1
|
Li S, Hou Z, Ye T, Song X, Hu X, Chen J. Saponin components in Polygala tenuifolia as potential candidate drugs for treating dementia. Front Pharmacol 2024; 15:1431894. [PMID: 39050746 PMCID: PMC11266144 DOI: 10.3389/fphar.2024.1431894] [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: 05/13/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Objective This study aims to elucidate the intervention effects of saponin components from Polygala tenuifolia Willd (Polygalaceae) on dementia, providing experimental evidence and new insights for the research and application of saponins in the field of dementia. Materials and Methods This review is based on a search of the PubMed, NCBI, and Google Scholar databases from their inception to 13 May 2024, using terms such as "P. tenuifolia," "P. tenuifolia and saponins," "toxicity," "dementia," "Alzheimer's disease," "Parkinson's disease dementia," and "vascular dementia." The article summarizes the saponin components of P. tenuifolia, including tenuigenin, tenuifolin, polygalasaponins XXXII, and onjisaponin B, as well as the pathophysiological mechanisms of dementia. Importantly, it highlights the potential mechanisms by which the active components of P. tenuifolia prevent and treat diseases and relevant clinical studies. Results The saponin components of P. tenuifolia can reduce β-amyloid accumulation, exhibit antioxidant effects, regulate neurotransmitters, improve synaptic function, possess anti-inflammatory properties, inhibit neuronal apoptosis, and modulate autophagy. Therefore, P. tenuifolia may play a role in the prevention and treatment of dementia. Conclusion The saponin components of P. tenuifolia have shown certain therapeutic effects on dementia. They can prevent and treat dementia through various mechanisms.
Collapse
Affiliation(s)
- Songzhe Li
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhitao Hou
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ting Ye
- The Second Hospital Affiliated Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xiaochen Song
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinying Hu
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Chen
- College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
2
|
Lee B, Yu MS, Song JG, Lee HM, Kim HW, Na D. Corydalis ternata Nakai Alleviates Cognitive Decline in Alzheimer's Disease by Reducing β-Amyloid and Neuroinflammation. Rejuvenation Res 2024; 27:87-101. [PMID: 38545769 DOI: 10.1089/rej.2023.0069] [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] [Indexed: 04/25/2024] Open
Abstract
Recently, natural herbs have gained increasing attention owing to their comparatively low toxicity levels and the abundance of historical medical documentation regarding their use. Nevertheless, owing to a lack of knowledge regarding these herbs and their compounds, attempts to find those that could be beneficial for treating diseases have often been ad hoc; thus, there is now a growing demand for an in silico method to identify beneficial herbs. In this study, we present a computational approach for identifying natural herbs specifically effective in treating cognitive decline in Alzheimer's disease (AD) sufferers, which analyzes the similarities between herbal compounds and known drugs targeting AD-related proteins. Our in silico method suggests that Corydalis ternata can improve cognitive decline in AD sufferers. Behavioral tests with an AD mouse model for the confirmation of the in silico prediction reveals that C. ternata significantly alleviated the cognitive decline (memory and motor functions) caused by neurodegeneration. Further pathology analyses reveal that C. ternata decreases the level of Aβ plaques, reduces neuroinflammation, and promotes autophagy flux, and thus C. ternata can be clinically effective for preventing mild cognitive impairment during the early stages of AD. These findings highlight the potential utility of our in silico method and the potential clinical application of the identified natural herb in treating and preventing AD.
Collapse
Affiliation(s)
- Bomi Lee
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jae Gwang Song
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung Wook Kim
- Department of Bio-Integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Kim KM, Hwang NH, Hyun JS, Shin D. Recent Advances in IRAK1: Pharmacological and Therapeutic Aspects. Molecules 2024; 29:2226. [PMID: 38792088 PMCID: PMC11123835 DOI: 10.3390/molecules29102226] [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: 03/04/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Interleukin receptor-associated kinase (IRAK) proteins are pivotal in interleukin-1 and Toll-like receptor-mediated signaling pathways. They play essential roles in innate immunity and inflammation. This review analyzes and discusses the physiological functions of IRAK1 and its associated diseases. IRAK1 is involved in a wide range of diseases such as dry eye, which highlights its potential as a therapeutic target under various conditions. Various IRAK1 inhibitors, including Pacritinib and Rosoxacin, show therapeutic potential against malignancies and inflammatory diseases. The covalent IRAK1 inhibitor JH-X-119-01 shows promise in B-cell lymphomas, emphasizing the significance of covalent bonds in its activity. Additionally, the emergence of selective IRAK1 degraders, such as JNJ-101, provides a novel strategy by targeting the scaffolding function of IRAK1. Thus, the evolving landscape of IRAK1-targeted approaches provides promising avenues for increasingly safe and effective therapeutic interventions for various diseases.
Collapse
Affiliation(s)
| | | | - Ja-Shil Hyun
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon 21935, Republic of Korea
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon 21935, Republic of Korea
| |
Collapse
|
4
|
Hans S, Stanton JE, Sauer AK, Shiels K, Saha SK, Lordan R, Tsoupras A, Zabetakis I, Grabrucker AM. Polar lipids modify Alzheimer's Disease pathology by reducing astrocyte pro-inflammatory signaling through platelet-activating factor receptor (PTAFR) modulation. Lipids Health Dis 2024; 23:113. [PMID: 38643113 PMCID: PMC11031880 DOI: 10.1186/s12944-024-02106-z] [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: 11/22/2023] [Accepted: 04/11/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Pro-inflammatory processes triggered by the accumulation of extracellular amyloid beta (Aβ) peptides are a well-described pathology in Alzheimer's disease (AD). Activated astrocytes surrounding Aβ plaques contribute to inflammation by secreting proinflammatory factors. While astrocytes may phagocytize Aβ and contribute to Aβ clearance, reactive astrocytes may also increase Aβ production. Therefore, identifying factors that can attenuate astrocyte activation and neuroinflammation and how these factors influence pro-inflammatory pathways is important for developing therapeutic and preventive strategies in AD. Here, we identify the platelet-activating factor receptor (PTAFR) pathway as a key mediator of astrocyte activation. Intriguingly, several polar lipids (PLs) have exhibited anti-inflammatory protective properties outside the central nervous system through their inhibitory effect on the PTAFR pathway. Thus, we additionally investigated whether different PLs also exert inhibitory effects on the PAF pathway in astrocytes and whether their presence influences astrocytic pro-inflammatory signaling and known AD pathologies in vitro. METHODS PLs from salmon and yogurt were extracted using novel food-grade techniques and their fatty acid profile was determined using LC/MS. The effect of PLs on parameters such as astrocyte activation and generation of oxygen species (ROS) was assessed. Additionally, effects of the secretome of astrocytes treated with these polar lipids on aged neurons was measured. RESULTS We show that PLs obtained from salmon and yogurt lower astrocyte activation, the generation of reactive oxygen species (ROS), and extracellular Aβ accumulation. Cell health of neurons exposed to the secretome of astrocytes treated with salmon-derived PLs and Aβ was less affected than those treated with astrocytes exposed to Aβ only. CONCLUSION Our results highlight a novel underlying mechanism, why consuming PL-rich foods such as fish and dairy may reduce the risk of developing dementia and associated disorders.
Collapse
Affiliation(s)
- Sakshi Hans
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
| | - Janelle E Stanton
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
| | - Ann Katrin Sauer
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland
| | - Katie Shiels
- Shannon Applied Biotechnology Centre, Technological University of the Shannon, Moylish Park, Limerick, V94E8YF, Ireland
| | - Sushanta Kumar Saha
- Shannon Applied Biotechnology Centre, Technological University of the Shannon, Moylish Park, Limerick, V94E8YF, Ireland
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Systems Pharmacology and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandros Tsoupras
- Hephaestus Laboratory, Department of Chemistry, School of Science, Democritus University of Thrace, Kavala University Campus, Kavala, GR65404, Greece
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, V94PH61, Ireland.
- Bernal Institute, University of Limerick, Analog Devices Building AD3-018, Castletroy, Limerick, V94PH61, Ireland.
- Health Research Institute (HRI), University of Limerick, Limerick, V94PH61, Ireland.
| |
Collapse
|
5
|
Berezutsky MA, Durnova NA, Andronova TA. [Ginkgolide B: mechanisms of neurobiological effects, prospects for use in the therapy of Alzheimer's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:22-27. [PMID: 38676673 DOI: 10.17116/jnevro202412404122] [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] [Indexed: 04/29/2024]
Abstract
The review presents an analysis of experimental data on the study of neurobiological effects of ginkgolide B, which may find application in the therapy of Alzheimer's disease (AD). Ginkgolide B is a diterpene trilactone isolated from the leaves of the relict woody plant Ginkgo biloba L., which has been used for thousands of years in traditional Chinese medicine as a neuroprotective agent. In recent years, this compound has attracted attention because of its wide range of neurobiological effects. The neuroprotective effect of ginkgolide B on brain neurons when exposed to various neurotoxins has been established. This compound has also been shown to effectively protect neurons from the effects of beta-amyloid. Studies have revealed the ability of ginkgolide B to reduce microglia activity and regulate neurotransmitter release. In vivo experiments have shown that this substance significantly increases the expression of brain-derived neurotrophic factor (BDNF) and improves cognitive functions, including memory and learning. It is concluded that ginkgolide B, apparently, may find application in the future as a multi-targeted agent of complex therapy of AD.
Collapse
Affiliation(s)
- M A Berezutsky
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - N A Durnova
- Razumovsky Saratov State Medical University, Saratov, Russia
| | - T A Andronova
- Razumovsky Saratov State Medical University, Saratov, Russia
| |
Collapse
|
6
|
Li J, Peng H, Zhang W, Li M, Wang N, Peng C, Zhang X, Li Y. Enhanced Nose-to-Brain Delivery of Combined Small Interfering RNAs Using Lesion-Recognizing Nanoparticles for the Synergistic Therapy of Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2023; 15:53177-53188. [PMID: 37939350 DOI: 10.1021/acsami.3c08756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Gene therapy has great potential in treating neurodegenerative diseases with complex pathologies. The combination of small interfering RNAs (siRNAs) targeting β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and caspase-3 will provide an effective treatment option for Alzheimer's disease (AD). To overcome the multiple physiological barriers and improve the therapeutic efficacy of siRNAs, lesion-recognizing nanoparticles (NPs) are constructed in this study for the synergistic treatment of AD. The lesion-recognizing NPs contain rabies virus glycoprotein peptide-modified mesenchymal stem cell-derived exosomes as the shell and a reactive oxygen species (ROS)-responsive polymer loaded with siRNAs as the core. After intranasal administration, the lesion-recognizing NPs cross the nasal mucosa and migrate to the affected brain areas. Furthermore, the NPs recognize the target cells and fuse with the cell membranes of neurons. The cores of NPs directly enter into the cytoplasm and achieve the controlled release of siRNAs in a high-ROS environment to downregulate the level of BACE1 and caspase-3 to ameliorate neurologic injury. In addition, lesion-recognizing NPs can significantly reduce the number of reactive astrocytes. Lesion-recognizing NPs have a positive effect on regulating the phase of neurons and astrocytes, which results in better restoration of memory deficits in 3 × Tg-AD mice. Therefore, this work provides a promising platform for neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Jiaxin Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Huan Peng
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Wen Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Muzi Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Nan Wang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Chen Peng
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xinyue Zhang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yan Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
7
|
Zhang Y, Chen D, Tian R, Yan X, Zhou Y. Resveratrol alleviates amyloid β-induced neuronal apoptosis, inflammation, and oxidative and endoplasmic reticulum stress by circ_0050263/miR-361-3p/PDE4A axis during Alzheimer's disease. Chem Biol Drug Des 2023; 102:1121-1132. [PMID: 37620166 DOI: 10.1111/cbdd.14313] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023]
Abstract
Resveratrol (Res) has been identified to reduce neurodegeneration. Circular RNAs (circRNAs) are stable noncoding RNAs that are considered to be ideal biomarkers for molecular targeting treatment. Here, this study focused on investigating the function and relationship of circ_0050263 and Res in Alzheimer's Disease (AD). Human neuroblastoma cell line SK-N-SH was exposed to amyloid-β (Aβ) to induce AD cell model in vitro. Cell viability, apoptosis, and inflammatory reaction were evaluated by CCK-8 assay, flow cytometery, and ELISA analysis. The oxidative stress and endoplasmic reticulum stress (ERS) were determined by detecting related markers. Levels of genes and proteins were detected by qRT-PCR and Western blot. Dual-luciferase reporter assay was adopted to verify the binding between miR-361-3p and circ_0050263 or PDE4A (Phosphodiesterase 4A). Subsequently, we found that Res treatment alleviated Aβ-induced apoptosis, inflammatory response, oxidative stress, and ERS in SK-N-SH cells. Circ_0050263 is a stable circRNA, which was increased by Aβ, but decreased by Res in SK-N-SH cells. Circ_0050263 overexpression reversed Res-induced neuroprotective effects. Mechanistically, circ_0050263 acted as a sponge for miR-361-3p, which targeted PDE4A. Circ_0050263 silencing abated Aβ-induced neuronal injury, which were counteracted by following PDE4A overexpression. Moreover, PDE4A upregulation could attenuate Res-mediated neuroprotective effects. In all, Res alleviated Aβ-induced neuronal apoptosis, inflammation, oxidative stress, and ERS via circ_0050263/miR-361-3p/PDE4A axis, providing new insights for AD therapy.
Collapse
Affiliation(s)
- Yanchun Zhang
- Department of Rehabilitation, Cangzhou Central Hospital, Cangzhou, China
| | - Deqiang Chen
- Department of CT Room, Cangzhou Central Hospital, Cangzhou, China
| | - Rui Tian
- Department of Rehabilitation, Cangzhou Central Hospital, Cangzhou, China
| | - Xinyue Yan
- Department of Rehabilitation, Cangzhou Central Hospital, Cangzhou, China
| | - Yingwen Zhou
- Department of Magnetic Resonance Imaging Room, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
8
|
Nanclares C, Noriega-Prieto JA, Labrada-Moncada FE, Cvetanovic M, Araque A, Kofuji P. Altered calcium signaling in Bergmann glia contributes to spinocerebellar ataxia type-1 in a mouse model of SCA1. Neurobiol Dis 2023; 187:106318. [PMID: 37802154 PMCID: PMC10624966 DOI: 10.1016/j.nbd.2023.106318] [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: 08/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a neurodegenerative disease caused by an abnormal expansion of glutamine (Q) encoding CAG repeats in the ATAXIN1 (ATXN1) gene and characterized by progressive cerebellar ataxia, dysarthria, and eventual deterioration of bulbar functions. SCA1 shows severe degeneration of cerebellar Purkinje cells (PCs) and activation of Bergmann glia (BG), a type of cerebellar astroglia closely associated with PCs. Combining electrophysiological recordings, calcium imaging techniques, and chemogenetic approaches, we have investigated the electrical intrinsic and synaptic properties of PCs and the physiological properties of BG in SCA1 mouse model expressing mutant ATXN1 only in PCs. PCs of SCA1 mice displayed lower spontaneous firing rate and larger slow afterhyperpolarization currents (sIAHP) than wildtype mice, whereas the properties of the synaptic inputs were unaffected. BG of SCA1 mice showed higher calcium hyperactivity and gliotransmission, manifested by higher frequency of NMDAR-mediated slow inward currents (SICs) in PC. Preventing the BG calcium hyperexcitability of SCA1 mice by loading BG with the calcium chelator BAPTA restored sIAHP and spontaneous firing rate of PCs to similar levels of wildtype mice. Moreover, mimicking the BG hyperactivity by activating BG expressing Gq-DREADDs in wildtype mice reproduced the SCA1 pathological phenotype of PCs, i.e., enhancement of sIAHP and decrease of spontaneous firing rate. These results indicate that the intrinsic electrical properties of PCs, but not their synaptic properties, were altered in SCA1 mice and that these alterations were associated with the hyperexcitability of BG. Moreover, preventing BG hyperexcitability in SCA1 mice and promoting BG hyperexcitability in wildtype mice prevented and mimicked, respectively, the pathological electrophysiological phenotype of PCs. Therefore, BG plays a relevant role in the dysfunction of the electrical intrinsic properties of PCs in SCA1 mice, suggesting that they may serve as potential targets for therapeutic approaches to treat the spinocerebellar ataxia type 1.
Collapse
Affiliation(s)
- Carmen Nanclares
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | - Marija Cvetanovic
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alfonso Araque
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Naik S, Katariya R, Shelke S, Patravale V, Umekar M, Kotagale N, Taksande B. Nattokinase prevents β-amyloid peptide (Aβ 1-42) induced neuropsychiatric complications, neuroinflammation and BDNF signalling disruption in mice. Eur J Pharmacol 2023; 952:175821. [PMID: 37263404 DOI: 10.1016/j.ejphar.2023.175821] [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: 01/18/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/03/2023]
Abstract
Alzheimer's disease (AD) is a chronic and progressive neurodegenerative disorder characterized by abnormal accumulation of extracellular β-amyloid (Aβ) plaques and neuronal damage. Although AD is typically considered a cognitive neurodegenerative disorder, almost all people diagnosed with AD develop neuropsychiatric complications at some stage in their life span. The present study investigated the effect of chronic Nattokinase (NK) administration on β-Amyloid peptide (Aβ1-42) induced neuropsychiatric conditions (depression-like behaviour, anxiety, and memory impairment) in mice. Aβ1-42 peptide injected mice demonstrated depression, anxiety, and impairment of cognitive abilities evaluated as increased immobility time in forced swim test (FST), decreased open arm time/entries in elevated plus maze (EPM) and reference and working memory error in radial arm maze (RAM) respectively with elevation in Interleukin-6 (IL-6), Tumour necrosis factor-α (TNF-α), reduction in Interleukin-10 (IL-10) and Brain-derived neurotrophic factor (BDNF) immunocontent within the hippocampus. Chronic administration of NK (50-100 mg/kg, i.p.) from day 8-27, prevented depression-like behaviour, anxiety, and memory impairment and normalized the neurochemical alteration within the hippocampus of mice injected with Aβ1-42 peptide. The effect of NK on psychiatric complications, learning, and memory was comparable to peripheral donepezil treatment. This study suggests that NK improves learning, memory impairment, and neuropsychiatric complications possibly through the downregulation of neuroinflammatory pathways and restoring BDNF signalling in AD.
Collapse
Affiliation(s)
- Shivraj Naik
- Pharmaceutical Science & Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Raj Katariya
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, MS, 441 002, India
| | - Shraddha Shelke
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, MS, 441 002, India
| | - Vandana Patravale
- Pharmaceutical Science & Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Milind Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, MS, 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, MS, 444604, India
| | - Brijesh Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, MS, 441 002, India.
| |
Collapse
|
10
|
O’Day DH. Alzheimer's Disease beyond Calcium Dysregulation: The Complex Interplay between Calmodulin, Calmodulin-Binding Proteins and Amyloid Beta from Disease Onset through Progression. Curr Issues Mol Biol 2023; 45:6246-6261. [PMID: 37623212 PMCID: PMC10453589 DOI: 10.3390/cimb45080393] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
A multifactorial syndrome, Alzheimer's disease is the main cause of dementia, but there is no existing therapy to prevent it or stop its progression. One of the earliest events of Alzheimer's disease is the disruption of calcium homeostasis but that is just a prelude to the disease's devastating impact. Calcium does not work alone but must interact with downstream cellular components of which the small regulatory protein calmodulin is central, if not primary. This review supports the idea that, due to calcium dyshomeostasis, calmodulin is a dominant regulatory protein that functions in all stages of Alzheimer's disease, and these regulatory events are impacted by amyloid beta. Amyloid beta not only binds to and regulates calmodulin but also multiple calmodulin-binding proteins involved in Alzheimer's. Together, they act on the regulation of calcium dyshomeostasis, neuroinflammation, amyloidogenesis, memory formation, neuronal plasticity and more. The complex interactions between calmodulin, its binding proteins and amyloid beta may explain why many therapies have failed or are doomed to failure unless they are considered.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
11
|
Das TK, Ganesh BP, Fatima-Shad K. Common Signaling Pathways Involved in Alzheimer's Disease and Stroke: Two Faces of the Same Coin. J Alzheimers Dis Rep 2023; 7:381-398. [PMID: 37220617 PMCID: PMC10200243 DOI: 10.3233/adr-220108] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Alzheimer's disease (AD) and stroke are two interrelated neurodegenerative disorders which are the leading cause of death and affect the neurons in the brain and central nervous system. Although amyloid-β aggregation, tau hyperphosphorylation, and inflammation are the hallmarks of AD, the exact cause and origin of AD are still undefined. Recent enormous fundamental discoveries suggest that the amyloid hypothesis of AD has not been proven and anti-amyloid therapies that remove amyloid deposition have not yet slowed cognitive decline. However, stroke, mainly ischemic stroke (IS), is caused by an interruption in the cerebral blood flow. Significant features of both disorders are the disruption of neuronal circuitry at different levels of cellular signaling, leading to the death of neurons and glial cells in the brain. Therefore, it is necessary to find out the common molecular mechanisms of these two diseases to understand their etiological connections. Here, we summarized the most common signaling cascades including autotoxicity, ApoE4, insulin signaling, inflammation, mTOR-autophagy, notch signaling, and microbiota-gut-brain axis, present in both AD and IS. These targeted signaling pathways reveal a better understanding of AD and IS and could provide a distinguished platform to develop improved therapeutics for these diseases.
Collapse
Affiliation(s)
- Tushar Kanti Das
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bhanu Priya Ganesh
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kaneez Fatima-Shad
- School of Life Sciences, University of Technology Sydney, NSW, Australia
- Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia
- School of Behavioral and Health Sciences, Faculty of Health Sciences, Australian Catholic University, NSW, Australia
| |
Collapse
|
12
|
The Role of the JAK/STAT Signaling Pathway in the Pathogenesis of Alzheimer's Disease: New Potential Treatment Target. Int J Mol Sci 2023; 24:ijms24010864. [PMID: 36614305 PMCID: PMC9821184 DOI: 10.3390/ijms24010864] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/19/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. However, emerging evidence suggests that neuroinflammation, mediated notably by activated neuroglial cells, neutrophils, and macrophages, also plays an important role in the pathogenesis of Alzheimer's disease. Therefore, understanding the interplay between the nervous and immune systems might be the key to the prevention or delay of Alzheimer's disease progression. One of the most important mechanisms determining gliogenic cell fate is the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway that is influenced by the overactivation of microglia and astrocytes. The JAK/STAT signaling pathway is one of the critical factors that promote neuroinflammation in neurodegenerative diseases such as Alzheimer's disease by initiating innate immunity, orchestrating adaptive immune mechanisms, and finally, constraining neuroinflammatory response. Since a chronic neuroinflammatory environment in the brain is a hallmark of Alzheimer's disease, understanding the process would allow establishing the underlying role of neuroinflammation, then estimating the prognosis of Alzheimer's disease development and finding a new potential treatment target. In this review, we highlight the recent advances in the potential role of JAK/STAT signaling in neurological diseases with a focus on discussing future research directions regarding novel therapeutic approaches and predictive biomarkers for Alzheimer's disease.
Collapse
|
13
|
Sarailoo M, Afshari S, Asghariazar V, Safarzadeh E, Dadkhah M. Cognitive Impairment and Neurodegenerative Diseases Development Associated with Organophosphate Pesticides Exposure: a Review Study. Neurotox Res 2022; 40:1624-1643. [PMID: 36066747 DOI: 10.1007/s12640-022-00552-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/22/2022] [Accepted: 07/27/2022] [Indexed: 10/14/2022]
Abstract
A significant body of literature emphasizes the role of insecticide, particularly organophosphates (OPs), as the major environmental factor in the etiology of neurodegenerative diseases. This review aims to study the relationship between OP insecticide exposure, cognitive impairment, and neurodegenerative disease development. Human populations, especially in developing countries, are frequently exposed to OPs due to their extensive applications. The involvement of various signaling pathways in OP neurotoxicity are reported, but the OP-induced cognitive impairment and link between OP exposure and the pathophysiology of neurodegenerative diseases are not clearly understood. In the present review, we have therefore aimed to come to new conclusions which may help to find protective and preventive strategies against OP neurotoxicity and may establish a possible link between organophosphate exposure, cognitive impairment, and OP-induced neurotoxicity. Moreover, we discuss the findings obtained from animal and human research providing some support for OP-induced cognitive impairment and neurodegenerative disorders.
Collapse
Affiliation(s)
- Mehdi Sarailoo
- Students Research Committee, School of Public Health, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Salva Afshari
- Students Research Committee, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Masoomeh Dadkhah
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
14
|
Li WY, Lee CY, Lee KM, Zhang G, Lyu A, Yue KKM. Advanced Glycation End-Product Precursor Methylglyoxal May Lead to Development of Alzheimer's Disease. Diabetes Metab Syndr Obes 2022; 15:3153-3166. [PMID: 36262805 PMCID: PMC9575592 DOI: 10.2147/dmso.s382927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/05/2022] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Diabetes mellitus (DM) is characterized by chronic hyperglycemia and diabetic complications. Exacerbated cortical neuronal degeneration was observed in Alzheimer's disease (AD) patients with DM. In fact, DM is now considered a risk factor of AD, as DM-induced activation of stress responses in the central nervous system (CNS) such as oxidative stress and neuroinflammation may lead to various neurodegenerative disorders. Methylglyoxal (MG) is one of the most reactive advanced glycation end-product (AGE) precursors. Abnormal accumulation of MG is observed in the serum of diabetic patients. As MG is reported to promote brain cells impairment in the CNS, and it is found that AGEs are abnormally increased in the brains of AD patients. Therefore, the effect of MG causing subsequent symptoms of AD was investigated. METHODS 5-week-old C57BL/6 mice were intraperitoneally injected with MG solution for 11 weeks. The Morris water maze (MWM) was used to examine the spatial learning ability and cognition of mice. After MG treatment, MTT assay, real-time PCR analyses, and Western blot were performed to assess the harvested astrocytes and hippocampi. RESULTS Significantly longer escape latency and reduced percentage time spent in the target quadrant were observed in the 9-week-MG-treated mice. We have found in both in vitro and in vivo models that MG induced astrogliosis, pro-inflammatory cytokines, AD-related markers, and ERK activation. Further, trend of normalization of the tested markers mRNA expressions were observed after ERK inhibition. CONCLUSION Our in vivo results suggested that MG could induce AD symptoms and in vitro results implied that ERK may regulate the promotion of inflammation and Aβ formation in MG-induced reactive astrocytes. Taken together, MG may participate in the dysfunction of brain cells resulting in possible diabetes-related neurodegeneration by promoting astrogliosis, Aβ production, and neuroinflammation through the ERK pathway. Our findings provide insight of targeting ERK as a therapeutic application for diabetes-induced AD.
Collapse
Affiliation(s)
- Wai Yin Li
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Cheuk Yan Lee
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Kwan Ming Lee
- Department of Biology, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Ge Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Aiping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, Hong Kong Baptist University, Hong Kong, People’s Republic of China
| | - Kevin Kin Man Yue
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People’s Republic of China
- Correspondence: Kevin Kin Man Yue, 4/F, Jockey Club School of Chinese Medicine Building, 7 Baptist University Road, Kowloon Tong, Kowloon, Tel +852 3411 2468, Email
| |
Collapse
|
15
|
Chen J, Li Q, Zhu J, Yuan Z, Wang T, Song J. GPR40 Agonist Ameliorate Pathological Neuroinflammation of Alzheimer's Disease via the Modulation of Gut Microbiota and Immune System, a Mini-Review. Neurotox Res 2021; 39:2175-2185. [PMID: 34505972 DOI: 10.1007/s12640-021-00408-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a central disease with high incidence, and its pathological process is closely associated with changes of some biological indicators in the periphery. Among them, the intestinal flora mainly causes a series of pathological changes such as inflammation through the immune system, which may contribute to the pathological process of AD. In this paper, we mainly focused the relationship between gut microbiota and immune system disorder in the neuropathology of AD, underlining the significance of the advanced mechanism of inflammatory response and providing a new direction for the treatment of AD.
Collapse
Affiliation(s)
- Jianheng Chen
- Department of Anesthesiology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Zhu
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zijing Yuan
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Wang
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Song
- Department of Nephrology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Li Y, Laws SM, Miles LA, Wiley JS, Huang X, Masters CL, Gu BJ. Genomics of Alzheimer's disease implicates the innate and adaptive immune systems. Cell Mol Life Sci 2021; 78:7397-7426. [PMID: 34708251 PMCID: PMC11073066 DOI: 10.1007/s00018-021-03986-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/14/2021] [Accepted: 10/16/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease characterised by cognitive impairment, behavioural alteration, and functional decline. Over 130 AD-associated susceptibility loci have been identified by genome-wide association studies (GWAS), while whole genome sequencing (WGS) and whole exome sequencing (WES) studies have identified AD-associated rare variants. These variants are enriched in APOE, TREM2, CR1, CD33, CLU, BIN1, CD2AP, PILRA, SCIMP, PICALM, SORL1, SPI1, RIN3, and more genes. Given that aging is the single largest risk factor for late-onset AD (LOAD), the accumulation of somatic mutations in the brain and blood of AD patients have also been explored. Collectively, these genetic findings implicate the role of innate and adaptive immunity in LOAD pathogenesis and suggest that a systemic failure of cell-mediated amyloid-β (Aβ) clearance contributes to AD onset and progression. AD-associated variants are particularly enriched in myeloid-specific regulatory regions, implying that AD risk variants are likely to perturbate the expression of myeloid-specific AD-associated genes to interfere Aβ clearance. Defective phagocytosis, endocytosis, and autophagy may drive Aβ accumulation, which may be related to naturally-occurring antibodies to Aβ (Nabs-Aβ) produced by adaptive responses. Passive immunisation is providing efficiency in clearing Aβ and slowing cognitive decline, such as aducanumab, donanemab, and lecanemab (ban2401). Causation of AD by impairment of the innate immunity and treatment using the tools of adaptive immunity is emerging as a new paradigm for AD, but immunotherapy that boosts the innate immune functions of myeloid cells is highly expected to modulate disease progression at asymptomatic stage.
Collapse
Affiliation(s)
- Yihan Li
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, 270 Joondalup Dr, Joondalup, WA, 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Dr, Joondalup, WA, 6027, Australia
| | - Luke A Miles
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - James S Wiley
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Xin Huang
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Ben J Gu
- The Florey Institute, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
17
|
Rather MA, Khan A, Alshahrani S, Rashid H, Qadri M, Rashid S, Alsaffar RM, Kamal MA, Rehman MU. Inflammation and Alzheimer's Disease: Mechanisms and Therapeutic Implications by Natural Products. Mediators Inflamm 2021; 2021:9982954. [PMID: 34381308 PMCID: PMC8352708 DOI: 10.1155/2021/9982954] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/24/2021] [Accepted: 07/10/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with no clear causative event making the disease difficult to diagnose and treat. The pathological hallmarks of AD include amyloid plaques, neurofibrillary tangles, and widespread neuronal loss. Amyloid-beta has been extensively studied and targeted to develop an effective disease-modifying therapy, but the success rate in clinical practice is minimal. Recently, neuroinflammation has been focused on as the event in AD progression to be targeted for therapies. Various mechanistic pathways including cytokines and chemokines, complement system, oxidative stress, and cyclooxygenase pathways are linked to neuroinflammation in the AD brain. Many cells including microglia, astrocytes, and oligodendrocytes work together to protect the brain from injury. This review is focused to better understand the AD inflammatory and immunoregulatory processes to develop novel anti-inflammatory drugs to slow down the progression of AD.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Tamil Nadu 608002, India
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hina Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Marwa Qadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Rana M. Alsaffar
- Department of Pharmacology & Toxicology, College of Pharmacy Girls Section, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770; Novel Global Community Educational Foundation, Australia
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
Nanclares C, Baraibar AM, Araque A, Kofuji P. Dysregulation of Astrocyte-Neuronal Communication in Alzheimer's Disease. Int J Mol Sci 2021; 22:7887. [PMID: 34360652 PMCID: PMC8346080 DOI: 10.3390/ijms22157887] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/16/2022] Open
Abstract
Recent studies implicate astrocytes in Alzheimer's disease (AD); however, their role in pathogenesis is poorly understood. Astrocytes have well-established functions in supportive functions such as extracellular ionic homeostasis, structural support, and neurovascular coupling. However, emerging research on astrocytic function in the healthy brain also indicates their role in regulating synaptic plasticity and neuronal excitability via the release of neuroactive substances named gliotransmitters. Here, we review how this "active" role of astrocytes at synapses could contribute to synaptic and neuronal network dysfunction and cognitive impairment in AD.
Collapse
Affiliation(s)
| | | | | | - Paulo Kofuji
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA; (C.N.); (A.M.B.); (A.A.)
| |
Collapse
|
19
|
Azabou E, Bao G, Costantino F, Jacota M, Lazizi C, Nkam L, Rottman M, Roux AL, Chevallier S, Grimaldi L, Breban M. Randomized Cross Over Study Assessing the Efficacy of Non-invasive Stimulation of the Vagus Nerve in Patients With Axial Spondyloarthritis Resistant to Biotherapies: The ESNV-SPA Study Protocol. Front Hum Neurosci 2021; 15:679775. [PMID: 34276328 PMCID: PMC8278783 DOI: 10.3389/fnhum.2021.679775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/31/2021] [Indexed: 01/04/2023] Open
Abstract
Axial spondyloarthritis (SpA), is a major cause of chronic pain and disability that profoundly alters the quality of life of patients. Nearly half of patients with SpA usually develop drug resistance. Non-pharmacological treatments targeting inflammation are an attractive alternative to drug administration. Vagus nerve stimulation (VNS), by promoting a cholinergic anti-inflammatory reflex holds promise for treating inflammatory disease. Inflammatory reflex signaling, which is enhanced by electrically stimulating the vagus nerve, significantly reduces cytokine production and attenuates disease severity in animal models of endotoxemia, sepsis, colitis, and other preclinical models of inflammatory diseases. It has been proposed that vagal efferent fibers release acetylcholine (Ach), which can interact with α7-subunit-containing nicotinic receptors expressed by tissue macrophages and other immune cells to rapidly inhibit the synthesis/release of pro-inflammatory cytokines such as TNFα, IL-1β, IL-6, and IL-18. External vagal nerve stimulation devices are now available that do not require surgery nor implantation to non-invasively stimulate the vagal nerve. This double-blind randomized cross-over clinical trial aims to study the change in SpA disease activity, according to Assessment in Ankylosing Spondylitis 20 (ASAS20) definition, after 12 weeks of non-invasive VNS treatment vs. non-specific dummy stimulation (control group). One hundred and twenty adult patients with drug resistant SpA, meeting the ASAS classification criteria, will be included in the study. Patients will be randomized into two parallel groups according to a cross over design: either active VNS for 12 weeks, then dummy stimulation for 12 weeks, or dummy stimulation for 12 weeks, then active VNS for 12 weeks. The two stimulation periods will be separated by a 4 weeks wash-out period. A transcutaneous auricular vagus nerve stimulator Tens Eco Plus SCHWA MEDICOTM France will be used in this study. The active VNS stimulation will be applied in the cymba conchae of the left ear upon the auricular branch of the vagus nerve, using low intensity (2–5 mA), once à week, during 1 h. Dummy stimulation will be performed under the same conditions and parameters as active VNS stimulation, but at an irrelevant anatomical site: the left ear lobule. This multicenter study was registered on ClinicalTrials.gov: NCT04286373.
Collapse
Affiliation(s)
- Eric Azabou
- Clinical Neurophysiology and Neuromodulation Unit, Department of Physiology, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Guillaume Bao
- Clinical Neurophysiology and Neuromodulation Unit, Department of Physiology, Raymond Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France
| | - Félicie Costantino
- Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France.,Rheumatology Department, AP-HP, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Laboratory of Excellence Inflamex, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Madalina Jacota
- Clinical Research Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Boulogne-Billancourt, France, University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Chanez Lazizi
- Clinical Research Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Boulogne-Billancourt, France, University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Lionelle Nkam
- Clinical Research Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Boulogne-Billancourt, France, University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Martin Rottman
- Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France.,Microbiology Laboratory, Raymond Poincaré Hospital, AP-HP Paris Saclay University, Paris, France
| | - Anne-Laure Roux
- Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France.,Microbiology Laboratory, Raymond Poincaré Hospital, AP-HP Paris Saclay University, Paris, France
| | - Sylvain Chevallier
- Versailles Engineering Systems Laboratory (LISV), University of Versailles Saint Quentin en Yvelines (UVSQ), Vélizy, France
| | - Lamiae Grimaldi
- Clinical Research Unit, Ambroise Paré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Boulogne-Billancourt, France, University of Versailles Saint-Quentin en Yvelines, Paris-Saclay University, Paris, France
| | - Maxime Breban
- Laboratory of Infection and Inflammation (2I)-Inserm UMR 1173, University of Versailles Saint-Quentin en Yvelines (UVSQ), Paris-Saclay University, Paris, France.,Rheumatology Department, AP-HP, Ambroise Paré Hospital, AP-HP, Boulogne-Billancourt, France.,Laboratory of Excellence Inflamex, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
20
|
Tautou M, Eddarkaoui S, Descamps F, Larchanché PE, El Bakali J, Goveas LM, Dumoulin M, Lamarre C, Blum D, Buée L, Melnyk P, Sergeant N. A ß-Secretase Modulator Decreases Tau Pathology and Preserves Short-Term Memory in a Mouse Model of Neurofibrillary Degeneration. Front Pharmacol 2021; 12:679335. [PMID: 34267657 PMCID: PMC8276176 DOI: 10.3389/fphar.2021.679335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/18/2021] [Indexed: 01/24/2023] Open
Abstract
Identifying which among several in cellulo pharmacological activities is necessary for the proper in vivo activity is essential for further drug development against Alzheimer’s disease pathophysiological processes. An in-depth structure–activity relationship–based study has been carried out, and two molecules, named MAGS02-14 and PEL24-199, that share a ß-secretase modulatory effect associated or not to a lysosomotropic activity in cellulo have been identified. In terms of chemical formulas, MAGS02-14 and PEL24-199 only differ from each other by a single nitrogen atom. The study aimed to elucidate the in vivo pharmacological effects of lysosomotropic and/or the ß-secretase modulatory activity in a tau pathology mouse model. To address this question, the THY-Tau22 transgenic model of tauopathy was treated with both compounds for 6 weeks in a curative paradigm. Short-term memory, tau burden, and inflammatory processes were analyzed using orthogonal methods, and PEL24-199, but not MAGS02-14, was shown to restore the short-term memory and reduce the neurofibrillary degenerating process. These effects were associated with a reduced phosphorylation of tau, an increased phosphatase expression, and decreased astrogliosis. Our results, therefore, suggest that the lysosomotropic activity may be nonessential for the effect on tau pathology.
Collapse
Affiliation(s)
- Marie Tautou
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Sabiha Eddarkaoui
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Florian Descamps
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Paul-Emmanuel Larchanché
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Jamal El Bakali
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Liesel Mary Goveas
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Mélanie Dumoulin
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Chloé Lamarre
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - David Blum
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Luc Buée
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Patricia Melnyk
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France
| | - Nicolas Sergeant
- Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience and Cognition, University of Lille, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
21
|
Wang J, Ding Y, Zhuang L, Wang Z, Xiao W, Zhu J. Ginkgolide B‑induced AMPK pathway activation protects astrocytes by regulating endoplasmic reticulum stress, oxidative stress and energy metabolism induced by Aβ1‑42. Mol Med Rep 2021; 23:457. [PMID: 33880582 PMCID: PMC8072312 DOI: 10.3892/mmr.2021.12096] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ginkgolide B (GB), the diterpenoid lactone compound isolated from the extracts of Ginkgo biloba leaves, significantly improves cognitive impairment, but its potential pharmacological effect on astrocytes induced by β-amyloid (Aβ)1-42 remains to be elucidated. The present study aimed to investigate the protective effect and mechanism of GB on astrocytes with Aβ1-42-induced apoptosis in Alzheimer's disease (AD). Astrocytes obtained from Sprague Dawley rats were randomly divided into control, Aβ, GB and GB + compound C groups. Cell viability and apoptosis were analyzed using Cell Counting Kit-8 and flow cytometry assays, respectively. Protein and mRNA expression levels were analyzed using western blotting and reverse transcription-quantitative PCR, respectively. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-Px), reactive oxygen species (ROS) and ATP were determined using the corresponding commercial kits. The findings revealed that GB attenuated Aβ1-42-induced apoptosis and the 5′ adenosine monophosphate- activated protein kinase (AMPK) inhibitor compound C reversed the protective effects of GB. In addition, GB reversed Aβ1-42-induced oxidative damage and energy metabolism disorders, including decreases in the levels of SOD, GSH-Px and ATP and increased the levels of MDA and ROS in astrocytes, while compound C reversed the anti-oxidative effect and the involvement of GB in maintaining energy metabolism in astrocytes. Finally, GB decreased the expression levels of the endoplasmic reticulum stress (ERS) proteins and the apoptotic protein CHOP and increased both mRNA and protein expression of the components of the energy metabolism-related AMPK/peroxisome proliferator-activated receptor γ coactivator 1α/peroxisome proliferator-activated receptor α and anti-oxidation-related nuclear respiratory factor 2/heme oxygenase 1/NAD(P)H dehydrogenase (quinone 1) pathways and downregulated the expression of β-secretase 1. However, compound C could antagonize these effects. In conclusion, the findings demonstrated that GB protected against Aβ1-42-induced apoptosis by inhibiting ERS, oxidative stress, energy metabolism disorders and Aβ1-42 production probably by activating AMPK signaling pathways. The findings provided an innovative insight into the treatment using GB as a therapeutic in Aβ1-42-related AD.
Collapse
Affiliation(s)
- Jing Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Yan Ding
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Linwu Zhuang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu 222000, P.R. China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, Jiangsu 222000, P.R. China
| | - Jingbo Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, P.R. China
| |
Collapse
|
22
|
Fleeman RM, Proctor EA. Astrocytic Propagation of Tau in the Context of Alzheimer's Disease. Front Cell Neurosci 2021; 15:645233. [PMID: 33815065 PMCID: PMC8010320 DOI: 10.3389/fncel.2021.645233] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/19/2021] [Indexed: 01/14/2023] Open
Abstract
More than 6 million Americans are currently living with Alzheimer's disease (AD), and the incidence is growing rapidly with our aging population. Numerous therapeutics have failed to make it to the clinic, potentially due to a focus on presumptive pathogenic proteins instead of cell-type-specific signaling mechanisms. The tau propagation hypothesis that inter-neuronal tau transfer drives AD pathology has recently garnered attention, as accumulation of pathological tau in the brain has high clinical significance in correlating with progression of cognitive AD symptoms. However, studies on tau pathology in AD are classically neuron-centric and have greatly overlooked cell-type specific effects of tau internalization, degradation, and propagation. While the contribution of microglia to tau processing and propagation is beginning to be recognized and understood, astrocytes, glial cells in the brain important for maintaining neuronal metabolic, synaptic, trophic, and immune function which can produce, internalize, degrade, and propagate tau are understudied in their ability to affect AD progression through tau pathology. Here, we showcase evidence for whether tau uptake by astrocytes may be beneficial or detrimental to neuronal health and how astrocytes and their immunometabolic functions may be key targets for future successful AD therapies.
Collapse
Affiliation(s)
- Rebecca M Fleeman
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| | - Elizabeth A Proctor
- Department of Neurosurgery, Department of Pharmacology, College of Medicine, Pennsylvania State University (PSU), Hershey, PA, United States.,Department of Biomedical Engineering, Department of Engineering Science and Mechanics, Center for Neural Engineering, Pennsylvania State University (PSU), University Park, PA, United States
| |
Collapse
|
23
|
Acioglu C, Li L, Elkabes S. Contribution of astrocytes to neuropathology of neurodegenerative diseases. Brain Res 2021; 1758:147291. [PMID: 33516810 DOI: 10.1016/j.brainres.2021.147291] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/10/2020] [Accepted: 01/05/2021] [Indexed: 02/08/2023]
Abstract
Classically, the loss of vulnerable neuronal populations in neurodegenerative diseases was considered to be the consequence of cell autonomous degeneration of neurons. However, progress in the understanding of glial function, the availability of improved animal models recapitulating the features of the human diseases, and the development of new approaches to derive glia and neurons from induced pluripotent stem cells obtained from patients, provided novel information that altered this view. Current evidence strongly supports the notion that non-cell autonomous mechanisms contribute to the demise of neurons in neurodegenerative disorders, and glia causally participate in the pathogenesis and progression of these diseases. In addition to microglia, astrocytes have emerged as key players in neurodegenerative diseases and will be the focus of the present review. Under the influence of pathological stimuli present in the microenvironment of the diseased CNS, astrocytes undergo morphological, transcriptional, and functional changes and become reactive. Reactive astrocytes are heterogeneous and exhibit neurotoxic (A1) or neuroprotective (A2) phenotypes. In recent years, single-cell or single-nucleus transcriptome analyses unraveled new, disease-specific phenotypes beyond A1/A2. These investigations highlighted the complexity of the astrocytic responses to CNS pathology. The present review will discuss the contribution of astrocytes to neurodegenerative diseases with particular emphasis on Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Some of the commonalties and differences in astrocyte-mediated mechanisms that possibly drive the pathogenesis or progression of the diseases will be summarized. The emerging view is that astrocytes are potential new targets for therapeutic interventions. A comprehensive understanding of astrocyte heterogeneity and disease-specific phenotypic complexity could facilitate the design of novel strategies to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Cigdem Acioglu
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Lun Li
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| | - Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, United States.
| |
Collapse
|
24
|
Reid MJ, Beltran-Lobo P, Johnson L, Perez-Nievas BG, Noble W. Astrocytes in Tauopathies. Front Neurol 2020; 11:572850. [PMID: 33071951 PMCID: PMC7542303 DOI: 10.3389/fneur.2020.572850] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the progressive accumulation across the brain of hyperphosphorylated aggregates of the microtubule-associated protein tau that vary in isoform composition, structural conformation and localization. Tau aggregates are most commonly deposited within neurons but can show differential association with astrocytes, depending on the disease. Astrocytes, the most abundant neural cells in the brain, play a major role in synapse and neuronal function, and are a key component of the glymphatic system and blood brain barrier. However, their contribution to tauopathy progression is not fully understood. Here we present a brief overview of the association of tau with astrocytes in tauopathies. We discuss findings that support a role for astrocytes in the uptake and spread of pathological tau, and we describe how alterations to astrocyte phenotype in tauopathies may cause functional alterations that impedes their ability to support neurons and/or cause neurotoxicity. The research reviewed here further highlights the importance of considering non-neuronal cells in neurodegeneration and suggests that astrocyte-directed targets that may have utility for therapeutic intervention in tauopathies.
Collapse
Affiliation(s)
- Matthew J Reid
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paula Beltran-Lobo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Louisa Johnson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Beatriz Gomez Perez-Nievas
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
25
|
Involvement of hippocampal agmatine in β1-42 amyloid induced memory impairment, neuroinflammation and BDNF signaling disruption in mice. Neurotoxicology 2020; 80:1-11. [DOI: 10.1016/j.neuro.2020.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 01/25/2023]
|
26
|
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are, respectively, the most prevalent and fastest growing neurodegenerative diseases worldwide. The former is primarily characterized by memory loss and the latter by the motor symptoms of tremor and bradykinesia. Both AD and PD are progressive diseases that share several key underlying mitochondrial, inflammatory, and other metabolic pathologies. This review will detail how these pathologies intersect with ketone body metabolism and signaling, and how ketone bodies, particularly d-β-hydroxybutyrate (βHB), may serve as a potential adjunctive nutritional therapy for two of the world's most devastating conditions.
Collapse
|
27
|
Gottipati MK, Zuidema JM, Gilbert RJ. Biomaterial strategies for creating in vitro astrocyte cultures resembling in vivo astrocyte morphologies and phenotypes. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 14:67-74. [PMID: 34296048 DOI: 10.1016/j.cobme.2020.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Astrocytes are dynamic cells residing in the central nervous system exhibiting many diverse functions. Astrocytes quickly change and present unique phenotypes in response to injury or disease. Here, we briefly summarize recent information regarding astrocyte morphology and function and provide brief insight into their phenotypic changes following injury or disease. We also present the utility of in vitro astrocyte cultures and present recent advances in biomaterial development that enable better recapitulation of their in vivo behavior and morphology.
Collapse
Affiliation(s)
- Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8 Street, Troy, NY. 12180.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8 Street, Troy, NY. 12180.,Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, 460 W. 12 Avenue, Columbus, OH. 43210
| | - Jonathan M Zuidema
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA. 92093
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8 Street, Troy, NY. 12180.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8 Street, Troy, NY. 12180
| |
Collapse
|
28
|
Liccardo D, Marzano F, Carraturo F, Guida M, Femminella GD, Bencivenga L, Agrimi J, Addonizio A, Melino I, Valletta A, Rengo C, Ferrara N, Rengo G, Cannavo A. Potential Bidirectional Relationship Between Periodontitis and Alzheimer's Disease. Front Physiol 2020; 11:683. [PMID: 32719612 PMCID: PMC7348667 DOI: 10.3389/fphys.2020.00683] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia in the elderly population, representing a global public health priority. Despite a large improvement in understanding the pathogenesis of AD, the etiology of this disorder remains still unclear, and no current treatment is able to prevent, slow, or stop its progression. Thus, there is a keen interest in the identification and modification of the risk factors and novel molecular mechanisms associated with the development and progression of AD. In this context, it is worth noting that several findings support the existence of a direct link between neuronal and non-neuronal inflammation/infection and AD progression. Importantly, recent studies are now supporting the existence of a direct relationship between periodontitis, a chronic inflammatory oral disease, and AD. The mechanisms underlying the association remain to be fully elucidated, however, it is generally accepted, although not confirmed, that oral pathogens can penetrate the bloodstream, inducing a low-grade systemic inflammation that negatively affects brain function. Indeed, a recent report demonstrated that oral pathogens and their toxic proteins infect the brain of AD patients. For instance, when AD progresses from the early to the more advanced stages, patients could no longer be able to adequately adhere to proper oral hygiene practices, thus leading to oral dysbiosis that, in turn, fuels infection, such as periodontitis. Therefore, in this review, we will provide an update on the emerging (preclinical and clinical) evidence that supports the relationship existing between periodontitis and AD. More in detail, we will discuss data attesting that periodontitis and AD share common risk factors and a similar hyper-inflammatory phenotype.
Collapse
Affiliation(s)
- Daniela Liccardo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Center for Translational Medicine, Temple University, Philadelphia, PA, United States
| | - Federica Marzano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | | | - Marco Guida
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | - Leonardo Bencivenga
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Jacopo Agrimi
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Armida Addonizio
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Imma Melino
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessandra Valletta
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, Naples, Italy
| | - Carlo Rengo
- Department of Prosthodontics and Dental Materials, School of Dental Medicine, University of Siena, Siena, Italy
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Istituti Clinici Scientifici ICS Maugeri - S.p.A.-Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Scientifico di Telese Terme, Telese, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Istituti Clinici Scientifici ICS Maugeri - S.p.A.-Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Scientifico di Telese Terme, Telese, Italy
| | - Alessandro Cannavo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
29
|
miR-194-5p inhibits LPS-induced astrocytes activation by directly targeting neurexophilin 1. Mol Cell Biochem 2020; 471:203-213. [PMID: 32533463 DOI: 10.1007/s11010-020-03780-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
Astrocytes are vitally involved in the development of neurodegenerative diseases and brain cancers. In this work, we investigated the potential ameliorative role of microRNA-194-5p (miR-194-5p) against lipopolysaccharide (LPS)-induced astrocytes activation and the mechanism underneath. Astrocytes were transfected with miR-194-5p mimic or inhibitor and subsequently induced with LPS. Cell proliferation was measured using MTT assay while Transwell assay was used for assessing cell migration. The concentrations of cyclooxygenase 2 (COX2) and cytokines (tumor necrosis factor-α (TNF-α), transforming growth factor β (TGF-β), interleukin (IL)-1β and IL-6) were determined by enzyme-linked immunosorbent assay (ELISA). Gene expression was assessed by quantitative reverse transcription PCR (RT-qPCR) while western blotting was used for quantifying relative protein expression. We found that miR-194-5p, downregulated in LPS-induced astrocytes, significantly inhibited LPS-induced cell proliferation and migration. In addition, miR-194-5p inhibited the release of COX2 and pro-inflammatory cytokines (TNF-α, TGF-β, IL-1β and IL-6). Moreover, the silencing of neurexophilin 1 (NXPH1), an in silico and mechanistically confirmed direct target of miR-194-5p, reverted the anti-inflammatory, anti-proliferative and anti-migratory effects of miR-194-5p. We anticipated that miR-194-5 inhibits the proliferation, invasion, and inflammatory reaction in LPS-induced astrocytes by directly targeting NXPH1. These findings hinted that miR-194-5p/NXPH1 axis exerts vital functions in astrocytes activation and neuroinflammation-associated diseases. This finding will open novel avenues for biomedical and neuroscience research.
Collapse
|
30
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Zhang L, Dong ZF, Zhang JY. Immunomodulatory role of mesenchymal stem cells in Alzheimer's disease. Life Sci 2020; 246:117405. [PMID: 32035129 DOI: 10.1016/j.lfs.2020.117405] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia and is characterized by gradual loss in memory, language, and cognitive function. The hallmarks of AD include extracellular amyloid deposition, intracellular neuronal fiber entanglement, and neuronal loss. Despite strenuous efforts toward improvement of AD, there remains a lack of effective treatment and current pharmaceutical therapies only alleviate the symptoms for a short period of time. Interestingly, some progress has been achieved in treatment of AD based on mesenchymal stem cell (MSC) transplantation in recent years. MSC transplantation, as a rising therapy, is used as an intervention in AD, because of the enormous potential of MSCs, including differentiation potency, immunoregulatory function, and no immunological rejection. Although numerous strategies have focused on the use of MSCs to replace apoptotic or degenerating neurons, recent studies have implied that MSC-immunoregulation, which modulates the activity state of microglia or astrocytes and mediates neuroinflammation via several transcription factors (NFs) signaling pathways, may act as a major mechanism for the therapeutic efficacy of MSC and be responsible for some of the satisfactory results. In this review, we will focus on the role of MSC-immunoregulation in MSC-based therapy for AD.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, China.
| | - Zhi-Fang Dong
- Department of Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, China.
| | - Jie-Yuan Zhang
- Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
32
|
Sung K, Jimenez-Sanchez M. Autophagy in Astrocytes and its Implications in Neurodegeneration. J Mol Biol 2020; 432:2605-2621. [PMID: 31931011 DOI: 10.1016/j.jmb.2019.12.041] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Autophagy is a major degradation pathway where double-membrane vesicles called autophagosomes deliver cytoplasmic content to the lysosome. Increasing evidence suggests that autophagy dysfunction contributes to the pathogenesis of neurodegenerative diseases. In addition, misfolded proteins that accumulate in these diseases and constitute a common pathological hallmark are substrates for autophagic degradation. Astrocytes, a major type of glial cells, are emerging as a critical component in most neurodegenerative diseases. This review will summarize the recent efforts to investigate the role that autophagy plays in astrocytes in the context of neurodegenerative diseases. While the field has mostly focused on the implications of autophagy in neurons, autophagy may also be involved in the clearance of disease-related proteins in astrocytes as well as in maintaining astrocyte function, which could impact the cell autonomous and non-cell autonomous contribution of astrocytes to neurodegeneration.
Collapse
Affiliation(s)
- Katherine Sung
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Maria Jimenez-Sanchez
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RX, UK.
| |
Collapse
|
33
|
Croft CL, Futch HS, Moore BD, Golde TE. Organotypic brain slice cultures to model neurodegenerative proteinopathies. Mol Neurodegener 2019; 14:45. [PMID: 31791377 PMCID: PMC6889333 DOI: 10.1186/s13024-019-0346-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/13/2019] [Indexed: 01/30/2023] Open
Abstract
Organotypic slice cultures of brain or spinal cord have been a longstanding tool in neuroscience research but their utility for understanding Alzheimer's disease (AD) and other neurodegenerative proteinopathies has only recently begun to be evaluated. Organotypic brain slice cultures (BSCs) represent a physiologically relevant three-dimensional model of the brain. BSCs support all the central nervous system (CNS) cell types and can be produced from brain areas involved in neurodegenerative disease. BSCs can be used to better understand the induction and significance of proteinopathies underlying the development and progression of AD and other neurodegenerative disorders, and in the future may serve as bridging technologies between cell culture and in vivo experiments for the development and evaluation of novel therapeutic targets and strategies. We review the initial development and general use of BSCs in neuroscience research and highlight the advantages of these cultures as an ex vivo model. Subsequently we focus on i) BSC-based modeling of AD and other neurodegenerative proteinopathies ii) use of BSCs to understand mechanisms underlying these diseases and iii) how BSCs can serve as tools to screen for suitable therapeutics prior to in vivo investigations. Finally, we will examine i) open questions regarding the use of such cultures and ii) how emerging technologies such as recombinant adeno-associated viruses (rAAV) may be combined with these models to advance translational research relevant to neurodegenerative disorders.
Collapse
Affiliation(s)
- C L Croft
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - H S Futch
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - B D Moore
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - T E Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA. .,Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA. .,McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
34
|
Bellozi PMQ, Gomes GF, da Silva MCM, Lima IVDA, Batista CRÁ, Carneiro Junior WDO, Dória JG, Vieira ÉLM, Vieira RP, de Freitas RP, Ferreira CN, Candelario-Jalil E, Wyss-Coray T, Ribeiro FM, de Oliveira ACP. A positive allosteric modulator of mGluR5 promotes neuroprotective effects in mouse models of Alzheimer's disease. Neuropharmacology 2019; 160:107785. [PMID: 31541651 DOI: 10.1016/j.neuropharm.2019.107785] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/06/2019] [Accepted: 09/17/2019] [Indexed: 12/26/2022]
Abstract
Alzheimer's Disease (AD) is the most prevalent neurodegenerative disorder. Despite advances in the understanding of its pathophysiology, none of the available therapies prevents disease progression. Excess glutamate plays an important role in excitotoxicity by activating ionotropic receptors. However, the mechanisms modulating neuronal cell survival/death via metabotropic glutamate receptors (mGluRs) are not completely understood. Recent data indicates that CDPPB, a positive allosteric modulator of mGluR5, has neuroprotective effects. Thus, this work aimed to investigate CDPPB treatment effects on amyloid-β (Aβ) induced pathological alterations in vitro and in vivo and in a transgenic mouse model of AD (T41 mice). Aβ induced cell death in primary cultures of hippocampal neurons, which was prevented by CDPPB. Male C57BL/6 mice underwent stereotaxic surgery for unilateral intra-hippocampal Aβ injection, which induced memory deficits, neurodegeneration, neuronal viability reduction and decrease of doublecortin-positive cells, a marker of immature neurons and neuronal proliferation. Treatment with CDPPB for 8 days reversed neurodegeneration and doublecortin-positive cells loss and recovered memory function. Fourteen months old T41 mice presented cognitive deficits, neuronal viability reduction, gliosis and Aβ accumulation. Treatment with CDPPB for 28 days increased neuronal viability (32.2% increase in NeuN+ cells) and reduced gliosis in CA1 region (Iba-1+ area by 31.3% and GFAP+ area by 37.5%) in transgenic animals, without inducing hepatotoxicity. However, it did not reverse cognitive deficit. Despite a four-week treatment did not prevent memory loss in aged transgenic mice, CDPPB is protective against Aβ stimulus. Therefore, this drug represents a potential candidate for further investigations as AD treatment.
Collapse
Affiliation(s)
| | - Giovanni Freitas Gomes
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | | | | | | | - Juliana Guimarães Dória
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Rafael Pinto Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Claudia Natália Ferreira
- Clinical Pathology Sector of COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | |
Collapse
|
35
|
Guzman-Martinez L, Maccioni RB, Andrade V, Navarrete LP, Pastor MG, Ramos-Escobar N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front Pharmacol 2019; 10:1008. [PMID: 31572186 PMCID: PMC6751310 DOI: 10.3389/fphar.2019.01008] [Citation(s) in RCA: 436] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/08/2019] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative diseases share the fact that they derive from altered proteins that undergo an unfolding process followed by formation of β-structures and a pathological tendency to self-aggregate in neuronal cells. This is a characteristic of tau protein in Alzheimer’s disease and several tauopathies associated with tau unfolding, α-synuclein in Parkinson’s disease, and huntingtin in Huntington disease. Usually, the self-aggregation products are toxic to these cells, and toxicity spreads all over different brain areas. We have postulated that these protein unfolding events are the molecular alterations that trigger several neurodegenerative disorders. Most interestingly, these events occur as a result of neuroinflammatory cascades involving alterations in the cross-talks between glial cells and neurons as a consequence of the activation of microglia and astrocytes. The model we have hypothesized for Alzheimer’s disease involves damage signals that promote glial activation, followed by nuclear factor NF-kβ activation, synthesis, and release of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1, IL-6, and IL-12 that affect neuronal receptors with an overactivation of protein kinases. These patterns of pathological events can be applied to several neurodegenerative disorders. In this context, the involvement of innate immunity seems to be a major paradigm in the pathogenesis of these diseases. This is an important element for the search for potential therapeutic approaches for all these brain disorders.
Collapse
Affiliation(s)
- Leonardo Guzman-Martinez
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Ricardo B Maccioni
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Andrade
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - Leonardo Patricio Navarrete
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| | - María Gabriela Pastor
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile.,Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Nicolas Ramos-Escobar
- Laboratory of Neuroscience, Faculty of Sciences, University of Chile & International Center for Biomedicine (ICC), Santiago, Chile
| |
Collapse
|
36
|
Liu PP, Xie Y, Meng XY, Kang JS. History and progress of hypotheses and clinical trials for Alzheimer's disease. Signal Transduct Target Ther 2019; 4:29. [PMID: 31637009 PMCID: PMC6799833 DOI: 10.1038/s41392-019-0063-8] [Citation(s) in RCA: 346] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/07/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive memory loss along with neuropsychiatric symptoms and a decline in activities of daily life. Its main pathological features are cerebral atrophy, amyloid plaques, and neurofibrillary tangles in the brains of patients. There are various descriptive hypotheses regarding the causes of AD, including the cholinergic hypothesis, amyloid hypothesis, tau propagation hypothesis, mitochondrial cascade hypothesis, calcium homeostasis hypothesis, neurovascular hypothesis, inflammatory hypothesis, metal ion hypothesis, and lymphatic system hypothesis. However, the ultimate etiology of AD remains obscure. In this review, we discuss the main hypotheses of AD and related clinical trials. Wealthy puzzles and lessons have made it possible to develop explanatory theories and identify potential strategies for therapeutic interventions for AD. The combination of hypometabolism and autophagy deficiency is likely to be a causative factor for AD. We further propose that fluoxetine, a selective serotonin reuptake inhibitor, has the potential to treat AD.
Collapse
Affiliation(s)
- Pei-Pei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yi Xie
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xiao-Yan Meng
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jian-Sheng Kang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
37
|
Li K, Li J, Zheng J, Qin S. Reactive Astrocytes in Neurodegenerative Diseases. Aging Dis 2019; 10:664-675. [PMID: 31165009 PMCID: PMC6538217 DOI: 10.14336/ad.2018.0720] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022] Open
Abstract
Astrocytes, the largest and most numerous glial cells in the central nervous system (CNS), play a variety of important roles in regulating homeostasis, increasing synaptic plasticity and providing neuroprotection, thus helping to maintain normal brain function. At the same time, astrocytes can participate in the inflammatory response and play a key role in the progression of neurodegenerative diseases. Reactive astrocytes are strongly induced by numerous pathological conditions in the CNS. Astrocyte reactivity is initially characterized by hypertrophy of soma and processes, triggered by different molecules. Recent studies have demonstrated that neuroinflammation and ischemia can elicit two different types of reactive astrocytes, termed A1s and A2s. However, in the case of astrocyte reactivity in different neurodegenerative diseases, the recently published research issues remain a high level of conflict and controversy. So far, we still know very little about whether and how the function or reactivity of astrocytes changes in the progression of different neurodegenerative diseases. In this review, we aimed to briefly discuss recent studies highlighting the complex contribution of astrocytes in the process of various neurodegenerative diseases, which may provide us with new prospects for the development of an excellent therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Kunyu Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jiatong Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jialin Zheng
- 2Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Song Qin
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
38
|
Tsoy A, Saliev T, Abzhanova E, Turgambayeva A, Kaiyrlykyzy A, Akishev M, Saparbayev S, Umbayev B, Askarova S. The Effects of Mobile Phone Radiofrequency Electromagnetic Fields on β-Amyloid-Induced Oxidative Stress in Human and Rat Primary Astrocytes. Neuroscience 2019; 408:46-57. [PMID: 30953670 DOI: 10.1016/j.neuroscience.2019.03.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 12/25/2022]
Abstract
Amyloid beta peptide (Aβ) is implicated in the development of pathological reactions associated with Alzheimer's disease (AD), such as oxidative stress, neuro-inflammation and death of brain cells. Current pharmacological approaches to treat AD are not able to control the deposition of Aβ and suppression of Aβ-induced cellular response. There is a growing body of evidence that exposure to radiofrequency electromagnetic field (RF-EMF) causes a decrease of beta-amyloid deposition in the brains and provides cognitive benefits to Alzheimer's Tg mice. Herein, we investigated the effects of mobile phone radiofrequency EMF of 918 MHz on reactive oxygen species (ROS) formation, mitochondrial membrane potential (MMP), activity of NADPH-oxidase, and phosphorylation of p38MAPK and ERK1/2 kinases in human and rat primary astrocytes in the presence of Aβ42 and H2O2. Our data demonstrate that EMF is able to reduce Aβ42- and H2O2-induced cellular ROS, abrogate Aβ₄₂-induced production of mitochondrial ROS and the co-localization between the cytosolic (p47-phox) and membrane (gp91-phox) subunits of NADPH oxidase, while increasing MMP, and inhibiting H2O2-induced phosphorylation of p38MAPK and ERK1/2 in primary astrocytes. Yet, EMF was not able to modulate alterations in the phosphorylation state of the MAPKs triggered by Aβ42. Our findings provide an insight into the mechanisms of cellular and molecular responses of astrocytes on RF-EMF exposure and indicate the therapeutic potential of RF-EMF for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Andrey Tsoy
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Timur Saliev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan; S.D. Asfendiyarov Kazakh National Medical University, Tole Bi Street 94, Almaty, 050000, Kazakhstan
| | - Elvira Abzhanova
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Anel Turgambayeva
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Aiym Kaiyrlykyzy
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Mars Akishev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Samat Saparbayev
- National Scientific Medical Center, 42 Abylai Khan Ave, Astana, 010000, Kazakhstan, 010009
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan
| | - Sholpan Askarova
- National Laboratory Astana, Nazarbayev University, 53 Kabanbay batyr Ave., Astana, 010000, Kazakhstan.
| |
Collapse
|
39
|
Gatt A, Lee H, Williams G, Thuret S, Ballard C. Expression of neurogenic markers in Alzheimer's disease: a systematic review and metatranscriptional analysis. Neurobiol Aging 2019; 76:166-180. [PMID: 30716542 DOI: 10.1016/j.neurobiolaging.2018.12.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by substantial neuronal loss and progressive brain atrophy. Animal studies have suggested that the process of adult neurogenesis might be altered at the earliest phases of disease onset. The relationship between AD progression and adult neurogenesis in the human brain is, however, not well understood. Here, we present a systematic review of the postmortem studies that investigated changes in human adult neurogenesis in the AD brain. We present findings from 11 postmortem studies that were identified by a systematic search within the literature, focusing on what markers of neurogenesis were used, which stages of AD were investigated, and whether the studies had any confounding information that could potentially hinder clear interpretation of the presented data. In addition, we also review studies that examined transcriptomic changes in human AD postmortem brains and reveal upregulated expression of neural progenitor and proliferation markers and downregulated expression of later neurogenic markers in AD. Taken together, the existing literature seems to suggest that the overall level of human adult neurogenesis is reduced during the later stages of AD, potentially due to failed maturation and integration of new-born neurons. Further investigations using complementary methods such as in vitro disease modeling will be helpful to understand the exact molecular mechanisms underlying such pattern of change and to determine whether neurogenesis can be an effective therapeutic target for early intervention.
Collapse
Affiliation(s)
- Ariana Gatt
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Hyunah Lee
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gareth Williams
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sandrine Thuret
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Clive Ballard
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
40
|
Chou CM, Fan CK. Significant apoptosis rather autophagy predominates in astrocytes caused by Toxocara canis larval excretory-secretory antigens. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2018; 53:250-258. [PMID: 30033092 DOI: 10.1016/j.jmii.2018.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND/PURPOSE Toxocariasis is a worldwide parasitic zoonosis and mainly caused by Toxocara canis. Humans can be infected by accidental ingestion of T. canis embryonated ova through contacting with contaminated food, water, or encapsulated larvae in paratenic hosts' viscera or meat. Since humans are the paratenic host of T. canis, the wandering and neuroinvasive larvae can cause mechanical tissue damage and the excretory-secretory antigens (TcES Ag) might induce neuroinflammatory responses in the brain. Human cerebral toxocariasis (CT) has been reported to cause several neurological symptoms and may develop into neurodegenerative diseases. However, the roles of astrocytes involved in the pathogenesis of CT remained largely unclear. METHODS This study intended to investigate the cytotoxic effects of TcES Ag on astrocytes as assessed by apoptosis and autophagy expression. RESULTS Our results showed TcES Ag treatment reduced cell viability and caused morphological changes. Expressions of autophagy associated proteins including Beclin 1, phosphor-mTOR and LC3-Ⅱ were not significantly changed; however, p62 as well as the cell survival protein, mTOR, was concomitantly decreased in TcES Ag treatment. Significantly accelerated cleaved caspase-3 and cytochrome c expression as well as enhanced caspase-9 and caspase-8 activation were found in astrocytes with TcES Ag treatment. Caspase-3 activity and apoptotic cells numbers were also increased as detected by fluorescence microscopy. CONCLUSION We concluded that TcES Ag may trigger astrocytes apoptosis predominantly through intrinsic and extrinsic pathways rather autophagy, revealing a novel role of astrocytes in the pathogenesis of CT.
Collapse
Affiliation(s)
- Chia-Mei Chou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan; Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan
| | - Chia-Kwung Fan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan; Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan; Research Center of International Tropical Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan; Tropical Medicine Division, International PhD Program in Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan.
| |
Collapse
|
41
|
Specific deletion connexin43 in astrocyte ameliorates cognitive dysfunction in APP/PS1 mice. Life Sci 2018; 208:175-191. [PMID: 30031059 DOI: 10.1016/j.lfs.2018.07.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/15/2018] [Accepted: 07/18/2018] [Indexed: 11/22/2022]
Abstract
Emerging data indicate an important role for connexin43 (Cx43) in cognitive function, but there is a lack of direct evidence of the role of astroglial Cx43 in cognitive dysfunction in Alzheimer's disease (AD). Here we evaluated the expression pattern of Cx43 in AD and found progressive upregulation of the mRNA and protein levels of Cx43. Subsequently, we generated an astroglial Cx43 knockout (KO) AD mouse model by crossbreeding Gfap (glial fibrillary acidic protein)-Cx43 KO mice with APP/PS1 mice. Then we assessed the cognitive function of 12-month-old APP (amyloid precursor protein)/PS1 (presenilin 1)/Gfap-Cx43 KO mice, which demonstrated that the deletion of astroglial Cx43 significantly ameliorated cognitive dysfunction. To further investigate the underlying mechanisms, we evaluated amyloid plaque formation, astrogliosis, and synaptic function. The number and area of amyloid plaques were not altered, but GFAP expression was significantly decreased and the number of synapses was markedly upregulated. These results suggest that deletion of astroglial Cx43 in APP/PS1 mice did not affect the formation of amyloid plaques but depressed astrogliosis and upregulated synaptic function. Moreover, levels of critical modulators of astroglial activation were also notably reduced, but those of pro- and anti-inflammatory cytokines were not altered. Furthermore, Cx43 regulation of postsynaptic elements targets mainly NMDAR (N-methyl-d-aspartate). In addition, the prevention of AD pathology was reversed by Cx43 re-expression. In sum, specific deletion of astroglial Cx43 in APP/PS1 mice improved cognitive dysfunction by decreasing astrogliosis and increasing synaptic function without affecting amyloid plaque formation or the inflammatory response.
Collapse
|
42
|
Edison P, Brooks DJ. Role of Neuroinflammation in the Trajectory of Alzheimer’s Disease and in vivo Quantification Using PET. J Alzheimers Dis 2018; 64:S339-S351. [DOI: 10.3233/jad-179929] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Paul Edison
- Neurology Imaging Unit, Department of Medicine, Imperial College London, London, UK
| | - David J. Brooks
- Department of Nuclear Medicine, Aarhus University, Denmark
- Institute of Neuroscience, University of Newcastle upon Tyne, UK
| |
Collapse
|
43
|
He M, Gu J, Zhu J, Wang X, Wang C, Duan C, Ni Y, Lu X, Li J. Up-regulation of Dyrk1b promote astrocyte activation following lipopolysaccharide-induced neuroinflammation. Neuropeptides 2018; 69:76-83. [PMID: 29751999 DOI: 10.1016/j.npep.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 02/11/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023]
Abstract
Astrocytes become activated in response to different stimulation. Dyrk1b is an arginine-directed serine/threonineprotein kinase that is expressed at elevated levels in many cancers but remains unknown in the pathologies of neuroinflammation. In this study, in vivo, we demonstrated that Dyrk1b expression was significantly increased and reached a peak at 12 h after LPS injection via Western blot. Double immunofluorescence staining showed that Dyrk1b co-located with GFAP and Ki67. In vitro, the expression of Dyrk1b, Ki67 and cyclinD1 was gradually increased and reached a peak at 12 h in a time-dependent manner after 1 μg/mL LPS stimulation. Knockdown of Dyrk1b significantly reduced the expression of Ki67 and cyclinD1. In addition, the data exhibited that silenced Dyrk1b decreased the expression of p-STAT3 in primary astrocyte cells, and Dyrk1b interacted with STAT3 in LPS-induced neuroinflammation. In conclusion, these results suggested that Dyrk1b is increased and may play a crucial role in regulating astrocyte cell activation via interact with STAT3 in LPS-induced neuroinflammation.
Collapse
Affiliation(s)
- Mingqing He
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, Jiangsu, China
| | - Jun Gu
- Department of Orthopaedics, XiShan People's Hospital, Wuxi 214011, Jiangsu, China
| | - Jinzhou Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Xiaoyan Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Chengniu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong 226001,Jiangsu, China
| | - Chengwei Duan
- The Second People's Hospital of Nantong, Nantong 226002, Jiangsu, China
| | - Yingjie Ni
- Department of Orthopaedics, XiShan People's Hospital, Wuxi 214011, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, The Affiliated Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Key Laboratory for Aging & Disease, Nanjing Medical University, Nanjing 210011, Jiangsu, China.
| | - Jianzhong Li
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
44
|
Extract of Fructus Schisandrae chinensis Inhibits Neuroinflammation Mediator Production from Microglia via NF-κ B and MAPK Pathways. Chin J Integr Med 2018; 25:131-138. [PMID: 29790065 DOI: 10.1007/s11655-018-3001-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 10/16/2022]
Abstract
OBJECTIVE To investigate the anti-neuroinflammation effect of extract of Fructus Schisandrae chinensis (EFSC) on lipopolysaccharide (LPS)-induced BV-2 cells and the possible involved mechanisms. METHODS Primary cortical neurons were isolated from embryonic (E17-18) cortices of Institute of Cancer Research (ICR) mouse fetuses. Primary microglia and astroglia were isolated from the frontal cortices of newborn ICR mouse. Different cells were cultured in specific culture medium. Cells were divided into 5 groups: control group, LPS group (treated with 1 μg/mL LPS only) and EFSC groups (treated with 1 μg/mL LPS and 100, 200 or 400 mg/mL EFSC, respectively). The effect of EFSC on cells viability was tested by methylthiazolyldiphenyltetrazolium bromide (MTT) colorimetric assay. EFSC-mediated inhibition of LPS-induced production of pro-inflammatory mediators, such as nitrite oxide (NO) and interleukin-6 (IL-6) were quantified and neuron-protection effect against microglia-mediated inflammation injury was tested by hoechst 33258 apoptosis assay and crystal violet staining assay. The expression of pro-inflammatory marker proteins was evaluated by Western blot analysis or immunofluorescence. RESULTS EFSC (200 and 400 mg/mL) reduced NO, IL-6, inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) expression in LPS-induced BV-2 cells (P<0.01 or P<0.05). EFSC (200 and 400 mg/mL) reduced the expression of NO in LPS-induced primary microglia and astroglia (P<0.01). In addition, EFSC alleviated cell apoptosis and inflammation injury in neurons exposed to microglia-conditioned medium (P<0.01). The mechanistic studies indicated EFSC could suppress nuclear factor (NF)-?B phosphorylation and its nuclear translocation (P<0.01). The anti-inflammatory effect of EFSC occurred through suppressed activation of mitogen-activated protein kinase (MAPK) pathway (P<0.01 or P<0.05). CONCLUSION EFSC acted as an anti-inflammatory agent in LPS-induced glia cells. These effects might be realized through blocking of NF-κB activity and inhibition of MAPK signaling pathways.
Collapse
|
45
|
Garwood CJ, Ratcliffe LE, Simpson JE, Heath PR, Ince PG, Wharton SB. Review: Astrocytes in Alzheimer's disease and other age-associated dementias: a supporting player with a central role. Neuropathol Appl Neurobiol 2018; 43:281-298. [PMID: 27442752 DOI: 10.1111/nan.12338] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 07/15/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
Astrocytes have essential roles in the central nervous system and are also implicated in the pathogenesis of neurodegenerative disease. Forming non-overlapping domains, astrocytes are highly complex cells. Immunohistochemistry to a variety of proteins can be used to study astrocytes in tissue, labelling different cellular components and sub-populations, including glial fibrillary acidic protein, ALDH1L1, CD44, NDRG2 and amino acid transporters, but none of these labels the entire astrocyte population. Increasing heterogeneity is recognized in the astrocyte population, a complexity that is relevant both to their normal function and pathogenic roles. They are involved in neuronal support, as active components of the tripartite synapse and in cell interactions within the neurovascular unit (NVU), where they are essential for blood-brain barrier maintenance and neurovascular coupling. Astrocytes change with age, and their responses may modulate the cellular effects of neurodegenerative pathologies, which alone do not explain all of the variance in statistical models of neurodegenerative dementias. Astrocytes respond to both the neurofibrillary tangles and plaques of Alzheimer's disease, to hyperphosphorylated tau and Aβ, eliciting an effect which may be neuroprotective or deleterious. Not only astrocyte hypertrophy, in the form of gliosis, occurs, but also astrocyte injury and atrophy. Loss of normal astrocyte functions may contribute to reduced support for neurones and dysfunction of the NVU. Understanding how astrocytes contribute to dementia requires an understanding of the underlying heterogeneity of astrocyte populations, and the complexity of their responses to pathology. Enhancing the supportive and neuroprotective components of the astrocyte response has potential translational applications in therapeutic approaches to dementia.
Collapse
Affiliation(s)
- C J Garwood
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - L E Ratcliffe
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - J E Simpson
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - P R Heath
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - P G Ince
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | - S B Wharton
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| |
Collapse
|
46
|
Zhou H, Qu Z, Zhang J, Jiang B, Liu C, Gao W. Shunaoxin dropping pill, a Chinese herb compound preparation, attenuates memory impairment in d-galactose-induced aging mice. RSC Adv 2018; 8:10163-10171. [PMID: 35540463 PMCID: PMC9078930 DOI: 10.1039/c7ra13726e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/19/2018] [Indexed: 11/30/2022] Open
Abstract
The Shunaoxin dropping pill (SNX) is derived from a traditional recipe. It has been used to treat cerebrovascular diseases in China since 2005 (approval number Z20050041). In this study, we used an in vitro H2O2-induced PC12 cell oxidative damage model and an in vivod-gal-induced mouse memory impairment model to investigate whether SNX had neuroprotective effects. In vitro, prior to exposure to 100 μM H2O2 for 2 h, PC12 cells were pre-treated with SNX 50 μg mL-1 for 24 h. Hoechst 33258 staining was used to confirm the effect of SNX on apoptosis in the PC12 cells. Our results demonstrate that H2O2 suppresses the proliferation of PC12 cells and induces cell death. Pretreatment with SNX attenuates H2O2-induced apoptosis in PC12 cells. In vivo, d-gal was administered (100 mg kg-1, subcutaneously (s.c.)) once daily for 8 weeks to induce memory deficit and neurotoxicity in the brain of an aging mouse. Then, SNX (320 mg kg-1) was simultaneously administered orally. The present study demonstrates that SNX can alleviate aging in the mouse brain induced by d-gal via improving behavioral performance, alleviating oxidative stress, inhibiting neuroinflammation, and reducing brain cell damage in the hippocampus. Overall, these data clearly demonstrate the neuroprotective effect of SNX from the in vitro and in vivo results. SNX may be considered a novel agent for easing aging and/or age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Hong Zhou
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China pharmgao@tju,edu.cn +86-22-87401895 +86-22-87401895
- No. 6 Traditional Chinese Medicine Factory, Tianjin Zhongxin Pharmaceutical Group Corporation Ltd. Tianjin 300401 China
| | - Zhuo Qu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China pharmgao@tju,edu.cn +86-22-87401895 +86-22-87401895
| | - Jingze Zhang
- Department of Pharmacy, Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Logistics University of Chinese People's Armed Police Forces Tianjin 300162 China
| | - Bingjie Jiang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China pharmgao@tju,edu.cn +86-22-87401895 +86-22-87401895
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics Tianjin 300193 China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University Weijin Road Tianjin 300072 China pharmgao@tju,edu.cn +86-22-87401895 +86-22-87401895
| |
Collapse
|
47
|
Wang S, Zhang X, Zhai L, Sheng X, Zheng W, Chu H, Zhang G. Atorvastatin Attenuates Cognitive Deficits and Neuroinflammation Induced by Aβ 1-42 Involving Modulation of TLR4/TRAF6/NF-κB Pathway. J Mol Neurosci 2018; 64:363-373. [PMID: 29417448 DOI: 10.1007/s12031-018-1032-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022]
Abstract
Inflammatory damage aggravates the progression of Alzheimer's disease (AD) and the mechanism of inflammatory damage may provide a new therapeutic window for the treatment of AD. Toll-like receptor 4 (TLR4)-mediated signaling can regulate the inflammatory process. However, changes in TLR4 signaling pathway induced by beta-amyloid (Aβ) have not been well characterized in brain, especially in the hippocampus. In the present study, we explored the changes of TLR4 signaling pathway induced by Aβ in the hippocampus and the role of atorvastatin in modulating this signal pathway and neurotoxicity induced by Aβ. Experimental AD rats were induced by intrahippocampal injection of Aβ1-42, and the rats were treated with atorvastatin by oral gavage from 3 weeks before to 6 days after injections of Aβ1-42. To determine the spatial learning and memory ability of rats in the AD models, Morris water maze (MWM) was performed. The expression of the glial fibrillary acidic protein (GFAP), ionized calcium binding adapter molecule-1 (Iba-1), TLR4, tumor necrosis factor receptor-associated factor 6 (TRAF6), and nuclear transcription factor (NF)-κB (NF-κB) protein in the hippocampus was detected by immunohistochemistry and Western blot. Compared to the control group, increased expression of TLR4, TRAF6, and NF-κB was observed in the hippocampus at 7 days post-injection of Aβ (P < 0.01). Furthermore, atorvastatin treatment significantly ameliorated cognitive deficits of rats, attenuated microglia and astrocyte activation, inhibited apoptosis, and down-regulated the expression of TLR4, TRAF6, and NF-κB, both at the mRNA and protein levels (P < 0.01). TLR4 signaling pathway is thus actively involved in Aβ-induced neuroinflammation and atorvastatin treatment can exert the therapeutic benefits for AD via the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Shan Wang
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China
| | - Xiaowei Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Liuyu Zhai
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China
| | - Xiaona Sheng
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China.
| | - Weina Zheng
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China
| | - Hongshan Chu
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China
| | - Guohua Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, No. 215 Hepingxi Road, Shijiazhuang, Hebei, 050000, China
| |
Collapse
|
48
|
Abstract
Extracellular exosomes are formed inside the cytoplasm of cells in compartments known as multivesicular bodies. Thus, exosomes contain cytoplasmic content. Multivesicular bodies fuse with the plasma membrane and release exosomes into the extracellular environment. Comprehensive research suggests that exosomes act as both inflammatory intermediaries and critical inducers of oxidative stress to drive progression of Alzheimer's disease. An important role of exosomes in Alzheimer's disease includes the formation of neurofibrillary tangles and beta-amyloid production, clearance, and accumulation. In addition, exosomes are involved in neuroinflammation and oxidative stress, which both act as triggers for beta-amyloid pathogenesis and tau hyperphosphorylation. Further, it has been shown that exosomes are strongly associated with beta-amyloid clearance. Thus, effective measures for regulating exosome metabolism may be novel drug targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Zhi-You Cai
- Department of Neurology, Chongqing General Hospital, Chongqing, China
| | - Ming Xiao
- Department of Anatomy, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Sohel H Quazi
- Department of Biological and Health Sciences, Texas A & M University-Kingsville, Kingsville, TX, USA
| | - Zun-Yu Ke
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
49
|
Ferrer I. Diversity of astroglial responses across human neurodegenerative disorders and brain aging. Brain Pathol 2017; 27:645-674. [PMID: 28804999 PMCID: PMC8029391 DOI: 10.1111/bpa.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022] Open
Abstract
Astrogliopathy refers to alterations of astrocytes occurring in diseases of the nervous system, and it implies the involvement of astrocytes as key elements in the pathogenesis and pathology of diseases and injuries of the central nervous system. Reactive astrocytosis refers to the response of astrocytes to different insults to the nervous system, whereas astrocytopathy indicates hypertrophy, atrophy/degeneration and loss of function and pathological remodeling occurring as a primary cause of a disease or as a factor contributing to the development and progression of a particular disease. Reactive astrocytosis secondary to neuron loss and astrocytopathy due to intrinsic alterations of astrocytes occur in neurodegenerative diseases, overlap each other, and, together with astrocyte senescence, contribute to disease-specific astrogliopathy in aging and neurodegenerative diseases with abnormal protein aggregates in old age. In addition to the well-known increase in glial fibrillary acidic protein and other proteins in reactive astrocytes, astrocytopathy is evidenced by deposition of abnormal proteins such as β-amyloid, hyper-phosphorylated tau, abnormal α-synuclein, mutated huntingtin, phosphorylated TDP-43 and mutated SOD1, and PrPres , in Alzheimer's disease, tauopathies, Lewy body diseases, Huntington's disease, amyotrophic lateral sclerosis and Creutzfeldt-Jakob disease, respectively. Astrocytopathy in these diseases can also be manifested by impaired glutamate transport; abnormal metabolism and release of neurotransmitters; altered potassium, calcium and water channels resulting in abnormal ion and water homeostasis; abnormal glucose metabolism; abnormal lipid and, particularly, cholesterol metabolism; increased oxidative damage and altered oxidative stress responses; increased production of cytokines and mediators of the inflammatory response; altered expression of connexins with deterioration of cell-to-cell networks and transfer of gliotransmitters; and worsening function of the blood brain barrier, among others. Increased knowledge of these aspects will permit a better understanding of brain aging and neurodegenerative diseases in old age as complex disorders in which neurons are not the only players.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental TherapeuticsUniversity of BarcelonaBarcelonaSpain
- Institute of NeuropathologyPathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de LlobregatBarcelonaSpain
- Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos IIIMadridSpain
| |
Collapse
|
50
|
Inhibition of glycogen synthase kinase-3 by BTA-EG 4 reduces tau abnormalities in an organotypic brain slice culture model of Alzheimer's disease. Sci Rep 2017; 7:7434. [PMID: 28785087 PMCID: PMC5547074 DOI: 10.1038/s41598-017-07906-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/29/2017] [Indexed: 11/08/2022] Open
Abstract
Organotypic brain slice culture models provide an alternative to early stage in vivo studies as an integrated tissue system that can recapitulate key disease features, thereby providing an excellent platform for drug screening. We recently described a novel organotypic 3xTg-AD mouse brain slice culture model with key Alzheimer’s disease-like changes. We now highlight the potential of this model for testing disease-modifying agents and show that results obtained following in vivo treatment are replicated in brain slice cultures from 3xTg-AD mice. Moreover, we describe novel effects of the amyloid-binding tetra (ethylene glycol) derivative of benzothiazole aniline, BTA-EG4, on tau. BTA-EG4 significantly reduced tau phosphorylation in the absence of any changes in the amounts of amyloid precursor protein, amyloid-β or synaptic proteins. The reduction in tau phosphorylation was associated with inactivation of the Alzheimer’s disease-relevant major tau kinase, GSK-3. These findings highlight the utility of 3xTg-AD brain slice cultures as a rapid and reliable in vitro method for drug screening prior to in vivo testing. Furthermore, we demonstrate novel tau-directed effects of BTA-EG4 that are likely related to the ability of this agent to inactivate GSK-3. Our findings support the further exploration of BTA-EG4 as a candidate therapeutic for Alzheimer’s disease.
Collapse
|