1
|
Wang S, Zhou Y, Wu Y, Lang Y, Mao Y, Pan G, Gao Z. Long-term aerobic exercise improves learning memory capacity and effects on oxidative stress levels and Keap1/Nrf2/GPX4 pathway in the hippocampus of APP/PS1 mice. Front Neurosci 2024; 18:1505650. [PMID: 39659881 PMCID: PMC11628528 DOI: 10.3389/fnins.2024.1505650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Objective To examine the effects of long-term aerobic exercise on oxidative stress and learning memory ability of APP/PS1 mice, focusing on the hippocampal Keap1, Nrf2, HO-1, and GPX4 proteins. Methods Thirty APP/PS1 double transgenic AD mice were randomly divided into three groups: model group, short-term exercise group, and long-term exercise group, with 10 mice in each group. Male non-transgenic mice of the same age served as the control group. The groups underwent swimming training for 6 weeks and 12 weeks, respectively. After the intervention, cognitive abilities were assessed using the Morris water maze test. Hippocampal tissue samples were analyzed for changes in superoxide dismutase (SOD) activity and malondialdehyde (MDA) content. ROS expression was observed using dihydroethidium probe, and Keap1, Nrf2, HO-1, and GPX4 protein levels were detected by Western blot analysis. Results Aerobic exercise significantly reduced the escape latency and increased both the time spent in the target quadrant and the number crossing the platform compared to the model group (p < 0.05). In the hippocampus, aerobic exercise significantly reduced the MDA content, while significantly increased SOD activity (p < 0.05). The level of ROS in the hippocampal region was significantly reduced by aerobic exercise (p < 0.05), with decreased Keap1 protein expression of and increased Nrf2, HO-1, GPX4 protein expression (p < 0.05). Conclusion Aerobic exercise enhances memory and learning abilities, improves cognitive function, and reduces the oxidative stress levels in the hippocampus of AD mice, which involves in the activation of Keap1/Nrf2/GPX4 pathway.
Collapse
Affiliation(s)
- Shiyan Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Ye Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yucheng Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yali Lang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yajun Mao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Guoyuan Pan
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Zhenzhen Gao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
2
|
Yang L, Nao J. Ferroptosis: a potential therapeutic target for Alzheimer's disease. Rev Neurosci 2022:revneuro-2022-0121. [PMID: 36514247 DOI: 10.1515/revneuro-2022-0121] [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: 10/03/2022] [Accepted: 10/30/2022] [Indexed: 12/15/2022]
Abstract
The most prevalent dementia-causing neurodegenerative condition is Alzheimer's disease (AD). The aberrant buildup of amyloid β and tau hyperphosphorylation are the two most well-known theories about the mechanisms underlying AD development. However, a significant number of pharmacological clinical studies conducted around the world based on the two aforementioned theories have not shown promising outcomes, and AD is still not effectively treated. Ferroptosis, a non-apoptotic programmed cell death defined by the buildup of deadly amounts of iron-dependent lipid peroxides, has received more attention in recent years. A wealth of data is emerging to support the role of iron in the pathophysiology of AD. Cell line and animal studies applying ferroptosis modulators to the treatment of AD have shown encouraging results. Based on these studies, we describe in this review the underlying mechanisms of ferroptosis; the role that ferroptosis plays in AD pathology; and summarise some of the research advances in the treatment of AD with ferroptosis modulators. We hope to contribute to the clinical management of AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
3
|
Ferrari F, Moretti A, Villa RF. Incretin-based drugs as potential therapy for neurodegenerative diseases: current status and perspectives. Pharmacol Ther 2022; 239:108277. [DOI: 10.1016/j.pharmthera.2022.108277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
|
4
|
Mechanism of Zhinao Capsule in Treating Alzheimer’s Disease Based on Network Pharmacology Analysis and Molecular Docking Validation. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:5708769. [PMID: 36032542 PMCID: PMC9410932 DOI: 10.1155/2022/5708769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/02/2022]
Abstract
Objective This study aimed to determine the active components of Zhinao capsule (ZNC) and the targets in treating Alzheimer's disease (AD) so as to investigate and explore the mechanism of ZNC for AD. Methods The active components and targets of ZNC were determined from the traditional Chinese medicine systems pharmacology database (TCMSP). The target genes of AD were searched for in GeneCards. Cytoscape was used to construct an herb-component-target-disease network. A protein-protein interaction (PPI) network was constructed by STRING. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the OmicShare. UCSF Chimera and SwissDock were used for molecular docking verification. Finally, four key target genes were validated by Western blotting. Results In total, 55 active components, 287 targets of active components, 1197 disease genes, and 134 common genes were screened, which were significantly enriched in 3975 terms of biological processes (BP), 284 terms of cellular components (CC), 433 terms of molecular functions (MF), and 245 signaling pathways. Caspase-3 (CASP3) and beta-sitosterol, tumor necrosis factor-alpha (TNF-α) and quercetin, vascular endothelial growth factor A (VEGFA) and baicalein, and mitogen-activated protein kinase 1 (MAPK1) and quercetin showed good-to-better docking. Moreover, ZNC not only downregulated CASP3 and TNF-α protein expression but also upregulated the protein expression of VEGFA and MAPK1. Conclusions The active components of ZNC, such as beta-sitosterol, quercetin, and baicalein may act on multiple targets like CASP3, VEGFA, MAPK1, and TNF-α to affect T cell receptor (TCR), TNF, and MAPK signaling pathway, thereby achieving the treatment of AD. This study provides a scientific basis for further exploring the potential mechanism of ZNC in the treatment of AD and a reference for its clinical application.
Collapse
|
5
|
The Role of Mitochondrial Quality Control in Cognitive Dysfunction in Diabetes. Neurochem Res 2022; 47:2158-2172. [PMID: 35661963 PMCID: PMC9352619 DOI: 10.1007/s11064-022-03631-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 12/26/2022]
Abstract
Type 2 diabetes (T2DM) is a well known risk factor for Alzheimer’s disease. Mitochondria are the center of intracellular energy metabolism and the main source of reactive oxygen species. Mitochondrial dysfunction has been identified as a key factor in diabetes-associated brain alterations contributing to neurodegenerative events. Defective insulin signaling may act in concert with neurodegenerative mechanisms leading to abnormalities in mitochondrial structure and function. Mitochondrial dysfunction triggers neuronal energy exhaustion and oxidative stress, leading to brain neuronal damage and cognitive impairment. The normality of mitochondrial function is basically maintained by mitochondrial quality control mechanisms. In T2DM, defects in the mitochondrial quality control pathway in the brain have been found to lead to mitochondrial dysfunction and cognitive impairment. Here, we discuss the association of mitochondrial dysfunction with T2DM and cognitive impairment. We also review the molecular mechanisms of mitochondrial quality control and impacts of mitochondrial quality control on the progression of cognitive impairment in T2DM.
Collapse
|
6
|
Jaragh-Alhadad LA, Falahati M. Copper oxide nanoparticles promote amyloid-β-triggered neurotoxicity through formation of oligomeric species as a prelude to Alzheimer's diseases. Int J Biol Macromol 2022; 207:121-129. [PMID: 35259430 DOI: 10.1016/j.ijbiomac.2022.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/12/2022] [Accepted: 03/02/2022] [Indexed: 12/28/2022]
Abstract
Protein oligomerization is involved in the progression of Alzheimer's disease (AD). In general, a particle that can accelerate protein oligomerization should be considered a toxic material. Several studies reported the progress of nanoparticles (NPs) such as copper oxide (CuO) in biomedical platforms, however, they may have the ability to promote the protein oligomerization process. Here, we aimed to study the effect of CuO NPs on amyloid β1-42 (Aβ1-42) oligomerization and relevant neurotoxicity. CuO NPs were synthesized by precipitation technique and characterized by several methods such as ThT, Congo red, CD spectroscopic methods, and TEM imaging. The outcomes indicated that the fabricated CuO NPs with a size of around 50 nm led to a remarkable acceleration in Aβ1-42 oligomerization in a concentration-dependent manner through shortening the nucleation step and promoting the fibrillization rate. Moreover, cellular assays revealed that Aβ1-42 oligomers aged with CuO NPs were more toxic than Aβ1-42 oligomers untreated against SH-SY5Y cells in triggering cell mortality, membrane leakage, oxidative stress, and apoptosis. In conclusion, this study provides important information about the adverse effects of CuO NPs against proteins in the central nervous system to promote the formation of cytotoxic oligomers.
Collapse
Affiliation(s)
- Laila Abdulmohsen Jaragh-Alhadad
- Department of Chemistry, College of Science, Kuwait University, Safat 13060, Kuwait; Cardiovascular and Metabolic Sciences Department, Learner Research Institute, Cleveland Clinic, OH 44195, USA.
| | - Mojtaba Falahati
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD Rotterdam, the Netherlands; Department of Nanotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Yun-Liang X, Bo Z. Protective Effect of Patchouli Alcohol Against SH-SY5Y Cell Injury Induced by Aβ 25-35 via the Reduction of Oxidative Stress and Apoptosis. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211031715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Patchouli alcohol (PA) has multiple pharmacological activities, but its protective effect against SH-SY5Y cell injury induced by Aβ25-35 has not been reported. It has been recorded that phosphatidylinositol 3-hydroxykinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway plays an important role in neuroprotection. The purpose of this study was to investigate the protective effect of PA against SH-SY5Y cell injury induced by Aβ25-35 and its underlying mechanism. The results showed that compared with that in the Aβ25-35-induced injury group, the survival rate of SH-SY5Y cells increased ( P < .01) in the different PA-treated groups and the lactic dehydrogenase activity decreased significantly ( P < .01) in the 10, 20, and 40 μg/mL PA groups; compared with those in the Aβ25-35-induced injury group, the malonyldialdehyde contents in SH-SY5Y cells decreased ( P < .05 or P < .01), while the superoxide dismutase, glutathione peroxidase, and catalase activities increased significantly ( P < .05 or P < .01) in the different PA-treated groups; compared with those in the Aβ25-35-induced injury group, the apoptosis rates, and the mRNA and protein levels of Caspase-3 and Bax in SH-SY5Y cells decreased ( P < .05 or P < .01), while the mRNA and protein levels of Bcl-2, and phosphorylated Akt (p-Akt) and phosphorylated mTOR protein levels increased significantly ( P < .05 or P < .01) in the different PA-treated groups. The above results indicate that PA can inhibit the oxidative stress and apoptosis of SH-SY5Y cells induced by Aβ25-35 by regulating the PI3K/Akt/mTOR pathway, to protect the SH-SY5Y cells from the injury induced by Aβ25-35.
Collapse
Affiliation(s)
- Xie Yun-Liang
- People’s Hospital of Suzhou New District, Suzhou, P. R. China
| | - Zhang Bo
- Affiliated Hospital of Beihua University, Jilin City, P. R. China
| |
Collapse
|
8
|
Li C, Zhang Y, Liu R, Mai Y. Anagliptin Protected against Hypoxia/Reperfusion-Induced Brain Vascular Endothelial Permeability by Increasing ZO-1. ACS OMEGA 2021; 6:7771-7777. [PMID: 33778288 PMCID: PMC7992143 DOI: 10.1021/acsomega.1c00242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 05/17/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral ischemia-reperfusion injury is commonly induced during the treatment of ischemic stroke and is reported to be related to the blood-brain barrier destruction and brain vascular endothelial cell dysfunction. Anagliptin is a novel antidiabetic agent recently reported to protect neurons from oxidative stress. In the present study, we aim to investigate the protective property of anagliptin against oxygen-glucose deprivation and reperfusion (OGD/R)-induced injury on endothelial cells and clarify the potential underlying mechanism. METHODS OGD/R modeling was established on bEnd.3 brain endothelial cells. Cell viability was detected using the MTT assay, and the mitochondrial reactive oxygen species (ROS) level was measured using the mitoses red staining assay. The endothelial monolayer permeability was determined using an FITC-dextran permeation assay. The expression levels of NOX-4 and ZO-1 were evaluated using qRT-PCR and Western blot assays. The expressions of MLC-2, p-MLC-2, and myosin light chain kinase (MLCK) were determined using Western blot. RESULTS First, the decreased cell viability, upregulated NOX-4, and elevated mitochondrial ROS level in the endothelial cells induced by OGD/R were reversed by treatment with anagliptin. Second, the enlarged endothelial permeability and the decreased expression level of ZO-1 in the endothelial cells induced by OGD/R were alleviated by anagliptin. Third, the downregulation of ZO-1 and enlarged brain endothelial monolayer permeability induced by OGD/R were ameliorated by an MLCK inhibitor, ML-7. Lastly, the elevated expressions of MLCK and p-MLC-2 induced by OGD/R were suppressed by anagliptin. CONCLUSION Anagliptin protected against hypoxia/reperfusion-induced brain vascular endothelial permeability by increasing the expression ZO-1, mediated by inhibition of the MLCK/MLC-2 signaling pathway.
Collapse
Affiliation(s)
- Chuo Li
- Department
of Neurology, Guangzhou Eighth People’s
Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510440, China
- . Phone/Fax: +86-020-36473145
| | - Yusheng Zhang
- Department
of Neurology and Stroke Center, The First
Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Rongrong Liu
- Department
of Neurology and Stroke Center, The First
Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Yuzhen Mai
- Department
of Neurology, Guangzhou Eighth People’s
Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510440, China
| |
Collapse
|
9
|
Studies of anticancer activity in vivo and in vitro behaviors of liposomes encapsulated iridium(III) complex. J Biol Inorg Chem 2021; 26:109-122. [PMID: 33475857 DOI: 10.1007/s00775-020-01841-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
Iridium(III) complexes have gained great attention in cancer treatment in recent years. In this paper, we designed and synthesized a new iridium(III) complex [Ir(piq)2(DQTT)](PF6) Ir1 (piq = 1-phenylisoquinoline, DQTT = 12-(1,4-dihydroquinoxalin-6-yl)-4,5,9,14-tetraazabenzo[b]triphenylene). The Ir1-loaded PEGylated liposomes (Lipo-Ir1) were prepared using the ethanol injection method. The anticancer activity of the complex and Lipo-Ir1 against SGC-7901 (human gastric adenocarcinoma), A549 (human lung carcinoma), HeLa (human cervical carcinoma), HepG2 (human hepatocellular carcinoma), BEL-7402 (human hepatocellular carcinoma), and normal NIH3T3 (mouse embryonic fibroblasts) was tested by the MTT method. The complex Ir1 shows moderate or low cytotoxicity against the selected cancer cells, whereas the Lipo-Ir1 exhibits high anticancer activity toward the same cancer cells. The apoptosis induced by Lipo-Ir1 was assayed by flow cytometry and Lipo-Ir1 induced apoptosis through increasing intracellular reactive-oxygen species levels, decreasing mitochondrial membrane potential, further promoting cytochrome c release and causing the increase of level of intracellular Ca2+. Western blot was used to detect the changes in Bcl-2 family protein and PI3K/AKT pathway proteins. The cloning experiments demonstrated that the Lipo-Ir1 can effectively inhibit cell proliferation. In vivo experiments, Lipo-Ir1 inhibited tumor growth in xenograft nude mice, and the percentage of tumor growth inhibition in vivo was 75.70%. Overall, the liposomes Lipo-Ir1 exhibits higher anticancer activity than Ir1 under the same conditions. These results indicated that Lipo-Ir1 may be a valuable resource for cancer therapy.
Collapse
|
10
|
Londero ÉP, Bressan CA, Pês TS, Saccol EMH, Baldisserotto B, Finamor IA, Pavanato MA. Rutin-added diet protects silver catfish liver against oxytetracycline-induced oxidative stress and apoptosis. Comp Biochem Physiol C Toxicol Pharmacol 2021; 239:108848. [PMID: 32777463 DOI: 10.1016/j.cbpc.2020.108848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
It is unknown whether the flavonoid rutin can protect the silver catfish liver in response to exposure to a known stressor, such as the prophylactic usage of the antimicrobial agent oxytetracycline. Thus, the current study aimed to assess the effect of rutin incorporation into the silver catfish diet formulation on oxytetracycline-induced liver oxidative stress and apoptosis. Fish were split into four groups as follows: control, rutin (1.5 g kg diet-1), oxytetracycline (0.1 g kg diet-1) and rutin+oxytetracycline (1.5 g kg diet-1 and 0.1 g kg diet-1, respectively). After two weeks of feeding with the different diets (standard, rutin-, oxytetracycline and rutin+oxytetracycline-added diets), fish were euthanized to collect the liver. Although the rutin-added diet was unable to recover glutathione peroxidase activity, ascorbic acid and reduced glutathione (GSH) levels, which were depleted due to oxytetracycline consumption, it markedly diminished the oxidized glutathione (GSSG) content, thus decreasing the GSSG to GSH ratio, an important index of oxidative stress. It also increased glutathione reductase and markedly augmented glucose-6-phosphate dehydrogenase activities, which were declined after oxytetracycline ingestion. Furthermore, the rutin-added diet reestablished superoxide dismutase and catalase activities and reduced lipid peroxidation, nitric oxide and superoxide anion levels as well, all changes resulting from oxytetracycline consumption. Finally, it also prevented oxytetracycline-induced apoptosis through increasing heat shock protein 70 and markedly decreasing high mobility group box 1 and, consequently, reducing cleaved caspase-3 protein levels. Therefore, in conclusion, the incorporation of this flavonoid to the silver catfish diet protected the liver against oxytetracycline-induced liver oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Érika P Londero
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil
| | - Caroline A Bressan
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil
| | - Tanise S Pês
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil
| | - Etiane M H Saccol
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil
| | | | - Isabela A Finamor
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil.
| | - Maria A Pavanato
- Department of Physiology and Pharmacology, Universidade Federal de Santa Maria, Brazil.
| |
Collapse
|
11
|
Yin Z, Wang X, Zheng S, Cao P, Chen Y, Yu M, Liao C, Zhang Z, Han J, Duan Y, Yang X, Zhang S. LongShengZhi Capsule Attenuates Alzheimer-Like Pathology in APP/PS1 Double Transgenic Mice by Reducing Neuronal Oxidative Stress and Inflammation. Front Aging Neurosci 2020; 12:582455. [PMID: 33328962 PMCID: PMC7719723 DOI: 10.3389/fnagi.2020.582455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 10/16/2020] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia in the elderly. It may be caused by oxidative stress, inflammation, and cerebrovascular dysfunctions in the brain. LongShengZhi Capsule (LSZ), a traditional Chinese medicine, has been approved by the China Food and Drug Administration for treatment of patients with cardiovascular/cerebrovascular disease. LSZ contains several neuroprotective ingredients, including Hirudo, Astmgali Radix, Carthami Flos (Honghua), Persicae Semen (Taoren), Acori Tatarinowii Rhizoma (Shichangpu), and Acanthopanax Senticosus (Ciwujia). In this study, we aimed to determine the effect of LSZ on the AD process. Double transgenic mice expressing the amyloid-β precursor protein and mutant human presenilin 1 (APP/PS1) to model AD were treated with LSZ for 7 months starting at 2 months of age. LSZ significantly improved the cognition of the mice without adverse effects, indicating its high degree of safety and efficacy after a long-term treatment. LSZ reduced AD biomarker Aβ plaque accumulation by inhibiting β-secretase and γ-secretase gene expression. LSZ also reduced p-Tau expression, cell death, and inflammation in the brain. Consistently, in vitro, LSZ ethanol extract enhanced neuronal viability by reducing L-glutamic acid-induced oxidative stress and inflammation in HT-22 cells. LSZ exerted antioxidative effects by enhancing superoxide dismutase and glutathione peroxidase expression, reduced Aβ accumulation by inhibiting β-secretase and γ-secretase mRNA expression, and decreased p-Tau level by inhibiting NF-κB-mediated inflammation. It also demonstrated neuroprotective effects by regulating the Fas cell surface death receptor/B-cell lymphoma 2/p53 pathway. Taken together, our study demonstrates the antioxidative stress, anti-inflammatory, and neuroprotective effects of LSZ in the AD-like pathological process and suggests it could be a potential medicine for AD treatment.
Collapse
Affiliation(s)
- Zequn Yin
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xuerui Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shihong Zheng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Peichang Cao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Maoyun Yu
- School of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | | | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China.,College of Life Sciences, Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Yajun Duan
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| |
Collapse
|
12
|
Jin Y, Zhao H, Hou Y, Song G. The effects of dipeptidyl peptidase-4 inhibitors and glucagon-like peptide 1 receptor agonists on cognitive functions in adults with type 2 diabetes mellitus: a systematic review and meta-analysis. Acta Diabetol 2020; 57:1129-1144. [PMID: 32300876 DOI: 10.1007/s00592-020-01529-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022]
Abstract
AIMS The effects of dipeptidyl peptidase-4 inhibitors (DPP-4 inhibitors/DPP-4I) and glucagon-like peptide 1 receptor agonists (GLP-1 RA) on cognition in patients with type 2 diabetes mellitus (T2DM) remain controversial. We aimed to explore this clinical issue through a systematic review and meta-analysis. METHODS PubMed, EMBASE and the Cochrane Library were searched, and data were expressed as mean difference (MD) or hazard ratio (HR)/odds ratio (OR) with a 95% confidence interval (CI). Heterogeneity was assessed using the Chi-squared test and the I2 statistic. The study was registered with PROSPERO (ID: CRD42019138777). RESULTS Eleven studies (n = 304,258 T2DM patients) were included in our review. In the DPP-4I group, six studies were enrolled to estimate ΔMini-Mental State Examination (MMSE) scores from baseline to the final evaluations after DPP-4I treatment, which showed no statistical difference (MD 0.20; 95% CI - 0.75 to 1.15, p = 0.68). ΔMMSE scores in the DPP-4I group and the other antidiabetic groups were compared, revealing no statistical difference (MD 0.57; 95% CI - 0.05 to 1.19, p = 0.07). Two cohort studies were pooled to determine the HRs for dementia, showing a lower risk of dementia after DPP-4I treatment (HR 0.52; 95% CI 0.29-0.93, p = 0.03). In the GLP-1 analogs group, two studies were included, one of which revealed a downward trend in the risk of dementia after GLP-1 analog treatment, while the other revealed no significant difference after incretins treatment. CONCLUSIONS Currently there is not enough irrefutable evidence to support the hypothesis of positive effects of incretins on cognition. Further clinical studies need to be performed.
Collapse
Affiliation(s)
- Yuxin Jin
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Hang Zhao
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Yilin Hou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China.
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, 050051, Hebei, People's Republic of China.
| |
Collapse
|
13
|
Hou L, Zhang L, Hong JS, Zhang D, Zhao J, Wang Q. Nicotinamide Adenine Dinucleotide Phosphate Oxidase and Neurodegenerative Diseases: Mechanisms and Therapy. Antioxid Redox Signal 2020; 33:374-393. [PMID: 31968994 DOI: 10.1089/ars.2019.8014] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: The growing incidence of neurodegenerative diseases significantly impacts the individuals who suffer from these disorders and is a major health concern globally. Although the specific mechanisms of neurodegenerative diseases are still far from being acknowledged, it is becoming clear that oxidative stress and neuroinflammation are critical contributing factors to the progression of neurodegeneration. Thus, it is conceivable that the inhibition of oxidative stress and neuroinflammation may represent promising therapeutic targets for the treatment of neurodegenerative diseases. Recent Advances: Recently, the strategy for neurodegenerative disease therapy has shifted from the use of antioxidants and conventional anti-inflammatory targets to upstream mediators due to the failure of most antioxidants and nonsteroidal anti-inflammatory drugs in clinical trials. Nicotinamide adenine dinucleotide phosphate oxidases (NOXs), a family of superoxide-producing enzyme complexes, have been identified as an upstream factor that controls both oxidative stress and neuroinflammation. Genetic inactivation or pharmacological inhibition of NOX enzymes displays potent neuroprotective effects in a broad spectrum of neurodegenerative disease models. Critical Issues: The detailed mechanisms of how NOX enzymes regulate oxidative stress and neuroinflammation still remain unclear. Moreover, the currently available inhibitors of NOX enzymes exhibit nonspecificity, off-target effects, unsuitable pharmacokinetic properties, and even high toxicity, markedly limiting their potential clinical applications. Future Directions: This review provides novel insights into the roles of NOXs in neurodegenerative pharmacology, and indicates the types of NOX enzyme inhibitors that should be identified and developed as candidates for future applications, which might reveal novel neurodegenerative disease therapies based on NOXs.
Collapse
Affiliation(s)
- Liyan Hou
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Dan Zhang
- State Key Laboratory of Natural Products and Functions, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| | - Qingshan Wang
- Institute of Toxicology, School of Public Health, Dalian Medical University, Dalian, China.,National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Can dipeptidyl peptidase-4 inhibitors treat cognitive disorders? Pharmacol Ther 2020; 212:107559. [PMID: 32380197 DOI: 10.1016/j.pharmthera.2020.107559] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022]
Abstract
The linkage of neurodegenerative diseases with insulin resistance (IR) and type 2 diabetes mellitus (T2DM), including oxidative stress, mitochondrial dysfunction, excessive inflammatory responses and abnormal protein processing, and the correlation between cerebrovascular diseases and hyperglycemia has opened a new window for novel therapeutics for these cognitive disorders. Various antidiabetic agents have been studied for their potential treatment of cognitive disorders, among which the dipeptidyl peptidase-4 (DPP-4) inhibitors have been investigated more recently. So far, DPP-4 inhibitors have demonstrated neuroprotection and cognitive improvements in animal models, and cognitive benefits in diabetic patients with or without cognitive impairments. This review aims to summarize the potential mechanisms, advantages and limitations, and currently available evidence for developing DPP-4 inhibitors as a treatment of cognitive disorders.
Collapse
|