1
|
Power SK, Venkatesan S, Qu S, McLaurin J, Lambe EK. Enhanced prefrontal nicotinic signaling as evidence of active compensation in Alzheimer's disease models. Transl Neurodegener 2024; 13:58. [PMID: 39623428 PMCID: PMC11613856 DOI: 10.1186/s40035-024-00452-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 08/22/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Cognitive reserve allows for resilience to neuropathology, potentially through active compensation. Here, we examine ex vivo electrophysiological evidence for active compensation in Alzheimer's disease (AD) focusing on the cholinergic innervation of layer 6 in prefrontal cortex. Cholinergic pathways are vulnerable to neuropathology in AD and its preclinical models, and their modulation of deep layer prefrontal cortex is essential for attention and executive function. METHODS We functionally interrogated cholinergic modulation of prefrontal layer 6 pyramidal neurons in two preclinical models: a compound transgenic AD mouse model that permits optogenetically-triggered release of endogenous acetylcholine and a transgenic AD rat model that closely recapitulates the human trajectory of AD. We then tested the impact of therapeutic interventions to further amplify the compensated responses and preserve the typical kinetic profile of cholinergic signaling. RESULTS In two AD models, we found potentially compensatory upregulation of functional cholinergic responses above non-transgenic controls after onset of pathology. To identify the locus of this enhanced cholinergic signal, we dissected key pre- and post-synaptic components with pharmacological strategies. We identified a significant and selective increase in post-synaptic nicotinic receptor signalling on prefrontal cortical neurons. To probe the additional impact of therapeutic intervention on the adapted circuit, we tested cholinergic and nicotinic-selective pro-cognitive treatments. Inhibition of acetylcholinesterase further enhanced endogenous cholinergic responses but greatly distorted their kinetics. Positive allosteric modulation of nicotinic receptors, by contrast, enhanced endogenous cholinergic responses and retained their rapid kinetics. CONCLUSIONS We demonstrate that functional nicotinic upregulation occurs within the prefrontal cortex in two AD models. Promisingly, this nicotinic signal can be further enhanced while preserving its rapid kinetic signature. Taken together, our work suggests that compensatory mechanisms are active within the prefrontal cortex that can be harnessed by nicotinic receptor positive allosteric modulation, highlighting a new direction for cognitive treatment in AD neuropathology.
Collapse
Affiliation(s)
- Saige K Power
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sridevi Venkatesan
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sarah Qu
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - JoAnne McLaurin
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Evelyn K Lambe
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Obstetrics and Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5G 1E2, Canada.
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, M5T 1R8, Canada.
| |
Collapse
|
2
|
Park S, Shin J, Kim K, Kim D, Lee WS, Lee J, Cho I, Park IW, Yoon S, Lee S, Kim HY, Lee JH, Hong KB, Kim Y. Modulation of Amyloid and Tau Aggregation to Alleviate Cognitive Impairment in a Transgenic Mouse Model of Alzheimer's Disease. ACS Pharmacol Transl Sci 2024; 7:2650-2661. [PMID: 39296253 PMCID: PMC11406698 DOI: 10.1021/acsptsci.4c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 09/21/2024]
Abstract
Aggregation of misfolded amyloid-β (Aβ) and hyperphosphorylated tau proteins to plaques and tangles, respectively, is the major drug target of Alzheimer's disease (AD), as the former is an onset biomarker and the latter is associated with neurodegeneration. Thus, we report a small molecule drug candidate, DN5355, with a dual-targeting function toward aggregates of both Aβ and tau. DN5355 was selected through a series of four screenings assessing 52 chemicals for their functions to inhibit and reverse the aggregation of Aβ and tau by utilizing thioflavin T. When orally administered to AD transgenic mouse model 5XFAD, DN5355 significantly reduced cerebral Aβ plaques and hyperphosphorylated tau tangles. In Y-maze spontaneous alteration and contextual fear conditioning tests, 5XFAD mice showed amelioration of cognitive deficits upon the oral administration of DN5355.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Jisu Shin
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Kyeonghwan Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Darong Kim
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Won Seok Lee
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Jusuk Lee
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Illhwan Cho
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - In Wook Park
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Soljee Yoon
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
- Department of Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
| | - Songmin Lee
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Hye Yun Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Ji Hoon Lee
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - Ki Bum Hong
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (KMEDIhub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
- Department of Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, Republic of Korea
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
3
|
Mihaylova R, Angelova VT, Tchekalarova J, Atanasova D, Ivanova P, Simeonova R. Tailored Melatonin- and Donepezil-Based Hybrids Targeting Pathognomonic Changes in Alzheimer's Disease: An In Vitro and In Vivo Investigation. Int J Mol Sci 2024; 25:5969. [PMID: 38892154 PMCID: PMC11172853 DOI: 10.3390/ijms25115969] [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: 04/07/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
A plethora of pathophysiological events have been shown to play a synergistic role in neurodegeneration, revealing multiple potential targets for the pharmacological modulation of Alzheimer's disease (AD). In continuation to our previous work on new indole- and/or donepezil-based hybrids as neuroprotective agents, the present study reports on the beneficial effects of lead compounds of the series on key pathognomonic features of AD in both cellular and in vivo models. An enzyme-linked immunosorbent assay (ELISA) was used to evaluate the anti-fibrillogenic properties of 15 selected derivatives and identify quantitative changes in the formation of neurotoxic β-amyloid (Aβ42) species in human neuronal cells in response to treatment. Among the most promising compounds were 3a and 3c, which have recently shown excellent antioxidant and anticholinesterase activities, and, therefore, have been subjected to further in vivo investigation in mice. An acute toxicity study was performed after intraperitoneal (i.p.) administration of both compounds, and 1/10 of the LD50 (35 mg/kg) was selected for subacute treatment (14 days) with scopolamine in mice. Donepezil (DNPZ) and/or galantamine (GAL) were used as reference drugs, aiming to establish any pharmacological superiority of the multifaceted approach in battling hallmark features of neurodegeneration. Our promising results give first insights into emerging disease-modifying strategies to combine multiple synergistic activities in a single molecule.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1431 Sofia, Bulgaria; (R.M.); (V.T.A.); (R.S.)
| | - Violina T. Angelova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1431 Sofia, Bulgaria; (R.M.); (V.T.A.); (R.S.)
| | - Jana Tchekalarova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (D.A.); (P.I.)
| | - Dimitrinka Atanasova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (D.A.); (P.I.)
- Department of Anatomy, Faculty of Medicine, Trakia University, 6003 Stara Zagora, Bulgaria
| | - Petja Ivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (D.A.); (P.I.)
| | - Rumyana Simeonova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1431 Sofia, Bulgaria; (R.M.); (V.T.A.); (R.S.)
| |
Collapse
|
4
|
Gurdon B, Yates SC, Csucs G, Groeneboom NE, Hadad N, Telpoukhovskaia M, Ouellette A, Ouellette T, O'Connell KMS, Singh S, Murdy TJ, Merchant E, Bjerke I, Kleven H, Schlegel U, Leergaard TB, Puchades MA, Bjaalie JG, Kaczorowski CC. Detecting the effect of genetic diversity on brain composition in an Alzheimer's disease mouse model. Commun Biol 2024; 7:605. [PMID: 38769398 PMCID: PMC11106287 DOI: 10.1038/s42003-024-06242-1] [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: 08/16/2023] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease (AD) is broadly characterized by neurodegeneration, pathology accumulation, and cognitive decline. There is considerable variation in the progression of clinical symptoms and pathology in humans, highlighting the importance of genetic diversity in the study of AD. To address this, we analyze cell composition and amyloid-beta deposition of 6- and 14-month-old AD-BXD mouse brains. We utilize the analytical QUINT workflow- a suite of software designed to support atlas-based quantification, which we expand to deliver a highly effective method for registering and quantifying cell and pathology changes in diverse disease models. In applying the expanded QUINT workflow, we quantify near-global age-related increases in microglia, astrocytes, and amyloid-beta, and we identify strain-specific regional variation in neuron load. To understand how individual differences in cell composition affect the interpretation of bulk gene expression in AD, we combine hippocampal immunohistochemistry analyses with bulk RNA-sequencing data. This approach allows us to categorize genes whose expression changes in response to AD in a cell and/or pathology load-dependent manner. Ultimately, our study demonstrates the use of the QUINT workflow to standardize the quantification of immunohistochemistry data in diverse mice, - providing valuable insights into regional variation in cellular load and amyloid deposition in the AD-BXD model.
Collapse
Affiliation(s)
- Brianna Gurdon
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
| | - Sharon C Yates
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gergely Csucs
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nicolaas E Groeneboom
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, ME, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | - Andrew Ouellette
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
| | - Tionna Ouellette
- The Jackson Laboratory, Bar Harbor, ME, USA
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA
| | - Kristen M S O'Connell
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA
| | - Surjeet Singh
- The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Ingvild Bjerke
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Heidi Kleven
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ulrike Schlegel
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Trygve B Leergaard
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maja A Puchades
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jan G Bjaalie
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Catherine C Kaczorowski
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA.
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA.
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Xing S, Tang X, Wang L, Wang J, Lv B, Wang X, Guo C, Zhao Y, Feng F, Liu W, Chen Y, Sun H. Optimizing drug-like properties of selective butyrylcholinesterase inhibitors for cognitive improvement: Enhancing aqueous solubility by disrupting molecular plane. Eur J Med Chem 2024; 268:116289. [PMID: 38452730 DOI: 10.1016/j.ejmech.2024.116289] [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: 01/09/2024] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
Most recently, worldwide interest in butyrylcholinesterase (BChE) as a potential target for treating Alzheimer's disease (AD) has increased. In this study, the previously obtained selective BChE inhibitors with benzimidazole-oxadiazole scaffold were further structurally modified to increase their aqueous solubility and pharmacokinetic (PK) characteristics. S16-1029 showed improved solubility (3280 μM, upgraded by 14 times) and PK parameters, including plasma exposure (AUC0-inf = 1729.95 ng/mL*h, upgraded by 2.6 times) and oral bioavailability (Fpo = 48.18%, upgraded by 2 times). S16-1029 also displayed weak or no inhibition against Cytochrome P450 (CYP450) and human ether a-go-go related gene (hERG) potassium channel. In vivo experiments on tissue distribution revealed that S16-1029 could cross the blood-brain barrier (BBB) and reach the central nervous system (CNS). In vivo cognitive improvement efficacy and good in vitro target inhibitory activity (eqBChE IC50 = 11.35 ± 4.84 nM, hBChE IC50 = 48.1 ± 11.4 nM) were also assured. The neuroprotective effects against several AD pathology characteristics allowed S16-1029 to successfully protect the CNS of progressed AD patients. According to the findings of this study, altering molecular planarity might be a viable strategy for improving the drug-like property of CNS-treating drugs.
Collapse
Affiliation(s)
- Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Leyan Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Jun Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Xiaolong Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Can Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Ye Zhao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China; Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| |
Collapse
|
6
|
Junior MSO, Reiche L, Daniele E, Kortebi I, Faiz M, Küry P. Star power: harnessing the reactive astrocyte response to promote remyelination in multiple sclerosis. Neural Regen Res 2024; 19:578-582. [PMID: 37721287 PMCID: PMC10581572 DOI: 10.4103/1673-5374.380879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 09/19/2023] Open
Abstract
Astrocytes are indispensable for central nervous system development and homeostasis. In response to injury and disease, astrocytes are integral to the immunological- and the, albeit limited, repair response. In this review, we will examine some of the functions reactive astrocytes play in the context of multiple sclerosis and related animal models. We will consider the heterogeneity or plasticity of astrocytes and the mechanisms by which they promote or mitigate demyelination. Finally, we will discuss a set of biomedical strategies that can stimulate astrocytes in their promyelinating response.
Collapse
Affiliation(s)
- Markley Silva Oliveira Junior
- Department of Neurology, Neuroregeneration laboratory, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Neuroregeneration laboratory, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Emerson Daniele
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
| | - Ines Kortebi
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
| | - Maryam Faiz
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
| | - Patrick Küry
- Department of Neurology, Neuroregeneration laboratory, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
7
|
Xiong R, Li B, Yu H, Fan T, Yu H, Yang Y, Wang JZ, Pi G, Yang X. Urolithin A Inhibits Anterior Basolateral Amygdala to Ventral Hippocampal CA1 Circuit to Ameliorate Amyloid-β-Impaired Social Ability. J Alzheimers Dis 2024; 99:1303-1316. [PMID: 38759018 DOI: 10.3233/jad-240298] [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: 05/19/2024]
Abstract
Background Anxiety and social withdrawal are highly prevalent among patients with Alzheimer's disease (AD). However, the neural circuit mechanisms underlying these symptoms remain elusive, and there is a need for effective prevention strategies. Objective This study aims to elucidate the neural circuitry mechanisms underlying social anxiety in AD. Methods We utilized 5xFAD mice and conducted a series of experiments including optogenetic manipulation, Tandem Mass Tag-labeled proteome analysis, behavioral assessments, and immunofluorescence staining. Results In 5xFAD mice, we observed significant amyloid-β (Aβ) accumulation in the anterior part of basolateral amygdala (aBLA). Behaviorally, 6-month-old 5xFAD mice displayed excessive social avoidance during social interaction. Concurrently, the pathway from aBLA to ventral hippocampal CA1 (vCA1) was significantly activated and exhibited a disorganized firing patterns during social interaction. By optogenetically inhibiting the aBLA-vCA1 pathway, we effectively improved the social ability of 5xFAD mice. In the presence of Aβ accumulation, we identified distinct changes in the protein network within the aBLA. Following one month of administration of Urolithin A (UA), we observed significant restoration of the abnormal protein network within the aBLA. UA treatment also attenuated the disorganized firings of the aBLA-vCA1 pathway, leading to an improvement in social ability. Conclusions The aBLA-vCA1 circuit is a vulnerable pathway in response to Aβ accumulation during the progression of AD and plays a crucial role in Aβ-induced social anxiety. Targeting the aBLA-vCA1 circuit and UA administration are both effective strategies for improving the Aβ-impaired social ability.
Collapse
Affiliation(s)
- Rui Xiong
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binrui Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haitao Yu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Tianceng Fan
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guilin Pi
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
8
|
Yuan WQ, Huang WP, Jiang YC, Xu H, Duan CS, Chen NH, Liu YJ, Fu XM. The function of astrocytes and their role in neurological diseases. Eur J Neurosci 2023; 58:3932-3961. [PMID: 37831013 DOI: 10.1111/ejn.16160] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
Astrocytes have countless links with neurons. Previously, astrocytes were only considered a scaffold of neurons; in fact, astrocytes perform a variety of functions, including providing support for neuronal structures and energy metabolism, offering isolation and protection and influencing the formation, function and elimination of synapses. Because of these functions, astrocytes play an critical role in central nervous system (CNS) diseases. The regulation of the secretiory factors, receptors, channels and pathways of astrocytes can effectively inhibit the occurrence and development of CNS diseases, such as neuromyelitis optica (NMO), multiple sclerosis, Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease. The expression of aquaporin 4 in AS is directly related to NMO and indirectly involved in the clearance of Aβ and tau proteins in AD. Connexin 43 has a bidirectional effect on glutamate diffusion at different stages of stroke. Interestingly, astrocytes reduce the occurrence of PD through multiple effects such as secretion of related factors, mitochondrial autophagy and aquaporin 4. Therefore, this review is focused on the structure and function of astrocytes and the correlation between astrocytes and CNS diseases and drug treatment to explore the new functions of astrocytes with the astrocytes as the target. This, in turn, would provide a reference for the development of new drugs to protect neurons and promote the recovery of nerve function.
Collapse
Affiliation(s)
- Wen-Qin Yuan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wei-Peng Huang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Pharmacy, Minzu University of China, Beijing, China
| | - Yang-Chao Jiang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Hao Xu
- College of Economics and Management, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Chong-Shen Duan
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying-Jiao Liu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiao-Mei Fu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
9
|
Liu GZ, Niu TT, Yu Q, Xu BL, Li XQ, Yuan BY, Yuan GB, Yang TT, Li HQ, Sun Y. Ginkgolide attenuates memory impairment and neuroinflammation by suppressing the NLRP3/caspase-1 pathway in Alzheimer's disease. Aging (Albany NY) 2023; 15:10237-10252. [PMID: 37793010 PMCID: PMC10599747 DOI: 10.18632/aging.205072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/17/2023] [Indexed: 10/06/2023]
Abstract
The NLRP3 inflammasome is involved in the neuroinflammatory pathway of Alzheimer's disease (AD). The aim of this study is to explore the roles and underlying mechanisms of ginkgolide (Baiyu®) on amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mice and a murine microglial cell line, BV-2. In the present study, the APP/PS1 mice were administered with ginkgolide, followed by a Morris water maze test. The mice were then euthanized to obtain brain tissue for histological and Aβ analysis. Additionally, BV-2 cells were pretreated with ginkgolide and then incubated with Aβ1-42 peptide. NLRP3, ASC, and caspase-1 mRNA and protein expression in brain tissue of mice and BV-2 cells were quantified by real-time PCR and western blotting, as well as reactive oxygen species (ROS) production, interleukin (IL)-1β and IL-18 levels by lucigenin technique and ELISA. Compared with the APP/PS1 mice, ginkgolide-treated mice demonstrated the shortened escape latency, reduced plaques, less inflammatory cell infiltration and neuron loss in the hippocampi of APP/PS1 mice. The levels of NLRP3, ASC, caspase-1, ROS, IL-1β, and IL-18 were also decreased in the brain tissue of APP/PS1 mice or Aβ1-42-treated BV-2 cells following ginkgolide treatment. Ginkgolide exerted protective effects on AD, at least partly by inactivating the NLRP3/caspase-1 pathway.
Collapse
Affiliation(s)
- Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Tian-Tong Niu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Qian Yu
- Beijing D.A. Medical Laboratory, Beijing 102600, China
| | - Bao-Lei Xu
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiao-Qing Li
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Bo-Yi Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Guo-Bin Yuan
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hui-Qin Li
- Research and Development Centre, Chengdu Baiyu Pharmaceutical Co., Ltd., Chengdu 611130, China
| | - Yi Sun
- Research and Development Centre, Chengdu Baiyu Pharmaceutical Co., Ltd., Chengdu 611130, China
| |
Collapse
|
10
|
Murayama MA. The past and present of therapeutic strategy for Alzheimer's diseases: potential for stem cell therapy. Exp Anim 2023; 72:285-293. [PMID: 36878603 PMCID: PMC10435354 DOI: 10.1538/expanim.22-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disease characterized by cognitive dysfunction and neuropsychiatric symptoms, is the most prevalent form of dementia among the elderly. Amyloid aggregation, tau hyperphosphorylation, and neural cell loss are the main pathological features. Various hypotheses have been proposed to explain the development of AD. Some therapeutic agents have shown clinical benefits in patients with AD; however, many of these agents have failed. The degree of neural cell loss is associated with the severity of AD. Adult neurogenesis, which governs cognitive and emotional behaviors, occurs in the hippocampus, and some research groups have reported that neural cell transplantation into the hippocampus improves cognitive dysfunction in AD model mice. Based on these clinical findings, stem cell therapy for patients with AD has recently attracted attention. This review provides past and present therapeutic strategies for the management and treatment of AD.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
11
|
Gomaa AA, Farghaly HSM, Makboul RM, Hussien AM, Nicola MA. Polyphenols from Conyza dioscoridis (L.) ameliorate Alzheimer’s disease- like alterations through multi-targeting activities in two animal models. BMC Complement Med Ther 2022; 22:288. [PMID: 36348329 PMCID: PMC9644610 DOI: 10.1186/s12906-022-03765-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022] Open
Abstract
Background Recent investigations suggested that anticancer agents may inhibit the progression of Alzheimer's disease (AD) pathology. Conyza dioscoridis (L.) was demonstrated to have anticancer, antioxidant, anti-inflammatory and antidiabetic effects. This study was carried out to investigate the efficacy of polyphenols from Conyza dioscoridis (L.) extract (PCDE) on AD. Methods Impacts of 3 doses of PCDE and donepezil, a reference drug, on the features of Alzheimer's disease in two animal models were investigated. Results PCDE ameliorated the memory and learning impairment shown in rats following a single dose of scopolamine (scopolamine model) or 17 weeks of high-fat/high-fructose(HF/Hfr) diet coupled with a single dose of streptozotocin, (25 mg/kg) (T2D model). They reduced significantly the high hippocampal cholinesterase activity in the two models of rats. Administration of PCDE for 8 weeks in the T2D model showed a significant reduction in hippocampal GSK-3β, caspase-3 activity and increase in the inhibited glutamate receptor expression (AMPA GluR1 subunit and NMDA receptor subunits NR1, NR2A, NR2B). A significant reduction of HOMA-insulin resistance and serum hypercholesterolemia was observed. The Tau hyperphosphorylation and Aβ 1–42 generation in the hippocampal of T2D rats were significantly decreased by PCDE. Modulation of the oxidative stress markers, (rise in GH and SOD; decrease in MDA levels) and a significant reduction of TNF-α and IL-1β in the hippocampus of T2D rats treated by PCDE extract were important findings in this study. The highest dose tested was 4% of the highest safe dose. Conclusion Our study suggests that PCDE is multi-targeting agent with multiple beneficial activities in combating features of AD. This study may provide a novel therapeutic strategy for AD treatment that warrants clinical studies.
Collapse
|