1
|
Lin Z, Sun M. Phytochemical regulation of CaMKII in Alzheimer's disease: A review of molecular mechanisms and therapeutic potential. Pharmacol Res 2025; 216:107790. [PMID: 40409522 DOI: 10.1016/j.phrs.2025.107790] [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: 01/29/2025] [Revised: 04/08/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder that leads to cognitive decline. CaMKII is a calcium-regulated kinase that is crucial for synaptic plasticity and memory. Phytochemicals with diverse origins, safety, and biological activity have attracted considerable attention in AD research. This systematic analysis of phytochemicals targeting CaMKII reveals their neuroprotective mechanisms against AD pathogenesis, highlighting CaMKII as a promising therapeutic target that warrants further preclinical investigation and drug development. We conducted a comprehensive review of the literature of phytochemicals that target CaMKII as a protective mechanism against AD. The search was conducted across multiple databases, including PubMed, Web of Science, China National Knowledge Internet, and Google Scholar, and covered the period from January 2000 to October 2024. A total of 301 articles were retrieved, of which 22 articles were included. The results showed that flavonoid, glycoside, terpene, and polyphenol analogs positively regulated CaMKII expression, whereas alkaloid analogs negatively regulated CaMKII expression. Different components of traditional Chinese medicine played different roles in CaMKII expression. Flavonoid compounds upregulated the expression of SYN, PSD-95, MAP2, and GluR1 to exert neuroprotective effects. Alkaloid and glycoside analogs inhibited Aβ deposition and tau hyperphosphorylation. Terpene analogs upregulated the SYN, PSD-95, NMDAR, BDNF, and PI3K/Akt signaling pathways to exert neuroprotection. Polyphenol analogs upregulated PSD-95, Munc18-1, SNAP25, SYN, and BDNF to exert neuroprotective effects. Emerging evidence demonstrates that select phytochemicals and traditional Chinese medicine compounds exert neuroprotective effects in AD by modulating CaMKII activity, thereby reducing Aβ accumulation, attenuating tau hyperphosphorylation, and enhancing synaptic plasticity, suggesting promising therapeutic potential.
Collapse
Affiliation(s)
- Zhongying Lin
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Miao Sun
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
2
|
Wu Y, Zeng M, Cao B, Zhang B, Zheng X, Feng W. Material basis and mechanism of Ephedra sinica in interfering with wind-chill cold. Int Immunopharmacol 2025; 152:114432. [PMID: 40058103 DOI: 10.1016/j.intimp.2025.114432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/18/2025] [Accepted: 03/03/2025] [Indexed: 03/24/2025]
Abstract
Ephedra sinica has a long history in medicine, i.e., as the first medicinal plant for treating wind-chill colds. However, few studies have shown its material basis and mechanism of action, so this article mainly focuses on these two issues. Rats were stimulated by fan blowing and low temperature for 7 days, following which alkaloid extract (SWJ), non-alkaloid extract (FSWJ), and polysaccharide extract (DT) from Ephedra sinica (MH) were administered for 4 days. First, the body temperature, sweat spots, activity status, and cough-related indexes were detected to screen the pharmacodynamic material basis of MH. Then, the immune cells, ROS, MDA, GSH-PX, SOD, IL-4, IgE, TRPM8, TLR4, p-P65/P65, acetylcholine, and CHRM3 levels were detected to explore the potential mechanism of the SWJ. In addition, BEAS-2B cells were cultured at 26 °C for 12 h to establish a cell injury model induced by cold stimulation. BEAS-2B cells were co-cultured with spleen cells, and the effects of ephedrine (MHJ) and pseudoephedrine (WMHJ) on immune cells and inflammatory factors in this system were detected. The TRPM8 inhibitor (AMG-333) was added 3 h before administration to detect the effects of MHJ and WMHJ on the expression levels of TRPM8, TLR4, and P-P65/P65. Animal experiments showed that SWJ, FSWJ, and DT reduced body temperature and relieved symptoms such as sweating difficulty, listlessness, and cough, and SWJ produced the best effect. Subsequently, it was found that SWJ transformed immune cells, weakened oxidative stress, downregulated IL-4, IgE, TRPM8, TLR4, and P-P65/P65, and upregulated ACH and CHRM3. Cell experiments showed that MHJ and WMHJ upregulated the Ths and Tcs and downregulated IL-6, TNF-α, TRPM8, TLR4, and P-P65/P65. The intervention of AMG-333 affected the TRPM8 and p-P65/P65, but AMG-333 did not regulate TLR4. Taken together, alkaloid extract is the material basis for MH to improve rats' wind-chill cold and may protect rats from injury by regulating the TRPM8/TLR4-NFκB pathway and CHRM3, with ephedrine and pseudoephedrine possibly playing an important role.
Collapse
Affiliation(s)
- Yuanyuan Wu
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Bing Cao
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Beibei Zhang
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| | - Weisheng Feng
- School of Pharmacy, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| |
Collapse
|
3
|
Hao F, Zeng M, Cao B, Liang X, Ye K, Jiao X, Feng W, Zheng X. Neobavaisoflavone Ameliorates Memory Deficits and Brain Damage in Aβ 25-35-Induced Mice by Regulating SIRT1. CNS Neurosci Ther 2024; 30:e70068. [PMID: 39392360 PMCID: PMC11469773 DOI: 10.1111/cns.70068] [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: 08/06/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a common chronic neurodegenerative disease in older people, and there is no specific treatment that can stop or reverse its progression. Neobavaisoflavone (NBIF) is a flavonoid that has been shown to have neuroprotective effects, but its role in AD has not been revealed. The present study investigated the role and mechanism of NBIF on Aβ25-35-induced brain injury. METHODS In this experiment, the AD mouse model was established by injection of Aβ25-35 peptides (200 μM, icv), and Donepezil (Don, 10 mg/kg/days), NBIF-L (15 mg/kg/days), and NBIF-H (30 mg/kg/days) were administered orally for 4 weeks. Learning memory, hippocampal pathological changes, pathological markers, apoptosis, oxidative stress, inflammation, immune cells were measured in mice. Network pharmacology combined with the GEO database led to the identification of SIRT1, a key target for NBIF intervention in AD, and levels of SIRT1, p-STAT3 and FOXO1 were measured. In addition, the antagonistic activity of SIRT1 transfection silencing against NBIF in Aβ25-35-induced in N9 cells and N2a-APP69 cells was investigated to assess whether the effects caused by NBIF were mediated by SIRT1. RESULTS The results showed that NBIF ameliorated learning memory and hippocampal neuronal damage, reduced pathological markers, apoptosis, oxidative stress and neuroinflammation, and modulated immune cells. SIRT1 is a key target for NBIF intervention in AD, and NBIF upregulates SIRT1 and reduces the expression levels of p-STAT3 and FOXO1. Furthermore, silencing SIRT1 effectively reduced the protective effect of NBIF on Aβ25-35-induced N9 cells and N2a-APP69 cells, which indicated that the protective effect of NBIF on AD is related to SIRT1. CONCLUSIONS NBIF ameliorated Aβ25-35-induced brain injury by inhibiting apoptosis, oxidative stress, and neuroinflammation, which may be mediated through SIRT1 signaling. These findings provide a rationale for NBIF in the treatment of AD and help facilitate the development of clinical therapeutic agents for AD.
Collapse
Affiliation(s)
- Fengxiao Hao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Mengnan Zeng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Bing Cao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xiwen Liang
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
| | - Kaili Ye
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xinmian Jiao
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Weisheng Feng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| | - Xiaoke Zheng
- College of PharmacyHenan University of Chinese MedicineZhengzhouChina
- The Engineering and Technology Center for Chinese Medicine Development of Henan ProvinceZhengzhouChina
- Co‐construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.RZhengzhouChina
| |
Collapse
|
4
|
Cao B, Zeng M, Hao F, Hao Z, Liang X, Zhang Z, Wu Y, Zhang Y, Wang R, Feng W, Zheng X. Cornus officinalis Sieb. Et Zucc. attenuates Aβ 25-35-induced mitochondrial damage and neuroinflammation in mice by modulating the ERK pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155709. [PMID: 38735197 DOI: 10.1016/j.phymed.2024.155709] [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: 05/03/2023] [Revised: 02/14/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024]
Abstract
BACKGROUND Cornus officinalis Sieb. Et Zucc. has the efficacy of tonifying the marrow and filling up the essence, breaking up the accumulation and opening up the orifices. Our research team found that CoS extracts were protective against Aβ25-35-induced memory impairment in mice. However, the pharmacodynamic components and mechanisms by which CoS improves AD have yet to be thoroughly explored and investigated. PURPOSE This study focused on exploring the bioactive components and pharmacodynamic mechanisms of CoS aqueous extract underlying mitochondrial damage and neuroinflammation to improve Aβ25-35-induced AD. METHODS AD mouse models were generated using Aβ25-35 brain injections. Different doses of CoS aqueous extract were orally administered to mice for 28 days. The cognitive function, neuronal and synaptic damage, mitochondrial damage (mitochondrial length, mitochondrial fusion fission-related protein expression), neuroglial activation, and immune inflammatory factor and ERK pathway-related protein levels of mice were assessed. The CoS aqueous extracts components were identified using UPLC-TQ/MS and screened for cellular activity. Midivi-1 (Drp1 inhibitor) or PD98059 (ERK inhibitor) was added to Aβ25-35-exposed PC12 cells to assess whether CoS and its active compounds mMorB and CorE regulate mitochondrial fission through ERK/Drp1. PC12-N9 cells were cocultured to investigate whether mMorB and CorE could regulate mitochondrial division through the ERK pathway to modulate neuroinflammation. RESULTS CoS improved exploration and memory in AD mice, reduced synaptic and mitochondrial damage in their hippocampus, and modulated disturbed mitochondrial dynamics. Moreover, CoS inhibited ERK pathway signaling and attenuated abnormal activation of glial cells and secondary immune inflammatory responses. Additionally, in vitro experiments revealed that CoS and its compounds 7β-O-methylmorroniside (mMorB) and Cornusdiridoid E (CorE) ameliorated mitochondrial injury caused by Aβ25-35 in PC12 cells through inhibition of the ERK/Drp1 pathway. Meanwhile, mMorB and CorE ameliorated cellular inflammation by inhibiting the Ras/ERK/CREB signaling pathway. CONCLUSION CoS aqueous extract ameliorates behavioral deficits and brain damage in Aβ25-35-induced AD mice by modulating the ERK pathway to attenuate mitochondrial damage and neuroinflammation, and the compounds mMorB and CorE are the therapeutically active ingredients.
Collapse
Affiliation(s)
- Bing Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Fengxiao Hao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Zhiyou Hao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Xiwen Liang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Zhenkai Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yuanyuan Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yuhan Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Ru Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, China.
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, China.
| |
Collapse
|
5
|
Tian J, Peng Q, Shen Y, Liu X, Li D, Li J, Guo S, Meng C, Xiao Y. Chondroitin sulphate modified MoS 2 nanoenzyme with multifunctional activities for treatment of Alzheimer's disease. Int J Biol Macromol 2024; 266:131425. [PMID: 38583830 DOI: 10.1016/j.ijbiomac.2024.131425] [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: 01/12/2024] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
Nano-MoS2 exhibit oxidoreductase-like activities, and has been shown to effectively eliminate excessive intracellular ROS and inhibit Aβ aggregation, thus demonstrating promising potential for anti-Alzheimer's disease (anti-AD) intervention. However, the low water dispersibility and high toxicity of nano-MoS2 limits its further application. In this study, we developed a chondroitin sulphate (CS)-modified MoS2 nanoenzyme (CS@MoS2) by harnessing the excellent biocompatibility of CS and the exceptional activities of nano-MoS2 to explore its potential in anti-AD research. Promisingly, CS@MoS2 significantly inhibited Aβ1-40 aggregation and prevented toxic injury in SH-SY5Y cells caused by Aβ1-40. In addition, CS@MoS2 protected these cells from oxidative stress damage by regulating ROS production, as well as promoting the activities of SOD and GSH-Px. CS@MoS2 also modulated the intracellular Ca2+ imbalance and downregulated Tau hyperphosphorylation by activating GSK-3β. CS@MoS2 suppressed p-NF-κB (p65) translocation to the nucleus by inhibiting MAPK phosphorylation, and modulated the expression of downstream anti- and proinflammatory cytokines. Owing to its multifunctional activities, CS@MoS2 effectively improved spatial learning, memory, and anxiety in D-gal/AlCl3-induced AD mice. Taken together, these results indicate that CS@MoS2 has significant potential for improving the therapeutic efficacy of the prevention and treatment of AD, while also presenting a novel framework for the application of nanoenzymes.
Collapse
Affiliation(s)
- Jialei Tian
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, Shandong, China
| | - Qian Peng
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, Shandong, China
| | - Yuzhen Shen
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, Shandong, China
| | - Xuan Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Delong Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China
| | - Jian Li
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, Shandong, China
| | - Shuyuan Guo
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, Shandong, China; Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, Shandong, China
| | - Caicai Meng
- The Second Affiliated Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian 271000, Shandong, China.
| | - Yuliang Xiao
- The Second Affiliated Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian 271000, Shandong, China.
| |
Collapse
|
6
|
Guo P, Zeng M, Liu M, Zhang Y, Jia J, Zhang Z, Liang S, Zheng X, Feng W. Isolation of Calenduloside E from Achyranthes bidentata Blume and its effects on LPS/D-GalN-induced acute liver injury in mice by regulating the AMPK-SIRT3 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155353. [PMID: 38241918 DOI: 10.1016/j.phymed.2024.155353] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Acute liver injury (ALI) is a frequent fatal liver disease with a high mortality. Calenduloside E (CE) is a pentacyclic triterpenoid derived from Achyranthes bidentata Blume. It has been found that liver injury is associated with mitochondrial dysfunction, and activation of the AMPK-SIRT3 signaling pathway protects the mitochondrial function to play a role in resistance to the disease. However, whether CE is protective against ALI through the AMPK-SIRT3 signaling pathway is unclear. PURPOSE To clarify the influences of Calenduloside E (CE) isolated from Achyranthes bidentata Blume on LPS/D-GalN-induced Acute liver injury (ALI). METHODS A mouse model of ALI was developed, intraperitoneal injection of 10 μg/kg LPS and 700 mg/kg D-GalN, histopathological, oxidative stress, and immune inflammation of the mice were monitored. The mechanism of CE influencing liver injury was investigated by examining the gut microbiota, mitochondrial dysfunction, and the AMPK-SIRT3 signaling pathway. The antagonistic effects of specific AMPK and SIRT3 blocker, as well as AMPKα1, AMPKα2, SIRT3 transfection-mediated silencing were investigated to confirm the role of the AMPK-SIRT3 signaling pathway in this process. RESULTS CE relieved liver pathological damage of mice and led to reduced oxidative stress and immune inflammation in mice, affected the balance of gut microbiota in mice with liver injury, as well as energy metabolism, and regulated mRNA and protein expressions of AMPK-SIRT3 signaling pathway. In addition, in vitro studies showed that CE relieved mitochondrial respiratory and protein expressions of AMPK-SIRT3 signaling pathway in LPS/D-GalN-induced AML12 and LX2 cells, and such effect was blocked by AMPK and SIRT3 inhibitors. Furthermore, silencing of AMPKα1, AMPKα2, and SIRT3 blocked the effects of CE. Overall, the influences of CE on mice with liver injury is tuned by the AMPK-SIRT3 signaling pathway. CONCLUSION CE mediates mitochondrial function and eventually regulate energy metabolism by regulating the AMPK-SIRT3 signaling pathway. The results of this study provide molecular evidences for application of CE in treatment of ALI and provide references to the drug development for ALI.
Collapse
Affiliation(s)
- Pengli Guo
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Mengnan Zeng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Meng Liu
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Yuhan Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Jufang Jia
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Ziyu Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Shulei Liang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China
| | - Xiaoke Zheng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| | - Weisheng Feng
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou 450046, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, 156 Jinshui East Road, Zhengzhou 450046, China.
| |
Collapse
|
7
|
Zeng M, Feng A, Ren Y, Zhang Q, Wang R, Zhang B, Liu M, Guo P, Zhang Y, Lyu J, Zheng X. Salvia miltiorrhiza Bunge extract and Przewalskin ameliorate Bleomycin-induced pulmonary fibrosis by inhibition of apoptosis, oxidative stress and collagen deposition via the TGF-β1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155339. [PMID: 38237513 DOI: 10.1016/j.phymed.2024.155339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/20/2023] [Accepted: 01/05/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Salvia miltiorrhiza Bunge (Labiatae) (DS) is a key part of the traditional Chinese medicine, whose roots are used to remove blood stasis, relieve pain, eliminate carbuncle and calm the nerves. Our research team found that the DS extract could significantly reverse LPS-induced lung injury, and five new diterpenoid quinones in DS extract with excellent lung protective activity for the first time. However, the material basis and mechanism of DS on pulmonary fibrosis (PF) needs to be explored in depth. OBJECTIVE Bleomycin (BLM) was employed to establish the PF model, and Transcriptome and Surface plasmon resonance (SPR) ligand fishing technology were used to explore the material basis and mechanism of DS on PF, and provided theoretical research for clinical treatment of PF. METHODS DS extract (24.58 or 49.16 mg/kg, i.g.) was administered daily from Day 8 to Day 28, followed by intratracheal BLM drip (5 mg/kg) to induce PF. Data about the influences of DS on PF were collected by transcriptome sequencing technology. Pulmonary ultrasound, airway responsiveness, lung damage, collagen deposition, and the levels of TNF-α, IL-1β, apoptosis, oxidative stress (OS), immune cells, TGF-β1, α-SMA, E-Cadherin and Collage Ⅰ were examined. The affinity component (Przewalskin) in DS extract targeted by TGF-β1 was fished by SPR ligand fishing technology. Furthermore, an in vivo PF mouse model and an in vitro TGF-β1 induced BEAS-2B cell model were established, to explore the mechanism of Przewalskin on PF from the apoptosis, OS and epithelial mesenchymal transformation pathway. RESULTS DS extract improved pulmonary ultrasound, reduced lung damage and collagen deposition, downregulated TNF-α, IL-1β, apoptosis, OS, TGF-β1, α-SMA, E-Cadherin and Collage Ⅰ, transformed immune cells following Bleomycin challenge. Furthermore, affinity component (Przewalskin) also improved pulmonary ultrasound and airway responsiveness, reduced lung damage and collagen deposition, downregulated TNF-α, IL-1β, apoptosis, OS in vivo and in vitro. CONCLUSION Analysis using a mouse model revealed that DS extract and Przewalskin can relieve clinical symptoms of PF, reduce lung injury and improve lung function. Meanwhile, DS extract and Przewalskin can improve BLM-induced PF by inhibition of, OS, apoptosis and collagen deposition might via the TGF-β1 pathway. This study provides references to identification of novel therapeutic targets, thereby facilitating drug development for PF.
Collapse
Affiliation(s)
- Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-construction of Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Zhengzhou, China
| | - Aozi Feng
- Department of Clinical Research, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yingjie Ren
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Qinqin Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Ru Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Pengli Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yuhan Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Jun Lyu
- Department of Clinical Research, the First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-construction of Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Zhengzhou, China.
| |
Collapse
|
8
|
Liu W, Li Y, Zhao T, Gong M, Wang X, Zhang Y, Xu L, Li W, Li Y, Jia J. The role of N-methyl-D-aspartate glutamate receptors in Alzheimer's disease: From pathophysiology to therapeutic approaches. Prog Neurobiol 2023; 231:102534. [PMID: 37783430 DOI: 10.1016/j.pneurobio.2023.102534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
N-Methyl-D-aspartate glutamate receptors (NMDARs) are involved in multiple physiopathological processes, including synaptic plasticity, neuronal network activities, excitotoxic events, and cognitive impairment. Abnormalities in NMDARs can initiate a cascade of pathological events, notably in Alzheimer's disease (AD) and even other neuropsychiatric disorders. The subunit composition of NMDARs is plastic, giving rise to a diverse array of receptor subtypes. While they are primarily found in neurons, NMDAR complexes, comprising both traditional and atypical subunits, are also present in non-neuronal cells, influencing the functions of various peripheral tissues. Furthermore, protein-protein interactions within NMDAR complexes has been linked with Aβ accumulation, tau phosphorylation, neuroinflammation, and mitochondrial dysfunction, all of which potentially served as an obligatory relay of cognitive impairment. Nonetheless, the precise mechanistic link remains to be fully elucidated. In this review, we provided an in-depth analysis of the structure and function of NMDAR, investigated their interactions with various pathogenic proteins, discussed the current landscape of NMDAR-based therapeutics, and highlighted the remaining challenges during drug development.
Collapse
Affiliation(s)
- Wenying Liu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Tan Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Min Gong
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Xuechu Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Yue Zhang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China
| | - Lingzhi Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, PR China; Beijing Key Laboratory of Geriatric Cognitive Disorders, PR China; Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, PR China; Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, PR China; Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, PR China.
| |
Collapse
|
9
|
Cao B, Zeng MN, Hao FX, Hao ZY, Zhang ZK, Liang XW, Wu YY, Zhang YH, Feng WS, Zheng XK. P-coumaric acid ameliorates Aβ 25-35-induced brain damage in mice by modulating gut microbiota and serum metabolites. Biomed Pharmacother 2023; 168:115825. [PMID: 37924791 DOI: 10.1016/j.biopha.2023.115825] [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: 08/02/2023] [Revised: 10/19/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease for which there is a lack of effective therapeutic drugs. There is great potential for natural products to be used in the development of anti-AD drugs. P-coumaric acid (PCA), a small molecule phenolic acid widely distributed in the plant kingdom, has pharmacological effects such as neuroprotection, but its anti-AD mechanism has not been fully elucidated. In the current study, we investigated the mechanism of PCA intervention in the Aβ25-35-induced AD model using gut microbiomics and serum metabolomics combined with in vitro and in vivo pharmacological experiments. PCA was found to ameliorate cognitive dysfunction and neuronal cell damage in Aβ25-35-injected mice as measured by behavioral, pathological and biochemical indicators. 16S rDNA sequencing and serum metabolomics showed that PCA reduced the abundance of pro-inflammatory-associated microbiota (morganella, holdemanella, fusicatenibacter and serratia) in the gut, which were closely associated with metabolites of the glucose metabolism, arachidonic acid metabolism, tyrosine metabolism and phospholipid metabolism pathways in serum. Next, in vivo and in vitro pharmacological investigations revealed that PCA regulated Aβ25-35-induced disruption of glucose metabolism through activation of PI3K/AKT/Glut1 signaling. Additionally, PCA ameliorated Aβ25-35-induced neuroinflammation by inhibiting nuclear translocation of NF-κB and by modulating upstream MAPK signaling. In conclusion, PCA ameliorated cognitive deficits in Aβ25-35-induced AD mice by regulating glucose metabolism and neuroinflammation, and the mechanism is related not only to restoring homeostasis of gut microbiota and serum metabolites, but also to PI3K/AKT/Glut1 and MAPK/NF-κB signaling.
Collapse
Affiliation(s)
- Bing Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Meng-Nan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Feng-Xiao Hao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Zhi-You Hao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Zhen-Kai Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Xi-Wen Liang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yuan-Yuan Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yu-Han Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Wei-Sheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, China.
| | - Xiao-Ke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China; The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, China.
| |
Collapse
|
10
|
Zeng M, Feng A, Wang L, Li K, Zhou J. Aralia saponin A isolated from Achyranthes bidentata Bl. ameliorates LPS/D-GalN induced acute liver injury via SPHK1/S1P/S1PR1 pathway in vivo and in vitro. Int Immunopharmacol 2023; 124:110912. [PMID: 37699301 DOI: 10.1016/j.intimp.2023.110912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVE Acute liver injury (ALI) refers to a disease in which the liver is affected by factors such as chemical substances, alcohol, and virus infection in a short time, resulting in damage to liver cells. Achyranthes bidentata Bl. with the hepatoprotective activity has attracted great attention. In this study, a pentacyclic triterpenoid (Aralia saponin A, AsA) was isolated from roots of Achyranthes bidentata Bl. and its anti-ALI activity, as well as the mechanisms, were investigated for the first time. METHODS AsA (10 or 20 mg/kg, i.g.) was administered over a period of 1 weeks, following which liver injury was induced by LPS (10 µg/kg)/D-GalN (700 mg/kg). H&E staining of liver, Aspartate amino transferase (AST), Alanine transaminase (ALT) and cytokines was employed to investigate ALI relevant features. The mitochondrial morphology and levels of mitochondrial membrane potential (MMP), oxidative stress balance, apoptosis, average fluorescence intensity of 2-DG, natural killer (NK) cells in liver tissues were determined to assess the oxidative stress damage and inflammatory injury. Transcriptomics and metabonomics analysis were employed to clarify the mechanisms. Additionally, the mRNA and protein expression levels of Sphingosine 1-phosphate (S1P), Sphingosine kinase-1 (SPKH1), Sphingosine 1 phosphate receptor 1 (S1PR1), Sphingosine 1 phosphate receptor 3 (S1PR3), TNF receptor associated factor 2 (TRAF-2), Phospho-NF- kappaB p65 (p-P65), NF- kappaB p65 (P65), Proto-oncogene ras (Ras), Ras-related C3 botulinum toxin substrate (Rac), Phospholipase C (PLC), Interleukin 6 (IL-6), Tumor necrosis factor α (TNF-α), Interleukin 1β (IL-1β), Vascular cell adhesion molecule 1 (Vcam1), CC chemokine ligand-2 (Ccl2) were analyzed. The mediating role of SPHK1 in the observed effects caused by AsA was assessed by investigatin SPHK1 transfection silencing/overexpression against AsA in AML12 cells induced by LPS/D-GalN. RESULTS AsA can ameliorate liver function, inflammation, mitochondrial structure and oxidative stress in the ALI model. Meanwhile, AsA led to downregulated expression of proteins associated with sphingolipid signaling pathway. Silencing of SPHK1 led to enhanced protective effects of AsA, while over-expression of SPHK1 led to degraded protective effects of AsA in LPS/D-GalN-induced AML12 cells, suggesting that ALI is regulated by active molecules of AsA by means of SPHK1 mediation. CONCLUSIONS AsA can ameliorate LPS/D-GalN-induced ALI by inhibiting inflammation and oxidative stress via the SPHK1/S1P/S1PR1 pathway. In this way, a molecular justification is provided for AsA application in ALI treatment.
Collapse
Affiliation(s)
- Mengnan Zeng
- College of Pharmacy, Huanghe S&T University, Zhengzhou 450000, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Aozi Feng
- Department of Clinical Research, Jinan University, Guangzhou 510632, China
| | - Li Wang
- College of Pharmacy, Huanghe S&T University, Zhengzhou 450000, China.
| | - Kun Li
- College of Pharmacy, Huanghe S&T University, Zhengzhou 450000, China
| | - Jihong Zhou
- College of Pharmacy, Huanghe S&T University, Zhengzhou 450000, China
| |
Collapse
|
11
|
Two polyphenols isolated from Corallodiscus flabellata B. L. Burtt ameliorate amyloid β-protein induced Alzheimer's disease neuronal injury by improving mitochondrial homeostasis. Behav Brain Res 2023; 440:114264. [PMID: 36535434 DOI: 10.1016/j.bbr.2022.114264] [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: 07/24/2022] [Revised: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Corallodiscus flabellata B. L. Burtt (CF) is a Chinese folk herb with reported potential for the treatment of Alzheimer's disease (AD). 3,4-Dihydroxyphenylethanol-8-O-[4-O-trans-caffeoyl-β-D-apiofuranosyl-(1→3)-β-D-glucopyranosyl (1→6)][1]-β-D-glucopyranoside (SDC-1-8) and hydroxytyrosol (HT) are two polyphenolic compounds isolated from CF. The aim of this study was to investigate the protective effects of SDC-1-8 and HT on an Aβ25-35-induced AD model and to study the underlying mechanism. The AD mouse model was established using a brain injection of amyloid β-protein 25-35 (Aβ25-35, 200 μM), followed by continuous administration of SDC-1-8 and HT for 4 weeks, and found that they improved cognitive dysfunction; ameliorated neuronal damage and apoptosis; decreased oxidative stress, and mitochondrial fission protein levels; and increased mitochondrial fusion protein levels in AD mice. Moreover, SDC-1-8 and HT inhibited mitochondrial membrane depolarization, reduced intracellular stored Ca2+ levels, enhanced mitochondrial respiration, increased mitochondrial fusion, and decreased mitochondrial division in Aβ25-35-induced PC12 cells even in the presence of mdivi-1. Furthermore, molecular docking simulations showed that SDC-1-8 and HT interacted with dynamin-related protein 1 with higher affinity than mitofusin 1. Thus, it is summarized that SDC-1-8 and HT may have neuroprotective effects by balancing the abnormalities of mitochondrial fission and fusion, and SDC-1-8 and HT are the components providing the therapeutic basis of CF.
Collapse
|
12
|
Two new secondary metabolites from Oreocharis auricula and their chemotaxonomic significance. BIOCHEM SYST ECOL 2022. [DOI: 10.1016/j.bse.2022.104477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|