1
|
Zhang W, Teng F, Lan X, Liu P, Wang A, Zhang F, Cui Z, Guan J, Sun X. A novel finding relates to the involvement of ATF3/DOCK8 in Alzheimer's disease pathogenesis. J Alzheimers Dis 2025:13872877251336266. [PMID: 40267290 DOI: 10.1177/13872877251336266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe involvement of microglia is likely to be pivotal in the pathogenesis of Alzheimer's disease (AD) by modulating the deposition of amyloid-β (Aβ) plaques. The deletion of Dedicator of cytokinesis 8 (DOCK8) has a protective effect in mouse with neurodegenerative diseases.ObjectiveTo explore the underlying mechanism of DOCK8 in AD.MethodsIn present study, we first the detected the expression of DOCK8 in the hippocampal tissue of APP/PS1 mice. Then, the expression of DOCK8 was knocked down in the hippocampal tissue of APP/PS1 mice, and the effects of DOCK8 down-regulation on cognitive function, the microglia migration around Aβ plaques, and the cell division cycle 42 (Cdc42)/p38 mitogen-activated protein kinase (MAPK) signaling pathway were detected. Next, the effects of DOCK8 knockdown on Aβ-induced migration and activation of BV-2 cells as well as the MAPK signaling pathway were detected. Finally, the transcriptional regulation of DOCK by transcription factor 3 (ATF3) was detected by a dual luciferase reporter assay.ResultsDOCK8 expression exerts a significant upregulation in the hippocampus of APP/PS1 mice. However, following the DOCK8 knockdown, there was a significant recovery in the results of the behavioral tests and a notable reduction in microglial expression. Moreover, the high expression of DOCK8 mediated by ATF3 successfully triggered the Cdc42/p38 MAPK signaling pathway, thereby enhancing the migration and recruitment of microglia towards senile plaques, accelerating the production of Aβ plaques.ConclusionsATF3-mediated high expression of DOCK8 accelerates the production of Aβ plaques, and participates in the pathogenesis of AD.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fei Teng
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xifa Lan
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
| | - Peihui Liu
- Neurointerventional Department, Huludao Central Hospital, Huludao, Liaoning, People's Republic of China
| | - Aiming Wang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fan Zhang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zhiqiang Cui
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jingwei Guan
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaohong Sun
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Science Experiment Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
2
|
Yang MX, Wang ZR, Zhang YL, Zhang ZN, Li YL, Wang R, Su Q, Guo JH. Albumin antagonizes Alzheimer's disease-related Tau pathology and enhances cognitive performance by inhibiting aberrant Tau aggregation. Exp Neurol 2025; 386:115155. [PMID: 39837459 DOI: 10.1016/j.expneurol.2025.115155] [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: 10/08/2024] [Revised: 01/04/2025] [Accepted: 01/17/2025] [Indexed: 01/23/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder primarily characterized by cognitive impairment, for which effective treatments remain lacking. Albumin (ALB) is an essential carrier protein found in various body fluids, playing crucial roles in anti-inflammatory processes, antioxidation, and signal transduction. Recent research indicates that ALB may play a significant role in the development and progression of AD, though its specific function is not yet fully understood. In this study, we observed a link between serum ALB levels and cognitive performance in the elderly. Administration of ALB intranasally was shown to enhance learning and memory in MAPT/P301S transgenic mice, markedly decreasing hyperphosphorylation of Tau protein and reducing neuronal apoptosis. In a neuronal cell model overexpressing Tau, ALB administration in vitro attenuated Tau-induced toxicity and reduced the production of phosphorylated Tau. Additionally, co-incubation of Tau with ALB significantly reduced the formation of neurofibrillary tangles. These results suggest that ALB improves AD-related cognitive function by preventing the pathological aggregation of Tau and reducing its abnormal phosphorylation. Furthermore, ALB's neuroprotective effect helps prevent neuronal apoptosis in the cortex and hippocampus, providing potential targets for AD prevention and treatment.
Collapse
Affiliation(s)
- Ming-Xuan Yang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Zhuo-Ran Wang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yan-Li Zhang
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan 030001, Shanxi, China
| | - Zhi-Na Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yan-Li Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Rui Wang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang 032200, Shanxi, China.
| | - Jun-Hong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
3
|
Li Y, Wang T, Li H, Jiang Y, Shen X, Kang N, Guo Z, Zhang R, Lu X, Kang T, Li M, Hou Y, Wu Y. Targeting LKB1-AMPK-SIRT1-induced autophagy and mitophagy pathways improves cerebrovascular homeostasis in APP/PS1 mice. Free Radic Biol Med 2025; 233:400-418. [PMID: 40180019 DOI: 10.1016/j.freeradbiomed.2025.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/14/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common and severe degenerative disorder of the central nervous system in the elderly, profoundly impacting patients' quality of life. However, effective therapeutic agents for AD are still lacking. Bazi Bushen capsule (BZBS) is a traditional Chinese herbal compound with potential neuroprotective effects, yet its underlying mechanisms remain poorly understood. METHODS In this study, we utilized APP/PS1 transgenic mice to assess the therapeutic efficacy of BZBS. Initially, we evaluated the spatial learning and memory of the mice using the Barnes maze. The brain microcirculation was assessed through a small-animal ultrasound system, two-photon in vivo imaging, and micro-computed tomography angiography. Molecular, biochemical, and pathological analyses were conducted on brain tissues. Through network pharmacology, we identified potential intervention pathways and targets for BZBS in the treatment of AD, which we subsequently validated both in vivo and in vitro. Additionally, we employed molecular virtual docking screening and biolayer interferometry to elucidate the direct interactions of ginsenoside Rg5 and ginsenoside Ro in BZBS with AMPK and LKB1 proteins. RESULTS The BZBS intervention significantly enhanced spatial learning and memory in APP/PS1 mice while decreasing Aβ deposition. Furthermore, BZBS protected cerebrovascular homeostasis and mitigated neuroinflammation, as evidenced by decreased blood-brain barrier permeability, increased expression of tight-junction proteins, and restored cerebral blood flow. Mechanistically, ginsenosides Rg5 and Ro in BZBS directly bind to AMPK and LKB1 proteins, activating the LKB1-AMPK-SIRT1 signaling pathway, promoting autophagy and mitochondrial autophagy, and alleviating oxidative stress damage in endothelial cells. CONCLUSIONS BZBS enhances autophagy-related activity, decreases Aβ deposition, and improves endothelial cell homeostasis through the activation of the LKB1-AMPK-SIRT1 signaling pathway, ultimately leading to improved cognitive function in mice with AD. This study highlights the importance of enhancing autophagic activity and maintaining cerebrovascular homeostasis in mitigating cognitive decline in AD, providing evidence and new insights into the application of compound medicines for treating age-related neurological disorders.
Collapse
Affiliation(s)
- Yawen Li
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Tongxing Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Hongrong Li
- Key Laboratory of State Administration of TCM (Cardio-Cerebral Vessel Collateral Disease), Shijiazhuang, 050035, China; Hebei Yiling Hospital, Shijiazhuang, 050035, China
| | - Yuning Jiang
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiaogang Shen
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Ning Kang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Zhifang Guo
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Runtao Zhang
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuan Lu
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Tianyu Kang
- Hebei Medical University, Shijiazhuang, 050017, China
| | - Mengnan Li
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| | - Yunlong Hou
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| | - Yiling Wu
- Hebei Medical University, Shijiazhuang, 050017, China; State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
| |
Collapse
|
4
|
Zhu L, Liu C, Wang Y, Zhu X, Wu L, Chen L, Zhou J, Wang F. METTL3/IGF2BP2/IκBα axis participates in neuroinflammation in Alzheimer's disease by regulating M1/M2 polarization of microglia. Neurochem Int 2025; 186:105964. [PMID: 40107503 DOI: 10.1016/j.neuint.2025.105964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Microglia-mediated neuroinflammation is closely related to the development of Alzheimer's disease (AD). This study further elucidated the regulatory mechanism of microglia polarization in AD. METHOD Microglia polarization was assessed using RT-qPCR, ELISA, and immunofluorescence (IF). Western blot (WB) analyzed inflammation-related, p-tau, and apoptosis-related proteins. Neuronal damage was evaluated by immunofluorescence, and neuronal apoptosis by flow cytometry and TUNEL assay. METTL3 and IκBα expression were detected using RT-qPCR and WB. N6-methyladenosine (m6A) levels were quantified with a colorimetric assay. RNA pull-down assay examined METTL3, IGF2BP2, and IκBα mRNA binding. IGF2BP expression was assessed by RT-qPCR. Learning and memory abilities were evaluated using morris water maze (MWM) test and novel object recognition (NOR) test. Inflammation-related proteins were detected using IF. RESULTS Stimulation with Aβ1-42 led to microglia M1 polarization, upregulation of inflammation-related proteins, and exacerbation of neuronal injury and apoptosis, along with increased p-tau expression in neurons. METTL3/IGF2BP2 modulated IκBα m6A modification through binding to IκBα mRNA, enhancing its expression. Enhanced METTL3 or IGF2BP2 expression suppressed M1 polarization, inflammation, and neuronal apoptosis in microglia, reversed by knockdown of IκBα. AD model mice exhibited cognitive impairments, neuroinflammation, and elevated M1 polarization. METTL3 or IGF2BP2 overexpression improved cognitive function, reduced neuroinflammation, and inhibited M1 polarization, and this effect was similarly reversed by knockdown of IκBα. CONCLUSION Our study demonstrates that the METTL3/IGF2BP2/IκBα axis is involved in neuroinflammation in AD by modulating microglia M1/M2 polarization, which sheds light on the treatment of AD.
Collapse
Affiliation(s)
- Ling Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Congyan Liu
- Department of pharmacy, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Yang Wang
- Department of radiology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Xuanang Zhu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lei Wu
- Department of neurology, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Lvan Chen
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China
| | - Jing Zhou
- College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| | - Fan Wang
- Department of neurosurgery, Jingmen Central Hospital, Jingmen Central Hospital affiliated to Jingchu University of Technology, Jingmen, 448000, China; College of Medical, Jingchu University of Technology, Jingmen, 448000, China.
| |
Collapse
|
5
|
Cai H, Hou F, Wang Y, Wu L, Wang Z, Wu M, Wang X, Hölscher C. Mitochondrial Calcium Uniporter knockdown improves the viability of HT22 hippocampal neurons in Alzheimer's disease. Eur J Pharmacol 2025; 991:177347. [PMID: 39914782 DOI: 10.1016/j.ejphar.2025.177347] [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: 10/23/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disorder that poses a serious threat because it has no cure. Recently, mitochondrial dysfunction has been shown to directly promote the development of AD. An imbalance in mitochondrial calcium (mCa2+) homeostasis is an important cause of mitochondrial dysfunction. Abnormal expression of mitochondrial calcium uniporter (MCU), a key channel responsible for mCa2+ uptake, can induce an imbalance in mCa2+ homeostasis, ultimately leading to mitochondrial dysfunction. Importantly, we observed a much higher expression level of MCU in the hippocampus of amyloid precursor protein (APP)/presenilin 1 (PS1)/tau transgenic mice than that in the hippocampus of control mice (C57), suggesting that MCU may be a target for the development of treatments for AD. Our recent study revealed the neuroprotective effect of MCU knockdown on hippocampal neurons in APP/PS1/tau mice. In the present study, we used MCU knockdown to investigate the cellular mechanisms involved in amyloid-β (Aβ)1-42 and okadaic acid (OA) cell models of AD. We found that MCU knockdown could increase the survival and decrease the apoptosis of these two cell models by lowering mCa2+ overload, further increasing the mitochondrial membrane potential (MMP) and ultimately reducing the overproduction of reactive oxygen species (ROS). This study showed that MCU knockdown could increase the proliferation and viability of HT22 hippocampal neurons, which might explain the neuroprotective effect of MCU knockdown on AD, potentially leading to the development of novel and effective therapies for AD.
Collapse
Affiliation(s)
- Hongyan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China.
| | - Fei Hou
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Yu Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Linhong Wu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China.
| | - Zhaojun Wang
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China; Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Meina Wu
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China; Key Laboratory of Cellular Physiology, Taiyuan, China; Department of Physiology, Shanxi Medical University, Taiyuan, China.
| | - Xiao Wang
- Department of Psychiatry, First Clinical Medical College of Shanxi Medical University, Taiyuan, China.
| | - Christian Hölscher
- Brain Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| |
Collapse
|
6
|
Kato R, Zhang L, Kinatukara N, Huang R, Asthana A, Weber C, Xia M, Xu X, Shah P. Investigating blood-brain barrier penetration and neurotoxicity of natural products for central nervous system drug development. Sci Rep 2025; 15:7431. [PMID: 40032960 PMCID: PMC11876671 DOI: 10.1038/s41598-025-90888-2] [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: 11/23/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Natural Products (NPs) are increasingly utilized worldwide for their potential therapeutic benefits, including central nervous system (CNS) disorders. Studies have shown açai berries mitigating Parkinson's disease progression through dopaminergic neuroprotection via Nrf-2 HO-1 pathways. Ashwagandha, an evergreen shrub, has shown potential as a therapeutic for neurodegenerative disorders via axonal regeneration in Aβ25-35-treated cortical neurons in vitro. In most cases, promising NPs are tested using in vitro assays or simpler systems during the early stages of drug discovery. However, a critical challenge lies in the lack of data on blood-brain barrier (BBB) penetration, which is a significant determinant for the successful development of CNS drugs. Our first goal was to test our in-house NP constituent library via the Parallel Artificial Membrane Permeability Assay (PAMPA-BBB), with the aim of understanding their BBB-penetration potential. Of the constituents tested, 255 were found to have moderate to high BBB permeability. Our next goal was to understand if these compounds could exhibit CNS toxicity. Neuronal viability and neurite outgrowth assays were performed with this subset to identify compounds with neurotoxicity potential. Around 35% of compounds tested showed neurite outgrowth inhibition. The habitual and widespread consumption of NPs underscores the importance of subjecting this subset of compounds to additional testing and validation in vivo to ascertain their potential detrimental effects. Understanding BBB permeability and assessing neurotoxicity mechanisms of NPs will significantly benefit the CNS drug discovery community.
Collapse
Affiliation(s)
- Rintaro Kato
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Li Zhang
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Nivedita Kinatukara
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Abhinav Asthana
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA
| | - Pranav Shah
- National Center for Advancing Translational Sciences (NCATS), 9808 Medical Center Drive, Rockville, MD, 20850, USA.
| |
Collapse
|
7
|
Nam YS, Gi EJ, Ko YS, Lee S, Cho HH. Trichostatin A suppresses hearing loss by reducing oxidative stress and inflammation in an Alport syndrome model. PLoS One 2025; 20:e0316033. [PMID: 39908276 DOI: 10.1371/journal.pone.0316033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/03/2024] [Indexed: 02/07/2025] Open
Abstract
Alport syndrome (AS) is a genetic disorder marked by mutations in type IV collagen, leading to kidney glomerular dysfunction. AS also involves the cochlea, causing late-onset progressive hearing loss. Currently, there are no therapeutic drugs to protect hearing from AS. HDAC inhibitors (HDACis) are chemical compounds that block the activity of histone deacetylase and are known to exert diverse biologic effects. We investigated the effect of Trichostatin A (TSA), an HDACi, to assess its potential to inhibit hearing deterioration in AS. Col4a3 knockout (KO) mice were treated with TSA at 3 weeks of age, and hearing levels were measured using auditory brainstem response (ABR). The results demonstrate that TSA significantly protects the hearing of KO mice compared to the untreated group. The TSA-treated group exhibited a reduction in the levels of oxidative stress markers 4-Hydroxynonenal and 3-Nitrotyrosine, along with a decrease in inflammatory cytokines, in both the mouse cochlea and in vitro HEI-OC1 (House Ear Institute-Organ of Corti 1) cell and HEK (Human Embryonic Kidney)293T cells. AS demonstrated a thickening in the stria vascular vessels, a phenomenon that TSA attenuated. Col4α3 deficiency showed down-regulation of Hemeoxygenase-1 (HO-1), a key anti-inflammatory molecule. TSA treatment induced HO-1 signaling, which contributed to the inhibition of oxidative stress and inflammatory cytokines. These findings suggest that TSA represents a promising candidate molecule for mitigating the progression of hearing loss in AS.
Collapse
Affiliation(s)
- Yoon Seok Nam
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Eun-Ji Gi
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Biomedical Science, College of Medicine, Chonnam National University Graduate School, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| | - Yoo-Seung Ko
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School and Chonnam National University Hospital, Gwangju, Republic of Korea
- Department of Biomedical Science, College of Medicine, Chonnam National University Graduate School, BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
8
|
Zhang Y, Li T, Miao J, Zhang Z, Yang M, Wang Z, Yang B, Zhang J, Li H, Su Q, Guo J. Gamma-glutamyl transferase 5 overexpression in cerebrovascular endothelial cells improves brain pathology, cognition, and behavior in APP/PS1 mice. Neural Regen Res 2025; 20:533-547. [PMID: 38819065 PMCID: PMC11317949 DOI: 10.4103/nrr.nrr-d-23-01525] [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: 09/10/2023] [Revised: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00030/figure1/v/2024-05-28T214302Z/r/image-tiff In patients with Alzheimer's disease, gamma-glutamyl transferase 5 (GGT5) expression has been observed to be downregulated in cerebrovascular endothelial cells. However, the functional role of GGT5 in the development of Alzheimer's disease remains unclear. This study aimed to explore the effect of GGT5 on cognitive function and brain pathology in an APP/PS1 mouse model of Alzheimer's disease, as well as the underlying mechanism. We observed a significant reduction in GGT5 expression in two in vitro models of Alzheimer's disease (Aβ1-42-treated hCMEC/D3 and bEnd.3 cells), as well as in the APP/PS1 mouse model. Additionally, injection of APP/PS1 mice with an adeno-associated virus encoding GGT5 enhanced hippocampal synaptic plasticity and mitigated cognitive deficits. Interestingly, increasing GGT5 expression in cerebrovascular endothelial cells reduced levels of both soluble and insoluble amyloid-β in the brains of APP/PS1 mice. This effect may be attributable to inhibition of the expression of β-site APP cleaving enzyme 1, which is mediated by nuclear factor-kappa B. Our findings demonstrate that GGT5 expression in cerebrovascular endothelial cells is inversely associated with Alzheimer's disease pathogenesis, and that GGT5 upregulation mitigates cognitive deficits in APP/PS1 mice. These findings suggest that GGT5 expression in cerebrovascular endothelial cells is a potential therapeutic target and biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi Province, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhina Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mingxuan Yang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhuoran Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Yang
- Department of Hernia and Abdominal Wall Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jiawei Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Haiting Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi Province, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
9
|
Ateya NH, Al-Taie SF, Jasim SA, Uthirapathy S, Chaudhary K, Rani P, Kundlas M, Naidu KS, Amer NA, Ahmed JK. Histone Deacetylation in Alzheimer's Diseases (AD); Hope or Hype. Cell Biochem Biophys 2025:10.1007/s12013-025-01670-0. [PMID: 39825060 DOI: 10.1007/s12013-025-01670-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
Histone acetylation is the process by which histone acetyltransferases (HATs) add an acetyl group to the N-terminal lysine residues of histones, resulting in a more open chromatin structure. Histone acetylation tends to increase gene expression more than methylation does. In the central nervous system (CNS), histone acetylation is essential for controlling the expression of genes linked to cognition and learning. Histone deacetylases (HDACs), "writing" enzymes (HATs), and "reading" enzymes with bromodomains that identify and localize to acetylated lysine residues are responsible for maintaining histone acetylation. By giving animals HDAC inhibitors (HDACis), it is possible to intentionally control the ratios of "writer" and "eraser" activity, which will change the acetylation of histones. In addition to making the chromatin more accessible, these histone acetylation alterations re-allocate the targeting of "readers," including the transcriptional co-activators, cAMP response element-binding protein (CBP), and bromodomain-containing protein 4 (Brd4) in the CNS. Conclusive evidence has shown that HDACs slow down the progression of Alzheimer's disease (AD) by reducing the amount of histone acetylation, decreasing the activity of genes linked to memory, supporting cognitive decline and Amyloid beta (Aβ) protein accumulation, influencing aberrant tau phosphorylation, and promoting the emergence of neurofibrillary tangles (NFTs). In this review, we have covered the therapeutic targets and functions of HDACs that might be useful in treating AD.
Collapse
Affiliation(s)
- Nabaa Hisham Ateya
- Biotechnology Department, College of Applied Science, Fallujah University, Al-Fallujah, Iraq
| | - Sarah F Al-Taie
- University of Baghdad, College of Science, Department of Biotechnology, Baghdad, Iraq
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Ramadi, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University Erbil, Kurdistan Region, Erbil, Iraq
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Pooja Rani
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Nevin Adel Amer
- Nursing Department, College of Applied Medical Sciences, Jouf University, Sakakah, Saudi Arabia
- Medical Surgical Nursing Department, Faculty of Nursing, Menofia University, Shibin el Kom, Saudi Arabia
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
10
|
Alkhatabi HA, Pushparaj PN. Untangling the complex mechanisms associated with Alzheimer's disease in elderly patients using high-throughput RNA sequencing data and next-generation knowledge discovery methods: Focus on potential gene signatures and drugs for dementia. Heliyon 2025; 11:e41266. [PMID: 39834440 PMCID: PMC11743088 DOI: 10.1016/j.heliyon.2024.e41266] [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: 02/21/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 01/22/2025] Open
Abstract
Objectives Alzheimer's disease (AD) is a complex neurodegenerative disorder that primarily affects elderly individuals. This study aimed to elucidate the intricate mechanisms underlying AD in elderly patients compared with healthy aged individuals using high-throughput RNA sequencing (RNA-seq) data and next-generation knowledge discovery methods (NGKD), with a focus on identifying potential therapeutic agents. Methods High-throughput RNA-seq data were obtained from the Gene Expression Omnibus (GEO) database (accession number: GSE104704). These data were derived from healthy and diseased human brains (eight young healthy brains [young], 10 aged healthy brains [Old], and 12 aged diseased brains [AD]). We used NGKD tools such as GEO RNA-seq Experiments Interactive Navigator (GREIN) to obtain differentially expressed genes (DEGs) by comparing the AD versus Old RNA-seq data and further filtered and normalized to obtain differentially regulated Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome and Panther pathways using ExpressAnalyst tool. Besides, WebGestalt was used to identify differentially regulated Gene Ontologies (GO) and the pre-ranked Gene Set Enrichment Analysis (GSEA) was performed using GSEA software. The X2K web tool was used to infer upstream regulator networks and X2K Appyter tool for obtaining transcription factors (TFs) and kinase network information. LFW1000 and L1000CDS2 tools were used to identify specific drugs that reverse AD-associated gene signatures in elderly patients. Results Our study revealed significant downregulation of pathways related to neuroactive receptor-ligand interaction, synaptic vesicle cycle, and neuronal system in elderly individuals with AD. GO analysis showed negative enrichment of functions related to cognition, potassium ion transport, receptor-ligand activity, SNARE binding, and primary lysosomes. The transcription factors SUZ12 and REST, along with increased MAPK signaling, were identified as key regulators of downregulated genes. Several drugs and natural products, including dihydroergocristine, mepacrine, gedunin, amlodipine, and disulfiram have been identified as potential therapeutic agents for reversing AD-associated gene signatures. Conclusions This comprehensive analysis of AD in elderly individuals using RNA-seq data and NGKD tools revealed multiple differentially regulated pathways, gene signatures, and potential drugs for dementia treatment. These findings highlight the complex molecular mechanisms underlying AD and provide insights into potential therapeutic strategies. Further research is needed to validate these findings and to develop personalized treatment approaches for AD in elderly patients.
Collapse
Affiliation(s)
- Hind A. Alkhatabi
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Institute of Genomic Medicine Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
11
|
González Molina LA, Dolga AM, Rots MG, Sarno F. The Promise of Epigenetic Editing for Treating Brain Disorders. Subcell Biochem 2025; 108:111-190. [PMID: 39820862 DOI: 10.1007/978-3-031-75980-2_4] [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: 01/19/2025]
Abstract
Brain disorders, especially neurodegenerative diseases, affect millions of people worldwide. There is no causal treatment available; therefore, there is an unmet clinical need for finding therapeutic options for these diseases. Epigenetic research has resulted in identification of various genomic loci with differential disease-specific epigenetic modifications, mainly DNA methylation. These biomarkers, although not yet translated into clinically approved options, offer therapeutic targets as epigenetic modifications are reversible. Indeed, clinical trials are designed to inhibit epigenetic writers, erasers, or readers using epigenetic drugs to interfere with epigenetic dysregulation in brain disorders. However, since such drugs elicit genome-wide effects and potentially cause toxicity, the recent developments in the field of epigenetic editing are gaining widespread attention. In this review, we provide examples of epigenetic biomarkers and epi-drugs, while describing efforts in the field of epigenetic editing, to eventually make a difference for the currently incurable brain disorders.
Collapse
Affiliation(s)
- Luis A González Molina
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Faculty of Science and Engineering, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Marianne G Rots
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Federica Sarno
- Epigenetic Editing, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
12
|
Sanchez-Fernandez E, Guerra-Ojeda S, Suarez A, Serna E, Mauricio MD. Histone Deacetylase Inhibitors as a Promising Treatment Against Myocardial Infarction: A Systematic Review. J Clin Med 2024; 13:7797. [PMID: 39768722 PMCID: PMC11728224 DOI: 10.3390/jcm13247797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Acute myocardial infarction (AMI) is a critical medical condition that requires immediate attention to minimise heart damage and improve survival rates. Early identification and prompt treatment are essential to save the patient's life. Currently, the treatment strategy focuses on restoring blood flow to the myocardium as quickly as possible. However, reperfusion activates several cellular cascades that contribute to organ dysfunction, resulting in the ischaemia/reperfusion (I/R) injury. The search for treatments against AMI and I/R injury is urgent due to the shortage of effective treatments at present. In this regard, histone deacetylase (HDAC) inhibitors emerge as a promising treatment against myocardial infarction. The objective of this systematic review is to analyse the effects of HDAC inhibitors on ventricular function, cardiac remodelling and infarct size, among other parameters, focusing on the signalling pathways that may mediate these cardiovascular effects and protect against AMI. Methods: Original experimental studies examining the effects of HDAC inhibitors on AMI were included in the review using the PubMed and Scopus databases. Non-experimental papers were excluded. The SYRCLE RoB tool was used to assess risk of bias and the results were summarised in a table and presented in sections according to the type of HDAC inhibitor used. Results: A total of 18 studies were included, 10 of them using trichostatin A (TSA) as an HDAC inhibitor and concluding that the treatment improved ventricular function, reduced infarct size, and inhibited myocardial hypertrophy and remodelling after AMI. Other HDAC inhibitors, such as suberoylanilide hydroxamic acid (SAHA), valproic acid (VPA), mocetinostat, givinostat, entinostat, apicidin, and RGFP966, were also analysed, showing antioxidant and anti-inflammatory effects, an improvement in cardiac function and remodelling, and a decrease in apoptosis, among other effects. Conclusions: HDAC inhibitors constitute a significant promise for the treatment of AMI due to their diverse cardioprotective effects. However, high risk of selection, performance, and detection bias in the in vivo studies means that their application in the clinical setting is still a long way off and more research is needed to better understand their benefits and possible side effects.
Collapse
Affiliation(s)
- Eduardo Sanchez-Fernandez
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (E.S.-F.); (S.G.-O.); (A.S.); (E.S.)
| | - Sol Guerra-Ojeda
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (E.S.-F.); (S.G.-O.); (A.S.); (E.S.)
- Instituto Investigación Hospital Clínico-INCLIVA, 46010 Valencia, Spain
| | - Andrea Suarez
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (E.S.-F.); (S.G.-O.); (A.S.); (E.S.)
- Instituto Investigación Hospital Clínico-INCLIVA, 46010 Valencia, Spain
| | - Eva Serna
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (E.S.-F.); (S.G.-O.); (A.S.); (E.S.)
- Instituto Investigación Hospital Clínico-INCLIVA, 46010 Valencia, Spain
| | - Maria D. Mauricio
- Department of Physiology, Universitat de Valencia, 46010 Valencia, Spain; (E.S.-F.); (S.G.-O.); (A.S.); (E.S.)
- Instituto Investigación Hospital Clínico-INCLIVA, 46010 Valencia, Spain
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBER-CV), 28029 Madrid, Spain
| |
Collapse
|
13
|
Gao QC, Liu GL, Wang Q, Zhang SX, Ji ZL, Wang ZJ, Wu MN, Yu Q, He PF. A promising drug repurposing approach for Alzheimer's treatment: Givinostat improves cognitive behavior and pathological features in APP/PS1 mice. Redox Biol 2024; 78:103420. [PMID: 39577323 PMCID: PMC11621940 DOI: 10.1016/j.redox.2024.103420] [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: 10/04/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, characterized by memory loss, speech and motor defects, personality changes, and psychological disorders. The exact cause of AD remains unclear. Current treatments focus on maintaining neurotransmitter levels or targeting β-amyloid (Aβ) protein, but these only alleviate symptoms and do not reverse the disease. Developing new drugs is time-consuming, costly, and has a high failure rate. Utilizing multi-omics for drug repositioning has emerged as a new strategy. Based on transcriptomic perturbation data of over 40,000 drugs in human cells from the LINCS-L1000 database, our study employed the Jaccard index and hypergeometric distribution test for reverse transcriptional feature matching analysis, identifying Givinostat as a potential treatment for AD. Our research found that Givinostat improved cognitive behavior and brain pathology in models and enhanced hippocampal synaptic plasticity. Transcriptome sequencing revealed increased expression of mitochondrial respiratory chain complex proteins in the brains of APP/PS1 mice after Givinostat treatment. Functionally, Givinostat restored mitochondrial membrane potential, reduced reactive oxygen species, and increased ATP content in Aβ-induced HT22 cells. Additionally, it improved mitochondrial morphology and quantity in the hippocampus of APP/PS1 mice and enhanced brain glucose metabolic activity. These effects are linked to Givinostat promoting mitochondrial biogenesis and improving mitochondrial function. In summary, Givinostat offers a promising new strategy for AD treatment by targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Qi-Chao Gao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Big Data for Clinical Decision Research in Shanxi Province, Taiyuan, China; Department of Physiology, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Taiyuan, China
| | - Ge-Liang Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Big Data for Clinical Decision Research in Shanxi Province, Taiyuan, China
| | - Qi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Big Data for Clinical Decision Research in Shanxi Province, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology and Immunology, The Second Hospital of Shanxi Medical University, Taiyuan, China; Department of Physiology, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Taiyuan, China
| | - Zhi-Lin Ji
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhao-Jun Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; Department of Physiology, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Taiyuan, China
| | - Mei-Na Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China; Department of Physiology, Shanxi Medical University, Key Laboratory of Cellular Physiology, Ministry of Education, Key Laboratory of Cellular Physiology in Shanxi Province, Taiyuan, China
| | - Qi Yu
- Key Laboratory of Big Data for Clinical Decision Research in Shanxi Province, Taiyuan, China; School of Management, Shanxi Medical University, Taiyuan, China.
| | - Pei-Feng He
- Key Laboratory of Big Data for Clinical Decision Research in Shanxi Province, Taiyuan, China; School of Management, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
14
|
Wei B, Shi Y, Yu X, Cai Y, Zhao Y, Song Y, Zhao Z, Huo M, Li L, Gao Q, Yu D, Wang B, Sun M. GR/P300 Regulates MKP1 Signaling Pathway and Mediates Depression-like Behavior in Prenatally Stressed Offspring. Mol Neurobiol 2024; 61:10613-10628. [PMID: 38769227 DOI: 10.1007/s12035-024-04244-y] [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: 10/13/2023] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Accumulating evidence suggests that prenatal stress (PNS) increases offspring susceptibility to depression, but the underlying mechanisms remain unclear. We constructed a mouse model of prenatal stress by spatially restraining pregnant mice from 09:00-11:00 daily on Days 5-20 of gestation. In this study, western blot analysis, quantitative real-time PCR (qRT‒PCR), immunofluorescence, immunoprecipitation, chromatin immunoprecipitation (ChIP), and mifepristone rescue assays were used to investigate alterations in the GR/P300-MKP1 and downstream ERK/CREB/TRKB pathways in the brains of prenatally stressed offspring to determine the pathogenesis of the reduced neurogenesis and depression-like behaviors in offspring induced by PNS. We found that prenatal stress leads to reduced hippocampal neurogenesis and depression-like behavior in offspring. Prenatal stress causes high levels of glucocorticoids to enter the fetus and activate the hypothalamic‒pituitary‒adrenal (HPA) axis, resulting in decreased hippocampal glucocorticoid receptor (GR) levels in offspring. Furthermore, the nuclear translocation of GR and P300 (an acetylation modifying enzyme) complex in the hippocampus of PNS offspring increased significantly. This GR/P300 complex upregulates MKP1, which is a negative regulator of the ERK/CREB/TRKB signaling pathway associated with depression. Interestingly, treatment with a GR antagonist (mifepristone, RU486) increased hippocampal GR levels and decreased MKP1 expression, thereby ameliorating abnormal neurogenesis and depression-like behavior in PNS offspring. In conclusion, our study suggested that the regulation of the MKP1 signaling pathway by GR/P300 is involved in depression-like behavior in prenatal stress-exposed offspring and provides new insights and ideas for the fetal hypothesis of mental health.
Collapse
Affiliation(s)
- Bin Wei
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yajun Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Xi Yu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yongle Cai
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yan Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Yueyang Song
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Zejun Zhao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Ming Huo
- Reproductive Medicine Center, The First Hospital of Lanzhou University, LanzhouGansu, 730000, China
| | - Lingjun Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Qinqin Gao
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China
| | - Dongyi Yu
- Center for Medical Genetics and Prenatal Diagnosis, Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250000, Shandong, China
| | - Bin Wang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou City, 215006, Jiangsu, China.
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory for Complex Severe and Rare Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China.
| |
Collapse
|
15
|
Zhang MJ, Zhong JH, Yi XJ. Retraction Note: Effect of Curcumin on Cognitive Behavior and Pathological Characteristics of the Hippocampus in Mice with Inherent Alzheimer's Disease. Bull Exp Biol Med 2024:10.1007/s10517-024-06251-4. [PMID: 39528854 DOI: 10.1007/s10517-024-06251-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- M J Zhang
- Guangzhou Vocational and Technical University of Science and Technology, Guang Zhou, Guang Dong, China.
| | - J H Zhong
- Department of Acupuncture and Moxibustion, Shangyou Hospital of Traditional Chinese Medicine, Ganzhou City, Jiangxi, China
| | - X J Yi
- Guangzhou Vocational and Technical University of Science and Technology, Guang Zhou, Guang Dong, China
| |
Collapse
|
16
|
Singh R, Rathore AS, Dilnashin H, Keshri PK, Gupta NK, Prakash SAS, Zahra W, Singh S, Singh SP. HAT and HDAC: Enzyme with Contradictory Action in Neurodegenerative Diseases. Mol Neurobiol 2024; 61:9110-9124. [PMID: 38587698 DOI: 10.1007/s12035-024-04115-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 03/08/2024] [Indexed: 04/09/2024]
Abstract
In view of the increasing risk of neurodegenerative diseases, epigenetics plays a fundamental role in the field of neuroscience. Several modifications have been studied including DNA methylation, histone acetylation, histone phosphorylation, etc. Histone acetylation and deacetylation regulate gene expression, and the regular activity of histone acetyltransferases (HATs) and histone deacetylases (HDACs) provides regulatory stages for gene expression and cell cycle. Imbalanced homeostasis in these enzymes causes a detrimental effect on neurophysiological function. Intriguingly, epigenetic remodelling via histone acetylation in certain brain areas has been found to play a key role in the neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. It has been demonstrated that a number of HATs have a role in crucial brain processes such regulating neuronal plasticity and memory formation. The most recent therapeutic methods involve the use of small molecules known as histone deacetylase (HDAC) inhibitors that antagonize HDAC activity thereby increase acetylation levels in order to prevent the loss of HAT function in neurodegenerative disorders. The target specificity of the HDAC inhibitors now in use raises concerns about their applicability, despite the fact that this strategy has demonstrated promising therapeutic outcomes. The aim of this review is to summarize the cross-linking between histone modification and its regulation in the pathogenesis of neurological disorders. Furthermore, these findings also support the notion of new pharmacotherapies that target particular areas of the brain using histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Richa Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Hagera Dilnashin
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Nitesh Kumar Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Singh Ankit Satya Prakash
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Walia Zahra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Shekhar Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221005 (U.P.), India.
| |
Collapse
|
17
|
Wang S, Huang J, Chen Y, Liang Y, Chen L, Ye D, Yang H, Hui Z, Wang X, Zhang Z, Zhu X. Qifu-yin activates the Keap1/Nrf2/ARE signaling and ameliorates synaptic injury and oxidative stress in APP/PS1 mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118497. [PMID: 38942156 DOI: 10.1016/j.jep.2024.118497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional medicinal formulation, Qifu-yin (QFY), has been widely prescribed for Alzheimer's disease (AD) treatment in China, yet the comprehensive mechanisms through which QFY mitigates AD pathology remain to be fully delineated. AIM OF THE STUDY This study aimed to explore the therapeutic implications of QFY on the synaptic injury and oxidative stress in the hippocampus of APPswe/PS1dE9 (APP/PS1) mice, with a concerted effort to elucidate the molecular mechanisms related to synaptic preservation and memory improvement. MATERIALS AND METHODS The components of QFY were identified by ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). The neuroprotective effects of QFY was evaluated using six-month-old male APP/PS1 mice. Subsequent to a 15 days of QFY regimen, spatial memory was assessed utilizing the Morris water maze (MWM) test. Amyloid-beta (Aβ) aggregation was detected via immunostaining, while the quantification of Aβ1-40 and Aβ1-42 was achieved through enzyme-linked immunosorbent assay (ELISA). Transmission electron microscopy (TEM) was used to investigate the synaptic structure and mitochondrial morphology. Golgi staining was applied to examine dendritic spine density. Reactive oxygen species (ROS), 3-nitrotyrosine (3-NT) and 4-hydroxy-nonenal (4-HNE) assays were employed to assess oxidative stress. The expression profiles of Aβ metabolism-associated enzymes and the Keap1/Nrf2/ARE signaling pathway were determined by Western blot. RESULTS A total of 20 principal compounds in QFY were identified. QFY mitigated memory deficits of APP/PS1 mice, including reducing escape latency and search distance and increasing the time and distance spent in the target quadrant. In addition, QFY increased platform crossings of APP/PS1 mice in the probe trial of MWM tests. TEM analysis showed that QFY increased synapse number in the CA1 region of APP/PS1 mice. Further studies indicated that QFY elevated the expression levels of Post synaptic density protein 95 (PSD95) and synaptophysin, and mitigated the loss of dendritic spine density in the hippocampus of APP/PS1 mice. QFY has been shown to ameliorated the structural abnormalities of mitochondria, including mitochondrial dissolution and degradation, up-regulate ATP synthesis and membrane potential in the hippocampus of APP/PS1 mice. Moreover, QFY activated the Keap1/Nrf2/ARE signaling pathway in the hippocampus of APP/PS1 mice, which might contribute to the neuroprotective effects of QFY. CONCLUSION QFY activates the Keap1/Nrf2/ARE signaling, and protects against synaptic and mitochondrial dysfunction in APP/PS1 mice, proposing a potential alternative therapeutic strategy for AD management.
Collapse
Affiliation(s)
- Sulei Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Jing Huang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Yanting Chen
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Ying Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Liqiu Chen
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Dan Ye
- Department of Neurology, The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, Changzhou, 213000, China.
| | - Hui Yang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, 210046, China.
| | - Zhen Hui
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Xiaomian Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Zhennian Zhang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210001, China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, China.
| |
Collapse
|
18
|
Htet M, Estay-Olmos C, Hu L, Wu Y, Powers BE, Campbell CD, Ahmed MR, Hohman TJ, Schneider JA, Bennett DA, Menon V, De Jager PL, Kaas GA, Colbran RJ, Greer CB. HEXIM1 is correlated with Alzheimer's disease pathology and regulates immediate early gene dynamics in neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615234. [PMID: 39386727 PMCID: PMC11463448 DOI: 10.1101/2024.09.27.615234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Impaired memory formation and recall is a distinguishing feature of Alzheimer's disease, and memory requires de novo gene transcription in neurons. Rapid and robust transcription of many genes is facilitated by the formation of a poised basal state, in which RNA polymerase II (RNAP2) has initiated transcription, but is paused just downstream of the gene promoter. Neuronal depolarization releases the paused RNAP2 to complete the synthesis of messenger RNA (mRNA) transcripts. Paused RNAP2 release is controlled by positive transcription elongation factor b (P-TEFb), which is sequestered into a larger inactive complex containing Hexamethylene bisacetamide inducible protein 1 (HEXIM1) under basal conditions. In this work, we find that neuronal expression of HEXIM1 mRNA is highly correlated with human Alzheimer's disease pathologies. Furthermore, P-TEFb regulation by HEXIM1 has a significant impact on the rapid induction of neuronal gene transcription, particularly in response to repeated depolarization. These data indicate that HEXIM1/P-TEFb has an important role in inducible gene transcription in neurons, and for setting and resetting the poised state that allows for the robust activation of genes necessary for synaptic plasticity. GRAPHICAL ABSTRACT
Collapse
|
19
|
Liang T, Liu S, Dang B, Luan X, Guo Y, Steimbach RR, Hu J, Lu L, Yue P, Wang R, Zheng M, Gao J, Yin X, Chen X. Multimechanism biological profiling of tetrahydro-β-carboline analogues as selective HDAC6 inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2024; 275:116624. [PMID: 38925015 DOI: 10.1016/j.ejmech.2024.116624] [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: 04/02/2024] [Revised: 06/08/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
With the intensive research on the pathogenesis of Alzheimer's disease (AD), inhibition of HDAC6 appears to be a potential therapeutic approach for AD. In this paper, a series of tetrahydro-β-carboline derivatives with hydroxamic acid group were fast synthesized. Among all, the most potent 15 selectively inhibited HDAC6 with IC50 of 15.2 nM and markedly increased acetylated alpha-tubulin levels. In cellular assay, 15 showed excellent neurotrophic effect by increasing the expression of GAP43 and Beta-3 tubulin markers. Besides, 15 showed neuroprotective effects in PC12 or SH-SY5Y cells against H2O2 and 6-OHDA injury through activation of Nrf2, catalase and Prx II, and significantly reduced H2O2-induced reactive oxygen species (ROS) production. In vivo, 15 significantly attenuated zebrafish anxiety-like behaviour and memory deficits in a SCOP-induced zebrafish model of AD. To sum up, multifunctional 15 might be a good lead to develop novel tetrahydrocarboline-based agents for the treatment of AD.
Collapse
Affiliation(s)
- Ting Liang
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Shiru Liu
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Baiyun Dang
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Xiaofa Luan
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Yifan Guo
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Raphael R Steimbach
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany; Biosciences Faculty, University of Heidelberg, 69120, Heidelberg, Germany
| | - Jiadong Hu
- School of Medicinal and Chemical Engineering, Yangling Vocational & Technical College, Yangling, 712100, PR China
| | - Long Lu
- School of Medicinal and Chemical Engineering, Yangling Vocational & Technical College, Yangling, 712100, PR China
| | - Peiyu Yue
- School of Medicinal and Chemical Engineering, Yangling Vocational & Technical College, Yangling, 712100, PR China
| | - Ruotian Wang
- School of Medicinal and Chemical Engineering, Yangling Vocational & Technical College, Yangling, 712100, PR China
| | - Meng Zheng
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China
| | - Jinming Gao
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China.
| | - Xia Yin
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China.
| | - Xin Chen
- Shaanxi Key Labotory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, 712100, PR China.
| |
Collapse
|
20
|
Reda A, Hategan LA, McLean TAB, Creighton SD, Luo JQ, Chen SES, Hua S, Winston S, Reeves I, Padmanabhan A, Dahi TA, Ramzan F, Brimble MA, Murphy PJ, Walters BJ, Stefanelli G, Zovkic IB. Role of the histone variant H2A.Z.1 in memory, transcription, and alternative splicing is mediated by lysine modification. Neuropsychopharmacology 2024; 49:1285-1295. [PMID: 38366138 PMCID: PMC11224360 DOI: 10.1038/s41386-024-01817-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/18/2024]
Abstract
Creating long-lasting memories requires learning-induced changes in gene expression, which are impacted by epigenetic modifications of DNA and associated histone proteins. Post-translational modifications (PTMs) of histones are key regulators of transcription, with different PTMs producing unique effects on gene activity and behavior. Although recent studies implicate histone variants as novel regulators of memory, effects of PTMs on the function of histone variants are rarely considered. We previously showed that the histone variant H2A.Z suppresses memory, but it is unclear if this role is impacted by H2A.Z acetylation, a PTM that is typically associated with positive effects on transcription and memory. To answer this question, we used a mutation approach to manipulate acetylation on H2A.Z without impacting acetylation of other histone types. Specifically, we used adeno-associated virus (AAV) constructs to overexpress mutated H2A.Z.1 isoforms that either mimic acetylation (acetyl-mimic) by replacing lysines 4, 7 and 11 with glutamine (KQ), or H2A.Z.1 with impaired acetylation (acetyl-defective) by replacing the same lysines with alanine (KA). Expressing the H2A.Z.1 acetyl-mimic (H2A.Z.1KQ) improved memory under weak learning conditions, whereas expressing the acetyl-defective H2A.Z.1KA generally impaired memory, indicating that the effect of H2A.Z.1 on memory depends on its acetylation status. RNA sequencing showed that H2A.Z.1KQ and H2A.Z.1KA uniquely impact the expression of different classes of genes in both females and males. Specifically, H2A.Z.1KA preferentially impacts genes involved in synaptic function, suggesting that acetyl-defective H2A.Z.1 impairs memory by altering synaptic regulation. Finally, we describe, for the first time, that H2A.Z is also involved in alternative splicing of neuronal genes, whereby H2A.Z depletion, as well as expression of H2A.Z.1 lysine mutants influence transcription and splicing of different gene targets, suggesting that H2A.Z.1 can impact behavior through effects on both splicing and gene expression. This is the first study to demonstrate that direct manipulation of H2A.Z post-translational modifications regulates memory, whereby acetylation adds another regulatory layer by which histone variants can fine tune higher brain functions through effects on gene expression and splicing.
Collapse
Affiliation(s)
- Anas Reda
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Luca A Hategan
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Timothy A B McLean
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Samantha D Creighton
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada
| | - Jian Qi Luo
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Sean En Si Chen
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Shan Hua
- Departments of Biology and Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Stephen Winston
- Department of Surgery and Graduate school of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Isaiah Reeves
- Department of Surgery and Graduate school of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Aditya Padmanabhan
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada
| | - Tarkan A Dahi
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada
| | - Firyal Ramzan
- Department of Biology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Mark A Brimble
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Patrick J Murphy
- Departments of Biology and Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brandon J Walters
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada
| | - Gilda Stefanelli
- Department of Biology, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| | - Iva B Zovkic
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, L5L 1C6, Canada.
| |
Collapse
|
21
|
Pereira M, Cruz MT, Fortuna A, Bicker J. Restoring the epigenome in Alzheimer's disease: advancing HDAC inhibitors as therapeutic agents. Drug Discov Today 2024; 29:104052. [PMID: 38830501 DOI: 10.1016/j.drudis.2024.104052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Current treatment options for Alzheimer's disease (AD) focus on symptom relief rather than halting disease progression. In this context, targeting histone deacetylation emerges as a promising therapeutic alternative. Dysregulation of histone deacetylase (HDAC) activity is present in AD, contributing to cognitive decline. Pharmacological HDAC inhibition has shown benefits in preclinical models, namely reduced amyloid beta plaque formation, lower phosphorylation and aggregation of tau protein, greater microtubule stability, less neuroinflammation, and improved metabolic homeostasis and cell survival. Nonetheless, clinical trials evidenced limitations such as insufficient selectivity or blood-brain barrier penetration. Hence, future innovative strategies are required to enhance their efficacy/safety.
Collapse
Affiliation(s)
- Márcia Pereira
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Maria Teresa Cruz
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
22
|
De Plano LM, Saitta A, Oddo S, Caccamo A. Epigenetic Changes in Alzheimer's Disease: DNA Methylation and Histone Modification. Cells 2024; 13:719. [PMID: 38667333 PMCID: PMC11049073 DOI: 10.3390/cells13080719] [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: 03/08/2024] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss, imposing a significant burden on affected individuals and their families. Despite the recent promising progress in therapeutic approaches, more needs to be done to understand the intricate molecular mechanisms underlying the development and progression of AD. Growing evidence points to epigenetic changes as playing a pivotal role in the pathogenesis of the disease. The dynamic interplay between genetic and environmental factors influences the epigenetic landscape in AD, altering gene expression patterns associated with key pathological events associated with disease pathogenesis. To this end, epigenetic alterations not only impact the expression of genes implicated in AD pathogenesis but also contribute to the dysregulation of crucial cellular processes, including synaptic plasticity, neuroinflammation, and oxidative stress. Understanding the complex epigenetic mechanisms in AD provides new avenues for therapeutic interventions. This review comprehensively examines the role of DNA methylation and histone modifications in the context of AD. It aims to contribute to a deeper understanding of AD pathogenesis and facilitate the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.S.); (S.O.)
| | | | | | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.S.); (S.O.)
| |
Collapse
|
23
|
Liu X, Li H, Wang Y, Li S, Ren W, Yuan J, Pang Y. Discovering common pathogenetic processes between tuberculosis and COVID-19 by bioinformatics and system biology approach. Heliyon 2024; 10:e28664. [PMID: 38596062 PMCID: PMC11002586 DOI: 10.1016/j.heliyon.2024.e28664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
Background SARS-CoV-2, the cause of the COVID-19 pandemic, poses a significant threat to humanity. Individuals with pulmonary tuberculosis (PTB) are at increased risk of developing severe COVID-19, due to long-term lung damage that heightens their susceptibility to full-blown disease. Methods Three COVID-19 datasets (GSE157103, GSE166253, and GSE171110) and one PTB dataset (GSE83456) were obtained from the Gene Expression Omnibus databases. Subsequently, data were subjected to weighted gene co-expression network analysis(WGCNA)followed by functional enrichment analysis using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. These analyses revealed two overlapping disease-specific modules, each comprising co-regulated genes with potentially related biological functions. Using Cytoscape, we visualised the interaction network containing common disease-related genes found within the intersection between modules and predicted transcription factors (TFs). Real-time qPCR was conducted to quantify expression levels of these genes in blood samples from COVID-19 and PTB patients. Finally, DisGeNET and the Drug Signatures database were employed to analyze these common genes, unveiling their connections to clinical disease features and potential drug treatments. Results Examination of the overlap between COVID-19 and PTB gene modules unveiled 11 common genes. Functional enrichment analyses using KEGG and GO shed light on potential functional relationships among these genes, providing insights into their potential roles in the heightened mortality of PTB patients due to SARS-CoV-2 infection. Furthermore, results of various bioinformatics-based analyses of common TFs and target genes led to identification of shared pathways and therapeutic targets for PTB patients with COVID-19, along with potential drug treatments for these patients. Conclusion Our results unveiled a potential biological connection between COVID-19 and PTB, as supported by results of functional enrichment analysis that highlighted potential biological processes and signaling pathways shared by both diseases. Building on these findings, we propose potential drug treatments for PTB patients with COVID-19, pending verification of drug safety and efficacy through laboratory and multicentre studies before clinical use.
Collapse
Affiliation(s)
| | | | | | - Shanshan Li
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Weicong Ren
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Jinfeng Yuan
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
24
|
Su Q, Ren YH, Liu GW, Gao YP, Zhang JX, Zhang JN, Pei XX, Li T. Trichostatin A relieves anxiety-and depression-like symptoms in APP/PS1 mice. Front Pharmacol 2024; 15:1333235. [PMID: 38572429 PMCID: PMC10987769 DOI: 10.3389/fphar.2024.1333235] [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: 11/04/2023] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Background Cognitive deficits and behavioral disorders such as anxiety and depression are common manifestations of Alzheimer's disease (AD). Our previous work demonstrated that Trichostatin A (TSA) could alleviate neuroinflammatory plaques and improve cognitive disorders. AD, anxiety, and depression are all associated with microglial inflammation. However, whether TSA could attenuate anxiety- and depression-like behaviors in APP/PS1 mice through anti-inflammatory signaling is still unclearly. Methods In the present study, all mice were subjected to the open field, elevated plus maze, and forced swim tests to assess anxiety- and depression-related behaviors after TSA administration. To understand the possible mechanisms underlying the behavioral effects observed, CST7 was measured in the hippocampus of mice and LPS-treated BV2 microglia. Results The results of this study indicated that TSA administration relieved the behaviors of depression and anxiety in APP/PS1 mice, and decreased CST7 levels in the hippocampus of APP/PS1 mice and LPS-induced BV2 cells. Conclusion Overall, these findings support the idea that TSA might be beneficial for reducing neurobehavioral disorders in AD and this could be due to suppression of CST7-related microglial inflammation.
Collapse
Affiliation(s)
- Qiang Su
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Yu-Hua Ren
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Guo-Wei Liu
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Yan-Ping Gao
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Jiu-Xuan Zhang
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Jin-Nan Zhang
- Department of Physiology, School of Basic Medicine, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Key Laboratory of Cell Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xia-Xia Pei
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi, China
| | - Tian Li
- Department of Physiology, School of Basic Medicine, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Key Laboratory of Cell Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
25
|
Zhang M, Wang W, Ye Q, Fu Y, Li X, Yang K, Gao F, Zhou A, Wei Y, Tian S, Li S, Wei F, Shi W, Li WD. Histone deacetylase inhibitors VPA and WT161 ameliorate the pathological features and cognitive impairments of the APP/PS1 Alzheimer's disease mouse model by regulating the expression of APP secretases. Alzheimers Res Ther 2024; 16:15. [PMID: 38245771 PMCID: PMC10799458 DOI: 10.1186/s13195-024-01384-0] [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: 07/08/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is a degenerative neurological disorder. Recent studies have indicated that histone deacetylases (HDACs) are among the most prominent epigenetic therapy targets and that HDAC inhibitors have therapeutic effects on AD. Here, we identified sodium valproate (VPA), a pan-HDAC inhibitor, and WT161, a novel HDAC6 selective inhibitor, as potential therapeutic agents for AD. Underlying molecular mechanisms were investigated. METHODS A cellular model, N2a-APPswe, was established via lentiviral infection, and the APPswe/PSEN1dE9 transgenic mouse model was employed in the study. LC-MS/MS was applied to quantify the concentration of WT161 in the mouse brain. Western blotting, immunohistochemical staining, thioflavin-S staining and ELISA were applied to detect protein expression in cells, tissues, or serum. RNA interference was utilized to knockdown the expression of specific genes in cells. The cognitive function of mice was assessed via the nest-building test, novel object recognition test and Morris water maze test. RESULTS Previous studies have focused mainly on the impact of HDAC inhibitors on histone deacetylase activity. Our study discovered that VPA and WT161 can downregulate the expression of multiple HDACs, such as HDAC1 and HDAC6, in both AD cell and mouse models. Moreover, they also affect the expression of APP and APP secretases (BACE1, PSEN1, ADAM10). RNA interference and subsequent vitamin C induction further confirmed that the expression of APP and APP secretases is indeed regulated by HDAC1 and HDAC6, with the JNK pathway being the intermediate link in this regulatory process. Through the above pathways, VPA and WT161 effectively reduced Aβ deposition in both AD cell and mouse models and significantly improved cognitive function in AD mice. CONCLUSIONS In general, we have discovered that the HDAC6-JNK-APP secretases cascade is an important pathway for VPA and WT161 to exert their therapeutic effects on AD. Investigations into the safety and efficacy of VPA and WT161 were also conducted, providing essential preclinical evidence for assessing these two epigenetic drugs for the treatment of AD.
Collapse
Affiliation(s)
- Miaomiao Zhang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Prenatal Diagnostic Center, Yiwu Maternity and Children Hospital, Yiwu, 322000, China
| | - Wanyao Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qun Ye
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yun Fu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350000, China
| | - Xuemin Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ke Yang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Fan Gao
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - An Zhou
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yonghui Wei
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shuang Tian
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Shen Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Fengjiang Wei
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Wentao Shi
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Wei-Dong Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
26
|
Wei B, Shi H, Yu X, Shi Y, Zeng H, Zhao Y, Zhao Z, Song Y, Sun M, Wang B. GR/Ahi1 regulates WDR68-DYRK1A binding and mediates cognitive impairment in prenatally stressed offspring. Cell Mol Life Sci 2024; 81:20. [PMID: 38195774 PMCID: PMC11073104 DOI: 10.1007/s00018-023-05075-1] [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: 08/09/2023] [Revised: 11/19/2023] [Accepted: 11/29/2023] [Indexed: 01/11/2024]
Abstract
Accumulating research shows that prenatal exposure to maternal stress increases the risk of behavioral and mental health problems for offspring later in life. However, how prenatal stress affects offspring behavior remains unknown. Here, we found that prenatal stress (PNS) leads to reduced Ahi1, decreased synaptic plasticity and cognitive impairment in offspring. Mechanistically, Ahi1 and GR stabilize each other, inhibit GR nuclear translocation, promote Ahi1 and WDR68 binding, and inhibit DYRK1A and WDR68 binding. When Ahi1 deletion or prenatal stress leads to hyperactivity of the HPA axis, it promotes the release of GC, leading to GR nuclear translocation and Ahi1 degradation, which further inhibits the binding of Ahi1 and WDR68, and promotes the binding of DYRK1A and WDR68, leading to elevated DYRK1A, reduced synaptic plasticity, and cognitive impairment. Interestingly, we identified RU486, an antagonist of GR, which increased Ahi1/GR levels and improved cognitive impairment and synaptic plasticity in PNS offspring. Our study contributes to understanding the signaling mechanisms of prenatal stress-mediated cognitive impairment in offspring.
Collapse
Affiliation(s)
- Bin Wei
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Haixia Shi
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xi Yu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yajun Shi
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Hongtao Zeng
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yan Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zejun Zhao
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yueyang Song
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Miao Sun
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Bin Wang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
27
|
Chen TJ, Hung HS, Cheng TL, Wang DC. Histone deacetylase inhibitor attenuates the effects of 27-hydroxycholesterol on the rat brain. Neurosci Lett 2024; 818:137533. [PMID: 37865186 DOI: 10.1016/j.neulet.2023.137533] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Hypercholesterolemia is a risk factor for Alzheimer's disease (AD). Plasma cholesterol does not pass the blood-brain barrier whereas its metabolite 27-hydroxycholesterol (27-OHC) can enter the brain. High 27-OHC in the brain has been suggested to mediate hypercholesterolemia-induced impairments of learning and memory through promoting amyloid-β accumulation and facilitating synaptic disruption. In AD brains, the activity of histone deacetylase (HDAC) is elevated. Treating AD animals with HDAC inhibitors decreases amyloid-β levels and synaptic damages, which leads to memory improvement. Whether HDAC activity is involved in the actions of 27-OHC is still uncertain. In this study, 4 weekly injections of 27-OHC/vehicle were given to rats followed by 3 daily injections of HDAC inhibitor trichostatin (TSA)/vehicle. The results of Morris water maze test reveal that all rats have intact spatial learning ability during the 5-d training phase. However, the behavioral performance during the probe trial was impaired by 27-OHC treatment, which was improved by adding TSA treatments. Furthermore, 27-OHC treatments reduced the hippocampal levels of acetylated histone H3, acetylated α tubulin, insulin-degrading enzyme and postsynaptic protein PSD-95, indicating that 27-OHC treatments may induce enhanced HDAC activity, decreased amyloid-β clearance and synaptic disruption. All reduced levels returned to the basal levels by adding TSA treatments. These findings support our hypothesis that HDAC activity is enhanced following long-term exposure to excess 27-OHC.
Collapse
Affiliation(s)
- Tsan-Ju Chen
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Hui-Shan Hung
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Lin Cheng
- Department of Physiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Dean-Chuan Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Sports Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
28
|
Wei W, Zhang Q, Jin T, Zhu L, Zhao J, Li F, Zhao S, Kong D, Hao J. Quantitative Proteomics Characterization of the Effect and Mechanism of Trichostatin A on the Hippocampus of Type II Diabetic Mice. Cell Mol Neurobiol 2023; 43:4309-4332. [PMID: 37864628 PMCID: PMC11407725 DOI: 10.1007/s10571-023-01424-7] [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: 07/28/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023]
Abstract
Diabetic encephalopathy (DE) is one of the complications of diabetes mellitus with mild-to-moderate cognitive impairment. Trichostatin A (TSA) has been revealed to show protective effect on central nervous systems in Alzheimer's disease (AD) and hypoxic-ischemic brain injury. However, the effect and molecular mechanism of TSA on cognitive function of DE are unknown. Here, we demonstrated that cognitive function was damaged in diabetic mice versus normal mice and treatment with TSA improved cognitive function in diabetic mice. Proteomic analysis of the hippocampus revealed 174 differentially expressed proteins in diabetic mice compared with normal mice. TSA treatment reversed the expression levels of 111 differentially expressed proteins grouped into functional clusters, including the longevity regulating pathway, the insulin signaling pathway, peroxisomes, protein processing in the endoplasmic reticulum, and ribosomes. Furthermore, protein-protein interaction network analysis of TSA-reversed proteins revealed that UBA52, CAT, RPL29, RPL35A, CANX, RPL37, and PRKAA2 were the main hub proteins. Multiple KEGG pathway-enriched CAT and PRKAA2 levels were significantly decreased in the hippocampus of diabetic mice versus normal mice, which was reversed by TSA administration. Finally, screening for potential similar or ancillary drugs for TSA treatment indicated that HDAC inhibitors ISOX, apicidin, and panobinostat were the most promising similar drugs, and the PI3K inhibitor GSK-1059615, the Aurora kinase inhibitor alisertib, and the nucleophosmin inhibitor avrainvillamide-analog-6 were the most promising ancillary drugs. In conclusion, our study revealed that CAT and PRKAA2 were the key proteins involved in the improvement of DE after TSA treatment. ISOX, apicidin, and panobinostat were promising similar drugs and that GSK-1059615, alisertib, and avrainvillamide-analog-6 were promising ancillary drugs to TSA in the treatment of DE.
Collapse
Affiliation(s)
- Wandi Wei
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Qingning Zhang
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tingting Jin
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Lin Zhu
- Department of Electromyogram, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jialing Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Fan Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Shijiazhuang, China
| | - Song Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China
| | - Dezhi Kong
- Department of Pharmacology of Chinese Materia Medica, Institution of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
- Center of Metabolic Diseases and Cancer Research, Institute of Medical and Health Science of Hebei Medical University, Shijiazhuang, China.
- Hebei Key Laboratory of Forensic Medicine, Shijiazhuang, China.
| |
Collapse
|
29
|
Jin M, Wei Z, Ramalingam N, Xiao M, Xu A, Yu X, Song Q, Liu W, Zhao J, Zhang D, Selkoe DJ, Li S. Activation of β 2-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28:4877-4888. [PMID: 37365243 DOI: 10.1038/s41380-023-02145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of β2-adrenergic receptor (β2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid β-protein (oAβ). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aβ-species. We found that pharmacological activation of β2-AR, but not β1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAβ-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in β2-AR knockout mice, suggesting that activating β2-AR prevents oAβ-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of β-ARs each decreased HDAC2, whereas Aβ oligomers increased HDAC2 levels in the hippocampus. Further, oAβ-induced inflammatory effects and neurite degeneration were prevented by either β2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of β2-AR is a novel potential therapeutic strategy to mitigate oAβ-mediated features of AD.
Collapse
Affiliation(s)
- Ming Jin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Meng Xiao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China
| | - Anqi Xu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Xiaohan Yu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Qingyang Song
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zhao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Henan Key Laboratory of Neurorestoratology, Xinxiang, Henan, 453100, China
| | - Dainan Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
30
|
Braz SO, Morgado MM, Pereira MI, Monteiro AC, Golonzhka O, Jarpe M, Brites P, Sousa MM, Nogueira-Rodrigues J. HDAC-6 inhibition ameliorates the early neuropathology in a mouse model of Krabbe disease. Front Mol Neurosci 2023; 16:1231659. [PMID: 37588057 PMCID: PMC10426153 DOI: 10.3389/fnmol.2023.1231659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/12/2023] [Indexed: 08/18/2023] Open
Abstract
Introduction In Krabbe disease (KD), mutations in β-galactosylceramidase (GALC), a lysosomal enzyme responsible for the catabolism of galactolipids, leads to the accumulation of its substrates galactocerebroside and psychosine. This neurologic condition is characterized by a severe and progressive demyelination together with neuron-autonomous defects and degeneration. Twitcher mice mimic the infantile form of KD, which is the most common form of the human disease. The Twitcher CNS and PNS present demyelination, axonal loss and neuronal defects including decreased levels of acetylated tubulin, decreased microtubule stability and impaired axonal transport. Methods We tested whether inhibiting the α-tubulin deacetylase HDAC6 with a specific inhibitor, ACY-738, was able to counteract the early neuropathology and neuronal defects of Twitcher mice. Results Our data show that delivery of ACY-738 corrects the low levels of acetylated tubulin in the Twitcher nervous system. Furthermore, it reverts the loss myelinated axons in the sciatic nerve and in the optic nerve when administered from birth to postnatal day 9, suggesting that the drug holds neuroprotective properties. The extended delivery of ACY-738 to Twitcher mice delayed axonal degeneration in the CNS and ameliorated the general presentation of the disease. ACY-738 was effective in rescuing neuronal defects of Twitcher neurons, stabilizing microtubule dynamics and increasing the axonal transport of mitochondria. Discussion Overall, our results support that ACY-738 has a neuroprotective effect in KD and should be considered as an add-on therapy combined with strategies targeting metabolic correction.
Collapse
Affiliation(s)
- Sandra O. Braz
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Marlene M. Morgado
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Marta I. Pereira
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Ana C. Monteiro
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Olga Golonzhka
- Acetylon Pharmaceuticals Inc., Boston, MA, United States
| | - Matthew Jarpe
- Acetylon Pharmaceuticals Inc., Boston, MA, United States
| | - Pedro Brites
- NeuroLipid Biology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Monica M. Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joana Nogueira-Rodrigues
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
31
|
de la Fuente AG, Pelucchi S, Mertens J, Di Luca M, Mauceri D, Marcello E. Novel therapeutic approaches to target neurodegeneration. Br J Pharmacol 2023; 180:1651-1673. [PMID: 36965025 PMCID: PMC10952850 DOI: 10.1111/bph.16078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Ageing is the main risk factor common to most primary neurodegenerative disorders. Indeed, age-related brain alterations have been long considered to predispose to neurodegeneration. Although protein misfolding and the accumulation of toxic protein aggregates have been considered as causative events in neurodegeneration, several other biological pathways affected by brain ageing also contribute to pathogenesis. Here, we discuss the evidence showing the involvement of the mechanisms controlling neuronal structure, gene expression, autophagy, cell metabolism and neuroinflammation in the onset and progression of neurodegenerative disorders. Furthermore, we review the therapeutic strategies currently under development or as future approaches designed to normalize these pathways, which may then increase brain resilience to cope with toxic protein species. In addition to therapies targeting the insoluble protein aggregates specifically associated with each neurodegenerative disorder, these novel pharmacological approaches may be part of combined therapies designed to rescue brain function.
Collapse
Affiliation(s)
- Alerie G. de la Fuente
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Instituto de Neurociencias CSIC‐UMHAlicanteSpain
- Wellcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Daniela Mauceri
- Institute of Anatomy and Cell BiologyDepartment of Molecular and Cellular Neuroscience, University of MarburgMarburgGermany
- Department of NeurobiologyInterdisciplinary Centre for Neurosciences (IZN), Heidelberg UniversityHeidelbergGermany
| | - Elena Marcello
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
32
|
Aleksandrova Y, Munkuev A, Mozhaitsev E, Suslov E, Tsypyshev D, Chaprov K, Begunov R, Volcho K, Salakhutdinov N, Neganova M. Elaboration of the Effective Multi-Target Therapeutic Platform for the Treatment of Alzheimer's Disease Based on Novel Monoterpene-Derived Hydroxamic Acids. Int J Mol Sci 2023; 24:ijms24119743. [PMID: 37298694 DOI: 10.3390/ijms24119743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Novel monoterpene-based hydroxamic acids of two structural types were synthesized for the first time. The first type consisted of compounds with a hydroxamate group directly bound to acyclic, monocyclic and bicyclic monoterpene scaffolds. The second type included hydroxamic acids connected with the monoterpene moiety through aliphatic (hexa/heptamethylene) or aromatic linkers. An in vitro analysis of biological activity demonstrated that some of these molecules had powerful HDAC6 inhibitory activity, with the presence of a linker area in the structure of compounds playing a key role. In particular, it was found that hydroxamic acids containing a hexa- and heptamethylene linker and (-)-perill fragment in the Cap group exhibit excellent inhibitory activity against HDAC6 with IC50 in the submicromolar range from 0.56 ± 0.01 µM to 0.74 ± 0.02 µM. The results of the study of antiradical activity demonstrated the presence of moderate ability for some hydroxamic acids to scavenge 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2ROO• radicals. The correlation coefficient between the DPPH radical scavenging activity and oxygen radical absorbance capacity (ORAC) value was R2 = 0.8400. In addition, compounds with an aromatic linker based on para-substituted cinnamic acids, having a monocyclic para-menthene skeleton as a Cap group, 35a, 38a, 35b and 38b, demonstrated a significant ability to suppress the aggregation of the pathological β-amyloid peptide 1-42. The 35a lead compound with a promising profile of biological activity, discovered in the in vitro experiments, demonstrated neuroprotective effects on in vivo models of Alzheimer's disease using 5xFAD transgenic mice. Together, the results obtained demonstrate a potential strategy for the use of monoterpene-derived hydroxamic acids for treatment of various aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| | - Aldar Munkuev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Evgenii Mozhaitsev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Evgenii Suslov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Dmitry Tsypyshev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Kirill Chaprov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| | - Roman Begunov
- Biology and Ecology Faculty of P. G. Demidov Yaroslavl State University, Matrosova Ave., 9, Yaroslavl 150003, Russia
| | - Konstantin Volcho
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Nariman Salakhutdinov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, Novosibirsk 630090, Russia
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, Chernogolovka 142432, Russia
| |
Collapse
|
33
|
Histone Modifications in Alzheimer's Disease. Genes (Basel) 2023; 14:genes14020347. [PMID: 36833274 PMCID: PMC9956192 DOI: 10.3390/genes14020347] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Since Late-onset Alzheimer's disease (LOAD) derives from a combination of genetic variants and environmental factors, epigenetic modifications have been predicted to play a role in the etiopathology of LOAD. Along with DNA methylation, histone modifications have been proposed as the main epigenetic modifications that contribute to the pathologic mechanisms of LOAD; however, little is known about how these mechanisms contribute to the disease's onset or progression. In this review, we highlighted the main histone modifications and their functional role, including histone acetylation, histone methylation, and histone phosphorylation, as well as changes in such histone modifications that occur in the aging process and mainly in Alzheimer's disease (AD). Furthermore, we pointed out the main epigenetic drugs tested for AD treatment, such as those based on histone deacetylase (HDAC) inhibitors. Finally, we remarked on the perspectives around the use of such epigenetics drugs for treating AD.
Collapse
|
34
|
TFEB acetylation promotes lysosome biogenesis and ameliorates Alzheimer's disease-relevant phenotypes in mice. J Biol Chem 2022; 298:102649. [PMID: 36441024 PMCID: PMC9694136 DOI: 10.1016/j.jbc.2022.102649] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
Lysosomes are one of the major centers for regulating cargo degradation and protein quality control. Transcription factor EB (TFEB)-promoted lysosome biogenesis enhances lysosome-mediated degradation and alleviates neurodegenerative diseases, but the mechanisms underlying TFEB modification and activation are still poorly understood. Here, we report essential roles of TFEB acetylation in TFEB nuclear translocation and lysosome biogenesis, which are independent of TFEB dephosphorylation. By screening small molecules, we find that Trichostatin A (TSA), the pan-inhibitor of histone deacetylases (HDACs), promotes nuclear translocation of TFEB. TSA enhances the staining of cells by LysoTracker Red and increases the expression of lysosomal and autophagic genes. We identify four novel acetylated lysine residues in TFEB, which are important for TFEB nuclear translocation and lysosome biogenesis. We show that TFEB acetylation is regulated by HDACs (HDAC5, HDAC6, and HDAC9) and lysine acetyltransferases (KATs), including ELP3, CREBBP, and HAT1. During TSA-induced cytosol-to-nucleus translocation of TFEB, acetylation is independent of TFEB dephosphorylation, since the mTORC1- or GSK3β-related phosphorylation sites on TFEB are still phosphorylated. Administration of TSA to APP/PS1 mice increases the expression of lysosomal and autophagic genes in mouse brains and also improves memory. Accordingly, the β-amyloid plaque burden is decreased. These results show that the acetylation of TFEB, as a novel mechanism of TFEB activation, promotes lysosome biogenesis and alleviates the pathogenesis of Alzheimer's disease. Our results also suggest that HDAC inhibition can promote lysosome biogenesis, and this may be a potential therapeutic approach for the treatment of neurodegenerative diseases and disorders related to HDAC hyperactivation.
Collapse
|
35
|
Li C, Ren J, Zhang M, Wang H, Yi F, Wu J, Tang Y. The heterogeneity of microglial activation and its epigenetic and non-coding RNA regulations in the immunopathogenesis of neurodegenerative diseases. Cell Mol Life Sci 2022; 79:511. [PMID: 36066650 PMCID: PMC11803019 DOI: 10.1007/s00018-022-04536-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 12/15/2022]
Abstract
Microglia are resident immune cells in the brain and play a central role in the development and surveillance of the nervous system. Extensive gliosis is a common pathological feature of several neurodegenerative diseases, such as Alzheimer's disease (AD), the most common cause of dementia. Microglia can respond to multiple inflammatory insults and later transform into different phenotypes, such as pro- and anti-inflammatory phenotypes, thereby exerting different functions. In recent years, an increasing number of studies based on both traditional bulk sequencing and novel single-cell/nuclear sequencing and multi-omics analysis, have shown that microglial phenotypes are highly heterogeneous and dynamic, depending on the severity and stage of the disease as well as the particular inflammatory milieu. Thus, redirecting microglial activation to beneficial and neuroprotective phenotypes promises to halt the progression of neurodegenerative diseases. To this end, an increasing number of studies have focused on unraveling heterogeneous microglial phenotypes and their underlying molecular mechanisms, including those due to epigenetic and non-coding RNA modulations. In this review, we summarize the epigenetic mechanisms in the form of DNA and histone modifications, as well as the general non-coding RNA regulations that modulate microglial activation during immunopathogenesis of neurodegenerative diseases and discuss promising research approaches in the microglial era.
Collapse
Affiliation(s)
- Chaoyi Li
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jie Ren
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mengfei Zhang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huakun Wang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Fang Yi
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Junjiao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Tang
- Aging Research Center, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, Hunan, China.
- The Biobank of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
36
|
Chen ZH, Li J, Zhao XX, Yang XH, Li J, Zou B, Zang WB, Ma RX, Wang YF, Yao Y. Saikogenin F From Bupleurum smithii Ameliorates Learning and Memory Impairment via Antiinflammation Effect in an Alzheimer’s Disease Mouse Model. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221111029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease associated with aging. Bupleurum smithii Wolff. is a Chinese folk medicine used to reduce fever and inflammation. Regarding the key role of neuroinflammation in AD pathogenesis, it was speculated that B. smithii may be the source of compounds that treat AD through anti-inflammatory effects. This study aimed to investigate the effects of saikogenin F, a natural active ingredient from B. smithii, on cognition impairment and neuroinflammation in AD mice induced by amyloid β (Aβ). The AD mice model was established by intracerebroventricular (i.c.v.) injection of Aβ, and different doses of saikogenin F (10, 20, and 40 mg/kg) were intragastrically administrated once daily. Results of behavioral experiments, including the novel object recognition (NOR) test, Y-maze test, and Morris water maze (MWZ) test, showed that saikogenin F could ameliorate Aβ-induced cognition impairment in AD mice. Enzyme linked immunosorbent assay (ELISA) results showed that tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and reactive oxygen species (ROS) levels in hippocampal tissue increased after Aβ injection, while saikogenin F could significantly reduce the concentrations of these inflammatory factors. Western blotting results revealed that the Aβ-induced reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits protein expression in mice hippocampus was remarkably downregulated by saikogenin F. Results of Iba-1 immunohistochemical staining showed that saikogenin F could effectively inhibit Aβ-induced activation of microglia in vivo. These results suggested that saikogenin F could relieve Aβ-induced cognitive impairment via inhibiting neuroinflammation and microglial activation. These effects may be achieved by inhibiting the expression of the NADPH oxidase subunits gp91phox and p47phox.
Collapse
Affiliation(s)
- Ze-Hui Chen
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Juan Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Ningxia Engineering and Technology Research Center for Modernization of Characteristic Chinese Medicine, and Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Xun-Xia Zhao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xin-He Yang
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jia Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Bin Zou
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Wei-Biao Zang
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Rui-Xia Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Ye-Feng Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
37
|
Ube2c-inhibition alleviated amyloid pathology and memory deficits in APP/PS1 mice model of AD. Prog Neurobiol 2022; 215:102298. [PMID: 35671859 DOI: 10.1016/j.pneurobio.2022.102298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/24/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
Autophagy is a major intracellular degradation pathway for the clearance of damaged organelles and misfolded peptides. Previous studies have indicated that autophagy is involved in the pathogenesis of neurodegenerative disease including Alzheimer's disease (AD). Defective autophagy and highly expressed ubiquitin-conjugating enzyme 2C (Ube2c) have been found in AD patients and mouse. However, little is known about the regulation of autophagy in AD. The association of Ube2c with autophagy, amyloid pathology and cognitive deficits in AD remains unclear. In the present study, we characterized over expression of Ube2c and declined autophagy in amyloid β (Aβ)-treated microglia and demonstrated the protective effects of agomelatine (AGO) in APP/PS1 mice. We found that knockdown of Ube2c with AAV2 encoding shUbe2c resulted in an obvious enhancement of autophagy in BV2 microglia cells, and an alleviation of Aβ pathology and memory deficits in APP/PS1 mice. Further, pharmacological inhibition of Ube2c by AGO significantly reduced Aβ plaques, improved synaptic plasticity and cognitive behaviors in APP/PS1 mice, as well as promoted autophagy in microglia. Our findings uncover a potent role of Ube2c over-expression and autophagy decline in the pathogenesis of AD, and suggest that regulation of Ube2c and autophagy may provide an important clue and a potential target for the novel therapeutics of AD.
Collapse
|
38
|
Pushparaj PN, Kalamegam G, Wali Sait KH, Rasool M. Decoding the Role of Astrocytes in the Entorhinal Cortex in Alzheimer’s Disease Using High-Dimensional Single-Nucleus RNA Sequencing Data and Next-Generation Knowledge Discovery Methodologies: Focus on Drugs and Natural Product Remedies for Dementia. Front Pharmacol 2022; 12:720170. [PMID: 35295737 PMCID: PMC8918735 DOI: 10.3389/fphar.2021.720170] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction: Alzheimer’s disease (AD) is a major cause of the development of cognitive decline and dementia. AD and associated dementias (ADRD) are the major contributors to the enormous burden of morbidity and mortality worldwide. To date, there are no robust therapies to alleviate or cure this debilitating disease. Most drug treatments focus on restoring the normal function of neurons and the cells that cause inflammation, such as microglia in the brain. However, the role of astrocytes, the brain’s housekeeping cells, in the development of AD and the initiation of dementia is still not well understood. Objective: To decipher the role of astrocytes in the entorhinal cortex of AD patients using single nuclear RNA sequencing (snRNASeq) datasets from the Single Cell RNA-seq Database for Alzheimer’s Disease (scREAD). The datasets were originally derived from astrocytes, isolated from the entorhinal cortex of AD brain and healthy brain to decipher disease-specific signaling pathways as well as drugs and natural products that reverse AD-specific signatures in astrocytes. Methods: We used snRNASeq datasets from the scREAD database originally derived from astrocytes isolated from the entorhinal cortex of AD and healthy brains from the Gene Expression Omnibus (GEO) (GSE138852 and GSE147528) and analyzed them using next-generation knowledge discovery (NGKD) platforms. scREAD is a user-friendly open-source interface available at https://bmbls.bmi.osumc.edu/scread/that enables more discovery-oriented strategies. snRNASeq data and metadata can also be visualized and downloaded via an interactive web application at adsn.ddnetbio.com. Differentially expressed genes (DEGs) for each snRNASeq dataset were analyzed using iPathwayGuide to compare and derive disease-specific pathways, gene ontologies, and in silico predictions of drugs and natural products that regulate AD -specific signatures in astrocytes. In addition, DEGs were analyzed using the L1000FWD and L1000CDS2 signature search programming interfaces (APIs) to identify additional drugs and natural products that mimic or reverse AD-specific gene signatures in astrocytes. Results: We found that PI3K/AKT signaling, Wnt signaling, neuroactive ligand-receptor interaction pathways, neurodegeneration pathways, etc. were significantly impaired in astrocytes from the entorhinal cortex of AD patients. Biological processes such as glutamate receptor signaling pathway, regulation of synapse organization, cell-cell adhesion via plasma membrane adhesion molecules, and chylomicrons were negatively enriched in the astrocytes from the entorhinal cortex of AD patients. Gene sets involved in cellular components such as postsynaptic membrane, synaptic membrane, postsynapse, and synapse part were negatively enriched (p < 0.01). Moreover, molecular functions such as glutamate receptor activity, neurotransmitter receptor activity, and extracellular ligand-gated ion channels were negatively regulated in the astrocytes of the entorhinal cortex of AD patients (p < 0.01). Moreover, the application of NGKD platforms revealed that antirheumatic drugs, vitamin-E, emetine, narciclasine, cephaeline, trichostatin A, withaferin A, dasatinib, etc. can potentially reverse gene signatures associated with AD. Conclusions: The present study highlights an innovative approach to use NGKD platforms to find unique disease-associated signaling pathways and specific synthetic drugs and natural products that can potentially reverse AD and ADRD-associated gene signatures.
Collapse
Affiliation(s)
- Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- *Correspondence: Peter Natesan Pushparaj, ; Mahmood Rasool,
| | - Gauthaman Kalamegam
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Khalid Hussain Wali Sait
- Department of Obstetrics and Gynaecology, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Peter Natesan Pushparaj, ; Mahmood Rasool,
| |
Collapse
|
39
|
Sharma R, Sharma S, Thakur A, Singh A, Singh J, Nepali K, Liou JP. The Role of Epigenetic Mechanisms in Autoimmune, Neurodegenerative, Cardiovascular, and Imprinting Disorders. Mini Rev Med Chem 2022; 22:1977-2011. [PMID: 35176978 DOI: 10.2174/1389557522666220217103441] [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: 09/10/2021] [Revised: 10/01/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic mutations like aberrant DNA methylation, histone modifications, or RNA silencing are found in a number of human diseases. This review article discusses the epigenetic mechanisms involved in neurodegenerative disorders, cardiovascular disorders, auto-immune disorder, and genomic imprinting disorders. In addition, emerging epigenetic therapeutic strategies for the treatment of such disorders are presented. Medicinal chemistry campaigns highlighting the efforts of the chemists invested towards the rational design of small molecule inhibitors have also been included. Pleasingly, several classes of epigenetic inhibitors, DNMT, HDAC, BET, HAT, and HMT inhibitors along with RNA based therapies have exhibited the potential to emerge as therapeutics in the longer run. It is quite hopeful that epigenetic modulator-based therapies will advance to clinical stage investigations by leaps and bounds.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Arshdeep Singh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jagjeet Singh
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia.,Department of Pharmacy, Rayat-Bahara Group of Institutes, Hoshiarpur, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
40
|
Li Y, Sang S, Ren W, Pei Y, Bian Y, Chen Y, Sun H. Inhibition of Histone Deacetylase 6 (HDAC6) as a therapeutic strategy for Alzheimer's disease: A review (2010-2020). Eur J Med Chem 2021; 226:113874. [PMID: 34619465 DOI: 10.1016/j.ejmech.2021.113874] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/13/2021] [Accepted: 09/25/2021] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which is characterized by the primary risk factor, age. Several attempts have been made to treat AD, while most of them end in failure. However, with the deepening study of pathogenesis of AD, the expression of HDAC6 in the hippocampus, which plays a major role of the memory formation, is becoming worth of notice. Neurofibrillary tangles (NFTs), a remarkable lesion in AD, has been characterized in association with the abnormal accumulation of hyperphosphorylated Tau, which is mainly caused by the high expression of HDAC6. On the other hand, the hypoacetylated tubulin induced by HDAC6 is also fatal for the neuronal transport, which is the key impact of the formation of axons and dendrites. Overall, the significantly increased expression of HDAC6 in brain regions is deleterious to neuron survival in AD patients. Based on the above research, the inhibition of HDAC6 seems to be a potential therapeutic method for the treatment of AD. Up to now, various types of HDAC6 inhibitors have been discovered. This review mainly analyzes the HDAC6 inhibitors reported amid 2010-2020 in terms of their structure, selectivity and pharmacological impact towards AD. And we aim at facilitating the design and development of better HDAC6 inhibitors in the future.
Collapse
Affiliation(s)
- Yunheng Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shenghu Sang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weijie Ren
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yaoyao Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Haopeng Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
41
|
Kaya-Tilki E, Dikmen M. Neuroprotective effects of some epigenetic modifying drugs' on Chlamydia pneumoniae-induced neuroinflammation: A novel model. PLoS One 2021; 16:e0260633. [PMID: 34847172 PMCID: PMC8631675 DOI: 10.1371/journal.pone.0260633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/14/2021] [Indexed: 12/02/2022] Open
Abstract
Chlamydia pneumoniae (Cpn) is a gram-negative intracellular pathogen that causes a variety of pulmonary diseases, and there is growing evidence that it may play a role in Alzheimer's disease (AD) pathogenesis. Cpn can interact functionally with host histones, altering the host's epigenetic regulatory system by introducing bacterial products into the host tissue and inducing a persistent inflammatory response. Because Cpn is difficult to propagate, isolate, and detect, a modified LPS-like neuroinflammation model was established using lyophilized cell free supernatant (CFS) obtained from infected cell cultures, and the effects of CFS were compared to LPS. The neuroprotective effects of Trichostatin A (TSA), givinostat, and RG108, which are effective on epigenetic mechanisms, and the antibiotic rifampin, were studied in this newly introduced model and in the presence of amyloid beta (Aβ) 1-42. The neuroprotective effects of the drugs, as well as the effects of CFS and LPS, were evaluated in Aβ-induced neurotoxicity using a real-time cell analysis system, total ROS, and apoptotic impact. TSA, RG108, givinostat, and rifampin all demonstrated neuroprotective effects in both this novel model and Aβ-induced neurotoxicity. The findings are expected to provide early evidence on neuroprotective actions against Cpn-induced neuroinflammation and Aβ-induced neurotoxicity, which could represent a new treatment option for AD, for which there are currently few treatment options.
Collapse
Affiliation(s)
- Elif Kaya-Tilki
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
42
|
Chen F, Yang D, Cheng XY, Yang H, Yang XH, Liu HT, Wang R, Zheng P, Yao Y, Li J. Astragaloside IV Ameliorates Cognitive Impairment and Neuroinflammation in an Oligomeric Aβ Induced Alzheimer's Disease Mouse Model via Inhibition of Microglial Activation and NADPH Oxidase Expression. Biol Pharm Bull 2021; 44:1688-1696. [PMID: 34433707 DOI: 10.1248/bpb.b21-00381] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Microglial activation and neuroinflammation induced by amyloid β (Aβ) play pivotal roles in Alzheimer's disease (AD) pathogenesis. Astragaloside IV (AS-IV) is one of the major active compounds of the traditional Chinese medicine Astmgali Radix. It has been reported that AS-IV could protect against Aβ-induced neuroinflammation and cognitive impairment, but the underlying mechanisms need to be further clarified. In this study, the therapeutic effects of AS-IV were investigated in an oligomeric Aβ (oAβ) induced AD mice model. The effects of AS-IV on microglial activation, neuronal damage and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression were further studied. Different doses of AS-IV were administered intragastrically once a day after intracerebroventricularly oAβ injection. Results of behavioral experiments including novel object recognition (NOR) test and Morris water maze (MWM) test revealed that AS-IV administration could significantly ameliorate oAβ-induced cognitive impairment in a dose dependent manner. Enzyme linked immunosorbent assay (ELISA) results showed that increased levels of reactive oxygen species (ROS), tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β) and IL-6 in hippocampal tissues induced by oAβ injection were remarkably inhibited after AS-IV treatment. OAβ induced microglial activation and neuronal damage was significantly suppressed in AS-IV-treated mice brain, observed in immunohistochemistry results. Furthermore, oAβ upregulated protein expression of NADPH oxidase subunits gp91phox, p47phox, p22phox and p67phox were remarkably reduced by AS-IV in Western blotting assay. These results revealed that AS-IV could ameliorate oAβ-induced cognitive impairment, neuroinflammation and neuronal damage, which were possibly mediated by inhibition of microglial activation and down-regulation of NADPH oxidase protein expression. Our findings provide new insights of AS-IV for the treatment of neuroinflammation related diseases such as AD.
Collapse
Affiliation(s)
- Fei Chen
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| | - Dan Yang
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| | - Xiao-Yu Cheng
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, the Second Affiliated Hospital, Soochow University
| | - Hui Yang
- Research Center of Medical Science and Technology, Ningxia Medical University
| | - Xin-He Yang
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| | - He-Tao Liu
- School of Basic Medical Sciences, Ningxia Medical University
| | - Rui Wang
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| | - Ping Zheng
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University
| | - Juan Li
- School of Pharmacy, Ningxia Engineering and Technology Research Center for Modernization of Traditional Chinese Medicine, and Key Laboratory of Traditional Chinese Medicine Modernization, Ministry of Education, Ningxia Medical University
| |
Collapse
|
43
|
Epigenetic Modulation of Microglia Function and Phenotypes in Neurodegenerative Diseases. Neural Plast 2021; 2021:9912686. [PMID: 34194489 PMCID: PMC8181095 DOI: 10.1155/2021/9912686] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia-mediated neuroinflammation is one of the most remarkable hallmarks of neurodegenerative diseases (NDDs), including AD, PD, and ALS. Accumulating evidence indicates that microglia play both neuroprotective and detrimental roles in the onset and progression of NDDs. Yet, the specific mechanisms of action surrounding microglia are not clear. Modulation of microglia function and phenotypes appears to be a potential strategy to reverse NDDs. Until recently, research into the epigenetic mechanisms of diseases has been gradually developed, making it possible to elucidate the molecular mechanisms underlying the epigenetic regulation of microglia in NDDs. This review highlights the function and phenotypes of microglia, elucidates the relationship between microglia, epigenetic modifications, and NDDs, as well as the possible mechanisms underlying the epigenetic modulation of microglia in NDDs with a focus on potential intervention strategies.
Collapse
|