staining of gastric mucosa for pathological changes and immunohistochemical staining of ET-1, epidermal growth factor receptors and inducible nitric oxide synthase were evaluated. A UPLC-QTOF-MS-based serum metabolomics approach was applied to explore the latent mechanism. A total of 21 potential metabolites involved in 7 metabolic pathways were identified. The study helps us to understand the pathogenesis of GU and to provide a potential natural anti-ulcer agent.
Collapse
|
28
|
Anti-Ageing Effect of Physalis alkekengi Ethyl Acetate Layer on a d-galactose-Induced Mouse Model through the Reduction of Cellular Senescence and Oxidative Stress. Int J Mol Sci 2020; 21:ijms21051836. [PMID: 32155871 PMCID: PMC7084245 DOI: 10.3390/ijms21051836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/31/2022] Open
Abstract
We aimed to study the effects of an ethyl acetate fraction of Physalis alkekengi (PAE) on d-galactose (d-gal)-induced senescence and the underlying mechanism. Firstly, analysis of the phytochemical composition revealed total flavonoids, total phenolics, total saponins, rutin, and luteolin contents of 71.72 ± 2.99 mg rutin equivalents/g, 40.19 ± 0.47 mg gallic acid equivalents/g, 128.13 ± 1.04 mg oleanolic acid equivalents/g, 1.67 ± 0.07 mg/g and 1.61 ± 0.01 mg/g, respectively. The mice were treated with d-gal for six weeks, and from the fifth week, the mice were administered with PAE by gavage once a day for five weeks. We found significant d-gal-induced ageing-related changes, such as learning and memory impairment in novel object recognition and Y-maze, fatigue in weight-loaded forced swimming, reduced thymus coefficient, and histopathological injury of the liver, spleen, and hippocampus. The PAE effectively protected from such changes. Further evaluation showed that PAE decreased the senescence-associated β-galactosidase of the liver, spleen, and hippocampus, as well as the oxidative stress of the liver, plasma, and brain. The abundance of flavonoids, phenols, and saponins in PAE may have contributed to the above results. Overall, this study showed the potential application of PAE for the prevention or treatment of ageing-associated disorders.
Collapse
|
29
|
Liu Y, Wu C, Hou Z, Fu X, Yuan L, Sun S, Zhang H, Yang D, Yao X, Yang J. Pseudoginsenoside-F11 Accelerates Microglial Phagocytosis of Myelin Debris and Attenuates Cerebral Ischemic Injury Through Complement Receptor 3. Neuroscience 2020; 426:33-49. [DOI: 10.1016/j.neuroscience.2019.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023]
|
30
|
Yuan L, Sun S, Pan X, Zheng L, Li Y, Yang J, Wu C. Pseudoginsenoside-F11 improves long-term neurological function and promotes neurogenesis after transient cerebral ischemia in mice. Neurochem Int 2020; 133:104586. [DOI: 10.1016/j.neuint.2019.104586] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022]
|
31
|
Pseudoginsenoside-F11 Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Suppressing Neutrophil Infiltration and Accelerating Neutrophil Clearance. Inflammation 2019; 42:1857-1868. [DOI: 10.1007/s10753-019-01047-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
32
|
Zhu X, Liu J, Huang S, Zhu W, Wang Y, Chen O, Xue J. Neuroprotective effects of isoliquiritigenin against cognitive impairment via suppression of synaptic dysfunction, neuronal injury, and neuroinflammation in rats with kainic acid-induced seizures. Int Immunopharmacol 2019; 72:358-366. [PMID: 31030091 DOI: 10.1016/j.intimp.2019.04.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/28/2019] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
Epileptogenesis is a dynamic process initiated by insults to brain and commonly accompanied by cognitive impairment. Isoliquiritigenin (ISL), a flavonoid in licorice, has a broad spectrum of biological effects including anti-inflammatory and antioxidant activities. However, the protective effects of ISL against cognitive impairment in epileptic processes and the underlying molecular mechanism are not well understood. To address these questions, we established an reproducible seizure model by intracerebroventricular injection of kainic acid (KA) in 21-day-old rats; ISL was intraperitoneally administered three times prior to KA injection, and changes in cognitive function; synaptic plasticity; neuronal injury; number of glial cells; and expression of pro-inflammatory cytokines and nuclear factor-like (NRF)2 signaling and NACHT, LRR, and PYD domains-containing protein (NLRP)3 inflammasome components in the hippocampus were examined. Rats with KA-induced seizures showed longer average escape latency and decreases in the number of platform crossings and average time spent in the target quadrant in the Morris water maze; ISL pretreatment reversed this decline in cognitive impairment and increased the protein levels of synaptophysin, postsynaptic density-95 and brain-derived neurotrophic factor while reducing the number of Fluoro Jade B-positive cells, microglia, and astrocytes; cleaved-Caspase-3 and -9 protein levels; and tumor necrosis factor-α, interleukin (IL)-1β, and IL-18 production. It also enhanced the nuclear localization of NRF2, hemeoxygenase-1, and NAD(P)H:quinone oxidoreductase (NQO) 1, and reversed the upregulation of NLRP3 inflammasome components NLRP3 and Caspase-1 induced by KA injection. Thus, ISL protects against cognitive impairment in KA-induced epileptic processes possibly through regulation of NRF2 signaling and the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Xiaobo Zhu
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China
| | - Jiankun Liu
- Department of Ophthalmology, the Second People's Hospital of Jinan City, Jinan 250000, China
| | - Shanying Huang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University, Jinan 250012, China
| | - Weiwei Zhu
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan 250000, China
| | - Yibiao Wang
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China
| | - Ou Chen
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China; Nursing School, Shandong University, Jinan 250012, China.
| | - Jiang Xue
- Department of Pediatrics, the Second Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|