1
|
Almohmadi NH, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Abdelaziz AM, Jabir MS, Alexiou A, Papadakis M, Batiha GES. Glutamatergic dysfunction in neurodegenerative diseases focusing on Parkinson's disease: Role of glutamate modulators. Brain Res Bull 2025; 225:111349. [PMID: 40252703 DOI: 10.1016/j.brainresbull.2025.111349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder resulting from the degeneration of dopamenergic neurons in the substantia nigra pars compacta (SNpc). Research has predominantly centered on understanding the dysfunction of dopaminergic neurotransmission in PD. Recently, more studies discussed the potential role of other neurotransmitters in PD neuropathology. One of the most important non-dopaminergic neurotransmitters involved in the pathogenesis of PD is glutamate, which is widely involved in glutamatergic neurotransmission in different brain regions, including SNpc. The development and progression of PD neuropathology and levodopa-induced dyskinesias (LID) are associated with glutamate neurotoxicity. Therefore, this review seeks to explore the possible involvement of glutamatergic signaling in PD development and assess the therapeutic potential of glutamate receptor antagonists in treating the disorder.
Collapse
Affiliation(s)
- Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq; Jabir ibn Hayyan Medical University Al-Ameer Qu, Po. Box (13), Kufa, Najaf, Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq.
| | - Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University-Arish Branch, Arish 45511, Egypt.
| | - Majid S Jabir
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia; University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Research & Development, Funogen, Athens, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten, Herdecke, Heusnerstrasse 40, Wuppertal 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, AlBeheira 22511, Egypt.
| |
Collapse
|
2
|
Lu Z, Jiang Z, Huang X, Chen Y, Feng L, Mai J, Lao L, Li L, Chen WH, Hu J. Anti-Alzheimer effects of an HDAC6 inhibitor, WY118, alone and in combination of lithium chloride: Synergistic suppression of ferroptosis via the modulation of tau phosphorylation and MAPK signaling. Eur J Pharmacol 2025; 997:177605. [PMID: 40204225 DOI: 10.1016/j.ejphar.2025.177605] [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: 02/06/2025] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder, and current therapies mainly offer symptomatic relief. Given that the pathophysiology of AD is multifaceted, a multimodal therapeutic strategy targeting multiple molecular pathways implicated in AD-related pathogenesis represents a pragmatic avenue for impeding the advancement of AD. In this study, we evaluated the anti-Alzheimer effects of an HDAC6 inhibitor WY118, both alone and in combination with lithium chloride (LiCl), a GSK-3β inhibitor, to synergistically suppress ferroptosis. The combination of compound WY118 and LiCl demonstrated significant synergistic effects in both cellular models of AD induced by glutamate and streptozotocin. The findings suggest that compound WY118, in particular in combination with LiCl, exhibits potent anti-Alzheimer effects by synergistically suppressing ferroptosis. Studies on the mechanism of action indicated that the combination treatment significantly reduced tau phosphorylation and inhibited p38 MAPK signaling. This combination therapy holds promise for developing more effective treatments for AD.
Collapse
Affiliation(s)
- Zhonghui Lu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Zixing Jiang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Xiaoling Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Yu Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Luanqi Feng
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Jielin Mai
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Linghui Lao
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Lanqing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China.
| | - Jinhui Hu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, 529020, PR China.
| |
Collapse
|
3
|
Kalra P, Grewal AK, Khan H, Singh TG. Unscrambling the cellular and molecular threads of Neuroplasticity: Insights into Alzheimer's disease pathogenesis. Neuroscience 2025; 571:74-88. [PMID: 39970983 DOI: 10.1016/j.neuroscience.2025.02.037] [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: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is predominantly the most recurring and devastating neurological condition among the elderly population, characterized by the accumulation of amyloid-β (Aβ) and phosphorylated tau proteins, and is accompanied by progressive decline of learning and memory. Due to its complex and multifactorial etiology, a wide variety of therapeutic interventions have been developed. Despite constant advancements in the field, effective treatments that ameliorate the severity of Alzheimer's symptoms or cease their progression are still insufficient. Mounting evidence suggests that synaptic dysfunction could be an essential component of AD pathogenesis as synapse signaling is impaired in the aging brain, which contributes to synaptic decline. Therefore, improving neuroplasticity such as synaptic plasticity or neurogenesis could be a promising therapeutic approach for alleviating the effects of AD. This article reviews the cellular and molecular threads of neuroplasticity as well as targets that restore neuronal survival and plasticity to provide functional recoveries, including receptors, downstream signaling pathways, ion channels, transporters, enzymes, and neurotrophic factors.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; University School of Pharmaceutical Sciences, Rayat Bahra University, Mohali, Punjab 140103, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
4
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
5
|
Soni P, Sharma SM, Pieper AA, Paul BD, Thomas B. Nrf2/Bach1 signaling axis: A promising multifaceted therapeutic strategy for Alzheimer's disease. Neurotherapeutics 2025; 22:e00586. [PMID: 40199685 DOI: 10.1016/j.neurot.2025.e00586] [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: 10/31/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, which continues to elude effective treatment despite decades of research and numerous clinical trials. While existing therapeutic strategies have primarily targeted neuropathological hallmarks such as amyloid plaques and tau tangles, they have failed to halt disease progression, leaving patients with limited options. This persistent failure reveals a critical gap in our understanding of AD and calls for a fresh perspective - one that goes beyond the traditional targets and dives deeper into the fundamental cellular processes that drive neurodegeneration. Recent advances in molecular biology underscore the significance of nuclear factor E2-related factor 2 (Nrf2), often termed the "guardian of redox homeostasis," in the pathophysiology of AD. Nrf2 orchestrates cellular responses to oxidative stress and neuroinflammation - two interlinked pathological features of AD. In the brains of AD patients, Nrf2 activity is diminished, weakening the brain's ability to counteract oxidative damage. Additionally, the BTB and CNC homology 1 (Bach1) protein, a transcriptional repressor of Nrf2, has emerged as a potential therapeutic target. Here, we review the current landscape of clinical trials in AD and identify the limitations of the conventional approaches. We then explore the prospects of a novel approach that combines Nrf2 activation with Bach1 inhibition to achieve a multipronged defense against oxidative stress, neuroinflammation, and other molecular culprits driving AD. This innovative strategy holds promise for synergistically modulating multiple neuroprotective pathways to advance AD treatment.
Collapse
Affiliation(s)
- Priyanka Soni
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Sudarshana M Sharma
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Bobby Thomas
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, SC, USA; Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA; Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
6
|
Bovis K, Davies-Branch M, Day PJR. Dysregulated Neurotransmission and the Role of Viruses in Alzheimer's Disease. ACS Chem Neurosci 2025; 16:982-987. [PMID: 40045566 PMCID: PMC11926781 DOI: 10.1021/acschemneuro.4c00763] [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: 11/13/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The causes of neurodegeneration remain elusive. There is growing evidence linking viral infection to dysregulated neurotransmission as a causative factor in Alzheimer's disease. Studies suggest that viral infection may result in dysregulated glutamatergic and l-arginine/NO neurotransmission that can initiate neurodegeneration and neuroinflammation within AD. This involves viral infection (HIV-1/HSV-1) altering glutamate biosynthesis and receptor activation resulting in excessive influxes of glutamate and subsequent dysregulation of Ca2+ influx that all contribute to reduced dendrite growth and tau phosphorylation. For l-arginine/NO neurotransmission, the mechanism derives from the "protective" antiviral mechanisms of NO that correlate with pathologies such as β-amyloid peptide accumulation and functional degeneration of hippocampal neurons, respectively. More research is required to underpin the direct mechanisms that viruses might impact to induce specific pathologies.
Collapse
Affiliation(s)
- Katherine Bovis
- Division
of Evolution, Infection & Genomic Sciences, University of Manchester, Manchester M13 9PL, U.K.
| | - Martha Davies-Branch
- Division
of Evolution, Infection & Genomic Sciences, University of Manchester, Manchester M13 9PL, U.K.
| | - Philip J. R. Day
- Division
of Evolution, Infection & Genomic Sciences, University of Manchester, Manchester M13 9PL, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, Manchester M1 7DN, U.K.
| |
Collapse
|
7
|
Shabani Sadr NK, Bakhtiarzadeh F, Shahpasand K, Mirnajafi-Zadeh J, Behmanesh M. Improving effects of melatonin on memory and synaptic potentiation in a mouse model of Alzheimer's-like disease: the involvement of glutamate homeostasis and mGluRs receptors. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:7. [PMID: 40102986 PMCID: PMC11916854 DOI: 10.1186/s12993-025-00271-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by progressive cognitive decline and synaptic dysfunction, largely driven by amyloid plaques and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau. These pathological hallmarks disrupt glutamate signaling, which is essential for synaptic plasticity and memory consolidation. This study investigates the therapeutic potential of melatonin on memory and synaptic plasticity in an AD-like mouse model, with a focus on its regulatory effects on glutamate homeostasis and metabotropic glutamate receptors (mGluRs). METHODS The study began with an in-silico bioinformatics analysis of RNA-seq datasets from hippocampal tissues of AD patients to identify differentially expressed genes (DEGs) related to glutamate signaling and tau pathology. An AD-like model was induced via intra-hippocampal injection of cis-phospho tau in C57BL/6 mice. Memory function was assessed using behavioral tests. Synaptic plasticity was evaluated using in vitro field potential recording of hippocampal slices. Histological analyses included Nissl staining for neuronal density, Luxol Fast Blue for myelin integrity, and immunofluorescence for tau hyperphosphorylation. Molecular studies employed qPCR and Western blot to assess glutamate-related markers and tau phosphorylation. Melatonin (10 mg/kg) was administered intraperitoneally, starting either two weeks (early intervention) or four weeks (late intervention) post-induction. RESULTS Key molecular targets in glutamate signaling pathways were identified using bioinformatics. AD-like mice displayed memory deficits and synaptic dysfunction. Melatonin improved cognitive function, especially with early intervention, as confirmed by behavioral tests. Histological studies revealed reduced neuronal loss, improved myelin integrity, and decreased tau hyperphosphorylation. Molecular findings showed restored mGluR expression and reduced GSK3 activity. Early intervention yielded superior outcomes, with partial restoration of synaptic plasticity observed in LTP recordings. CONCLUSIONS These findings underscore the neuroprotective properties of melatonin, mediated by its ability to modulate glutamate signaling and mGluR activity, offering new insights into its potential as a therapeutic agent for AD. Additionally, the results suggest that earlier administration of melatonin may significantly enhance its efficacy, highlighting the importance of timely intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Narjes Khatoun Shabani Sadr
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran, P.O Box 14115-154
| | - Fatemeh Bakhtiarzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran, P.O. Box, 14115-331
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran, P.O. Box, 14115-331
- Institute for Brain and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran, P.O Box 14115-154.
- Institute for Brain and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
8
|
Gupta S, Gupta AK, Mehan S, Khan Z, Gupta GD, Narula AS. Disruptions in cellular communication: Molecular interplay between glutamate/NMDA signalling and MAPK pathways in neurological disorders. Neuroscience 2025; 569:331-353. [PMID: 39809360 DOI: 10.1016/j.neuroscience.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Neurological disorders significantly impact the central nervous system, contributing to a growing public health crisis globally. The spectrum of these disorders includes neurodevelopmental and neurodegenerative diseases. This manuscript reviews the crucial roles of cellular signalling pathways in the pathophysiology of these conditions, focusing primarily on glutaminase/glutamate/NMDA receptor signalling, alongside the mitogen-activated protein kinase (MAPK) pathways-ERK1/2, C-JNK, and P38 MAPK. Activation of these pathways is often correlated with neuronal excitotoxicity, apoptosis, and inflammation, leading to many other pathological conditions such as traumatic brain injury, stroke, and brain tumor. The interplay between glutamate overstimulation and MAPK signalling exacerbates neurodegenerative processes, underscoring the complexity of cellular communication in maintaining neuronal health. Dysfunctional signalling alters synaptic plasticity and neuronal survival, contributing to cognitive impairments in various neurological diseases. The manuscript emphasizes the potential of targeting these signalling pathways for therapeutic interventions, promoting neuroprotection and reducing neuroinflammation. Incorporating insights from precision medicine and innovative drug delivery systems could enhance treatment efficacy. Overall, understanding the intricate mechanisms of these pathways is essential for developing effective strategies to mitigate the impact of neurological disorders and improve patient outcomes. This review highlights the necessity for further exploration into these signalling cascades to facilitate advancements in therapeutic approaches, ensuring better prognoses for individuals affected by neurological conditions.
Collapse
Affiliation(s)
- Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Abhishek Kumar Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India. https://mehanneuroscience.org
| | - Zuber Khan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
9
|
Talebi M, Ayatollahi SA, As’Habi MA, Kobarfard F, Khoramjouy M, Boroujeni FN, Faizi M, Ghassempour A. Investigating the neuroprotective effects of Dracocephalum moldavica extract and its effect on metabolomic profile of rat model of sporadic Alzheimer's disease. Heliyon 2025; 11:e42412. [PMID: 39981356 PMCID: PMC11840490 DOI: 10.1016/j.heliyon.2025.e42412] [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: 08/10/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive condition marked by multiple underlying mechanisms. Therefore, the investigation of natural products that can target multiple pathways presents a potential gate for the understanding and management of AD. This study aimed to assess the neuroprotective effects of the hydroalcoholic extract of Dracocephalum moldavica (DM) on cognitive impairment, biomarker changes, and putative metabolic pathways in a rat model of AD induced by intracerebroventricular streptozotocin (ICV-STZ). The DM extract was standardized and quantified based on examining total phenolic, total flavonoid, rosmarinic acid, and quercetin contents using colorimetry and high-performance liquid chromatography (HPLC) methods. The antioxidant potential of the extract was evaluated by 2,2-Diphenyl-1-picrylhydrazyl and nitric oxide radical scavenging assays. Male Wistar rats were injected with STZ (3 mg/kg, single dose, bilateral ICV) to induce a sporadic AD (sAD) model. Following model induction, rats were orally administered with DM extract (100, 200, and 400 mg/kg/day) or donepezil (5 mg/kg/day) for 21 days. Cognitive function was assessed using the radial arm water maze behavioral test. The histopathological evaluations were conducted in the cortex and hippocampus regions. Matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS) was used to assess metabolite changes in various brain regions. DM extract significantly attenuated cognitive dysfunction induced by ICV-STZ according to behavioral and histopathological investigations. Thirty-two discriminating metabolites related to the amino acid metabolism; the glutamate/gamma-aminobutyric acid/glutamine cycle; nucleotide metabolism; lipid metabolism (glycerophospholipids, sphingomyelins, ceramides, phosphatidylserines, and prostaglandins), and glucose metabolic pathways were identified in the brains of rats with sAD simultaneously for the first time in this model. Polyphenols in DM extract may contribute to the regulation of these pathways. After treatment with DM extract, 10 metabolites from the 32 identified ones were altered in the brain tissue of a rat model of sAD, most commonly at doses of 200 and 400 mg/kg. In conclusion, this study demonstrates the neuroprotective potential of DM by upregulation/downregulation of various pathophysiological biomarkers such as adenine, glycerophosphoglycerol, inosine, prostaglandins, and sphingomyelin induced by ICV-STZ in sAD. These findings are consistent with cognitive behavioral results and histopathological outcomes.
Collapse
Affiliation(s)
- Marjan Talebi
- Student Research Committee, Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Abdulmajid Ayatollahi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Pharmacognosy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali As’Habi
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G.C., Evin, Tehran, Iran
| | - Farzad Kobarfard
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Khoramjouy
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ghassempour
- Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G.C., Evin, Tehran, Iran
| |
Collapse
|
10
|
Bay S, Rodina A, Haut F, Roychowdhury T, Argyrousi EK, Staniszewski A, Han K, Sharma S, Chakrabarty S, Digwal CS, Stanisavljevic A, Labuza A, Alldred MJ, Panchal P, SanthaSeela A, Tuffery L, Li Z, Hashmi A, Rosiek E, Chan E, Monetti M, Sasaguri H, Saido TC, Schneider JA, Bennett DA, Fraser PE, Erdjument-Bromage H, Neubert TA, Ginsberg SD, Arancio O, Chiosis G. Systems-Level Interactome Mapping Reveals Actionable Protein Network Dysregulation Across the Alzheimer's Disease Spectrum. RESEARCH SQUARE 2025:rs.3.rs-5930673. [PMID: 39989971 PMCID: PMC11844643 DOI: 10.21203/rs.3.rs-5930673/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) progresses as a continuum, from preclinical stages to late-stage cognitive decline, yet the molecular mechanisms driving this progression remain poorly understood. Here, we provide a systems-level map of protein-protein interaction (PPI) network dysfunction across the AD spectrum and uncover epichaperomes-stable scaffolding platforms formed by chaperones and co-factors-as central drivers of this process. Using over 100 human brain specimens, mouse models, and human neurons, we show that epichaperomes emerge early, even in preclinical AD, and progressively disrupt multiple PPI networks critical for synaptic function and neuroplasticity. Glutamatergic neurons, essential for learning and memory, exhibit heightened vulnerability, with their dysfunction driven by protein sequestration into epichaperome scaffolds, independent of changes in protein expression. Notably, pharmacological disruption of epichaperomes with PU-AD restores PPI network integrity and reverses synaptic and cognitive deficits, directly linking epichaperome-driven network dysfunction to AD pathology. These findings establish epichaperomes as key mediators of molecular collapse in AD and identify network-centric intervention strategies as a promising avenue for disease-modifying therapies.
Collapse
Affiliation(s)
- Sadik Bay
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Florence Haut
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elentina K Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
| | - Kyung Han
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Souparna Chakrabarty
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Amanda Labuza
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anand SanthaSeela
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Laura Tuffery
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zhuoning Li
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arsalan Hashmi
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Rosiek
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Chan
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mara Monetti
- Proteomics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, University of Toronto, Toronto, ON M5R 0A3, Canada
| | - Hediye Erdjument-Bromage
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A Neubert
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Stephen D Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Division of Solid Tumors, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
11
|
Mohammad ZB, Yudin SCY, Goldberg BJ, Serra KL, Klegeris A. Exploring neuroglial signaling: diversity of molecules implicated in microglia-to-astrocyte neuroimmune communication. Rev Neurosci 2025; 36:91-117. [PMID: 39240134 PMCID: PMC11717358 DOI: 10.1515/revneuro-2024-0081] [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: 06/10/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Effective communication between different cell types is essential for brain health, and dysregulation of this process leads to neuropathologies. Brain glial cells, including microglia and astrocytes, orchestrate immune defense and neuroimmune responses under pathological conditions during which interglial communication is indispensable. Our appreciation of the complexity of these processes is rapidly increasing due to recent advances in molecular biology techniques, which have identified numerous phenotypic states of both microglia and astrocytes. This review focuses on microglia-to-astrocyte communication facilitated by secreted neuroimmune modulators. The combinations of interleukin (IL)-1α, tumor necrosis factor (TNF), plus complement component C1q as well as IL-1β plus TNF are already well-established microglia-derived stimuli that induce reactive phenotypes in astrocytes. However, given the large number of inflammatory mediators secreted by microglia and the rapidly increasing number of distinct functional states recognized in astrocytes, it can be hypothesized that many more intercellular signaling molecules exist. This review identifies the following group of cytokines and gliotransmitters that, while not established as interglial mediators yet, are known to be released by microglia and elicit functional responses in astrocytes: IL-10, IL-12, IL-18, transforming growth factor (TGF)-β, interferon (IFN)-γ, C-C motif chemokine ligand (CCL)5, adenosine triphosphate (ATP), l-glutamate, and prostaglandin E2 (PGE2). The review of molecular mechanisms engaged by these mediators reveals complex, partially overlapping signaling pathways implicated in numerous neuropathologies. Additionally, lack of human-specific studies is identified as a significant knowledge gap. Further research on microglia-to-astrocyte communication is warranted, as it could discover novel interglial signaling-targeted therapies for diverse neurological disorders.
Collapse
Affiliation(s)
- Zainab B. Mohammad
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Samantha C. Y. Yudin
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Benjamin J. Goldberg
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Kursti L. Serra
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| | - Andis Klegeris
- Laboratory of Cellular and Molecular Pharmacology, Department of Biology, University of British Columbia Okanagan Campus, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
12
|
Rubino E, Italia M, Giorgio E, Boschi S, Dimartino P, Pippucci T, Roveta F, Cambria CM, Elia G, Marcinnò A, Gallone S, Rogaeva E, Antonucci F, Brusco A, Gardoni F, Rainero I. Exome sequencing reveals a rare damaging variant in GRIN2C in familial late-onset Alzheimer's disease. Alzheimers Res Ther 2025; 17:21. [PMID: 39810256 PMCID: PMC11730494 DOI: 10.1186/s13195-024-01661-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with both genetic and environmental factors contributing to its pathogenesis. While early-onset AD has well-established genetic determinants, the genetic basis for late-onset AD remains less clear. This study investigates a large Italian family with late-onset autosomal dominant AD, identifying a novel rare missense variant in GRIN2C gene associated with the disease, and evaluates the functional impact of this variant. METHODS Affected and unaffected members from a Northern Italian family were included. Genomic DNA from family members was extracted and initially screened for pathogenic mutations in APP, PSEN1, and PSEN2, and screened for 77 genes associated with neurodegenerative conditions using NeuroX array assay. Exome sequencing was performed on three affected individuals and two healthy relatives. Bioinformatics analyses were conducted. Functional analysis was performed using primary neuronal cultures, and the impact of the variant was assessed through immunocytochemistry and electrophysiology. RESULTS Pathogenic variants were not identified in APP, PSEN1, or PSEN2, nor in the 77 genes in NeuroX array assay. Exome Sequencing revealed the c.3215C > T p.(A1072V) variant in GRIN2C gene (NM 000835.6), encoding for the glutamate ionotropic receptor N-methyl-D-aspartate receptor (NMDA) type subunit 2C (GluN2C). This variant segregated in 6 available AD patients in the family and was absent in 9 healthy relatives. Primary rat hippocampal neurons overexpressing GluN2CA1072V showed an increase in NMDAR-induced currents, suggesting altered glutamatergic transmission. Surface expression assays demonstrated an elevated surface/total ratio of the mutant GluN2C, correlating with the increased NMDAR current. Additionally, immunocytochemistry revealed in neurons expressing the mutant variant a reduced colocalization between the GluN2C subunit and 14-3-3 proteins, which are known to facilitate membrane trafficking of NMDARs. DISCUSSION We identified a rare missense variant in GRIN2C associated with late-onset autosomal dominant Alzheimer's disease. These findings highlight the role of GluN2C-containing NMDARs in glutamatergic signaling and their potential contribution to AD pathogenesis.
Collapse
Affiliation(s)
- Elisa Rubino
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy.
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy.
| | - Maria Italia
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, 20133, Italy
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, Pavia, 27100, Italy
- Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, 27100, Italy
| | - Silvia Boschi
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Paola Dimartino
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, Pavia, 27100, Italy
| | - Tommaso Pippucci
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria, Via Albertoni 15, Bologna, 40138, Italy
| | - Fausto Roveta
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Clara Maria Cambria
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Via Festa del Perdono 7, Milan, 20122, Italy
| | - Gabriella Elia
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Andrea Marcinnò
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
| | - Salvatore Gallone
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, King's College Circle 1, Toronto, ON, M5S1A8, Canada
| | - Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Via Festa del Perdono 7, Milan, 20122, Italy
- Institute of Neuroscience, IN-CNR, Via Raoul Follereau 3, Vedano al Lambro, 20854, Italy
| | - Alfredo Brusco
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
- Medical Genetics Unit, Città della Salute e della Scienza di Torino, University Hospital, Via Santena 19, Turin, 10126, Italy
- Molecular Biotechnology Center Guido Tarone, University of Turin, Piazza Nizza 44B, Turin, 10126, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Balzaretti 9, Milan, 20133, Italy
| | - Innocenzo Rainero
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Via Cherasco 15, Turin, 10126, Italy
- Center for Alzheimer's Disease and Related Dementias, Department of Neuroscience and Mental Health, AOU Città della Salute e della Scienza di Torino, University Hospital, Via Cherasco 15, Turin, 10126, Italy
| |
Collapse
|
13
|
Puranik N, Song M. Glutamate: Molecular Mechanisms and Signaling Pathway in Alzheimer's Disease, a Potential Therapeutic Target. Molecules 2024; 29:5744. [PMID: 39683904 DOI: 10.3390/molecules29235744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Gamma-glutamate is an important excitatory neurotransmitter in the central nervous system (CNS), which plays an important role in transmitting synapses, plasticity, and other brain activities. Nevertheless, alterations in the glutamatergic signaling pathway are now accepted as a central element in Alzheimer's disease (AD) pathophysiology. One of the most prevalent types of dementia in older adults is AD, a progressive neurodegenerative illness brought on by a persistent decline in cognitive function. Since AD has been shown to be multifactorial, a variety of pharmaceutical targets may be used to treat the condition. N-methyl-D-aspartic acid receptor (NMDAR) antagonists and acetylcholinesterase inhibitors (AChEIs) are two drug classes that the Food and Drug Administration has authorized for the treatment of AD. The AChEIs approved to treat AD are galantamine, donepezil, and rivastigmine. However, memantine is the only non-competitive NMDAR antagonist that has been authorized for the treatment of AD. This review aims to outline the involvement of glutamate (GLU) at the molecular level and the signaling pathways that are associated with AD to demonstrate the drug target therapeutic potential of glutamate and its receptor. We will also consider the opinion of the leading authorities working in this area, the drawback of the existing therapeutic strategies, and the direction for the further investigation.
Collapse
Affiliation(s)
- Nidhi Puranik
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
14
|
Li Y, Yang X. A β-mediated synaptic glutamate dynamics and calcium dynamics in astrocytes associated with Alzheimer's disease. Cogn Neurodyn 2024; 18:3401-3426. [PMID: 39712135 PMCID: PMC11655814 DOI: 10.1007/s11571-024-10064-6] [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: 09/01/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 12/24/2024] Open
Abstract
The accumulation of amyloid β peptide A β is assumed to be one of the main causes of Alzheimer's disease AD . There is increasing evidence that astrocytes are the primary targets of Aβ. Aβ can cause abnormal synaptic glutamate, aberrant extrasynaptic glutamate, and astrocytic calcium dysregulation through astrocyte glutamate transporters facing the synaptic cleft (GLT-syn), astrocyte glutamate transporters facing the extrasynaptic space (GLT-ess), metabotropic glutamate receptors in astrocytes (mGluR), N-methyl-D-aspartate receptors in astrocytes (NMDAR), and glutamatergic gliotransmitter release (Glio-Rel). However, it is difficult to experimentally identify the extent to which each pathway affects synaptic glutamate, extrasynaptic glutamate, and astrocytic calcium signaling. Motivated by these findings, this work established a concise mathematical model of astrocyteCa 2 + dynamics, including the above Aβ-mediated glutamate-related multiple pathways. The model results presented the extent to which five mechanisms acted upon by Aβ affect synaptic glutamate, extrasynaptic glutamate, and astrocytic intracellularCa 2 + signals. We found that GLT-syn is the main pathway through which Aβ affects synaptic glutamate. GLT-ess and Glio-Rel are the main pathways through which A β affects extrasynaptic glutamate. GLT-syn, mGluR, and NMDAR are the main pathways through which Aβ affects astrocytic intracellularCa 2 + signals. Additionally, we discovered a strong, monotonically increasing relationship between the mean glutamate concentration and the meanCa 2 + oscillation amplitude (or frequency). Our results may have therapeutic implications for slowing cell death induced by the combination of glutamate imbalance andCa 2 + dysregulation in AD.
Collapse
Affiliation(s)
- YuPeng Li
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, 710119 People’s Republic of China
| | - XiaoLi Yang
- School of Mathematics and Statistics, Shaanxi Normal University, Xi’an, 710119 People’s Republic of China
| |
Collapse
|
15
|
Al-Mazidi S. Molecular physiology unlocks the mystery that relates cognitive impairment with the retina in schizophrenia and autism spectrum disorders: a perspective review. Front Psychiatry 2024; 15:1495017. [PMID: 39588547 PMCID: PMC11586360 DOI: 10.3389/fpsyt.2024.1495017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Schizophrenia and Autism spectrum disorders (SSD and ASD) are neurodevelopmental disorders involving cognitive impairment. Timely diagnosis is important for early intervention; currently, no tools are available to help with early diagnosis. Molecular biomarkers of cognitive impairment have been extensively studied, but clinical correlation is crucial in screening for cognitive impairment in SSD and ASD. There has been growing interest in examining the retina to scan for neurological disorders since the retina is the only part of the central nervous system that can be directly imaged non-invasively and in a timely manner. This review discusses biomarkers of cognitive impairment and their correlation to the retina in SSD and ASD. It also discusses the possible involvement of the retina and molecular biomarkers, specifically Disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and ciliary neurotrophic factor (CNTF) in the pathophysiology of SSD and ASD. A protocol for early diagnosing cognitive impairment and its severity in SSD and ASD is also suggested. This review also mentions insights into the potential use of molecular biomarkers of cognitive impairment to enhance cognitive performance in ASD and SSD and areas where more research is needed to solve the mystery of the relationship between the retina and cognitive impairment in neurodevelopmental psychiatric disorders.
Collapse
Affiliation(s)
- Sarah Al-Mazidi
- Department of Anatomy and Physiology, Imam Muhammad ibn Saud Islamic University, Riyadh, Saudi Arabia
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Gao Y, Zhang H, Hu Y. Baseline serum glutamate: Implications for diagnosis and prediction in mild cognitive impairment and Alzheimer's disease of the Alzheimer's Disease Neuroimaging Initiative. J Clin Neurosci 2024; 129:110828. [PMID: 39265358 DOI: 10.1016/j.jocn.2024.110828] [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: 05/11/2024] [Revised: 07/28/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
PURPOSE Numerous studies have highlighted a close link between metabolic imbalances and Alzheimer's Disease (AD). The advancement of metabolomics has recently enabled the exploration of characteristic metabolic changes associated with AD. Studies indicate that serum glutamate (Glu) levels may correlate with mild cognitive impairment (MCI) and AD. This study aims to further elucidate the characteristics of baseline serum Glu levels in MCI and AD. METHODS This study included 783 participants from the Alzheimer's Disease Neuroimaging Initiative-1 (ADNI-1) cohort, categorized into cognitively normal (CN, n = 224), stable MCI (sMCI, n = 181), progressive MCI (pMCI, n = 193), and AD (n = 185). The study aimed to analyze the diagnostic value of baseline serum Glu, to explore its predictive capability for the progression from CN to MCI or AD, and from MCI to AD, and to analyze the relationship between serum Glu and cerebrospinal fluid (CSF) biomarkers and cognitive functions in different diagnostic groups. RESULTS Compared to the CN and sMCI groups, the pMCI group showed significantly lower levels of serum Glu, and the AD group also had lower Glu levels compared to the sMCI group. However, serum Glu did not show significant diagnostic value for MCI and AD. Lower levels of serum Glu could predict the progression from MCI to AD. CONCLUSION Serum Glu levels can predict the progression from MCI to AD, suggesting that it could provide new insights into the pathophysiological mechanisms of AD. However, serum Glu may not be an ideal peripheral biomarker for AD.
Collapse
Affiliation(s)
- Ying Gao
- Department of General Medical Wards Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Engineering Research Center of Stem Cell Therapy. Chongqing 400014, China
| | - Hua Zhang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuming Hu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
17
|
Singh NAK, Prasad S. Ellagic Acid Reverses Alterations in the Expression of AMPA Receptor and Its Scaffolding Proteins in the Cerebral Cortex and Memory Decline in STZ-sporadic Alzheimer' s Disease Mouse Model. Psychopharmacology (Berl) 2024; 241:2117-2131. [PMID: 38842699 DOI: 10.1007/s00213-024-06622-9] [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: 12/19/2023] [Accepted: 05/18/2024] [Indexed: 06/07/2024]
Abstract
RATIONALE Alzheimer's disease (AD), an age-dependent devastating neuropsychiatric disorder, is a leading cause of learning, memory and intellectual disabilities. Current therapeutic approaches for the amelioration of the anomalies of AD are not effective. OBJECTIVE In the present study, the molecular mechanisms underlying sporadic AD (sAD), the memory related behavioral analysis and neuroprotective effects of Ellagic acid (EA) were investigated. METHOD sAD mouse model was developed by intracerebroventricular (ICV) injection of Streptozotocin (STZ). The efficacy of EA, a naturally occurring polyphenol, in amelioration of anomalies associated with sAD was assessed. EA was administered once daily for 28 days at a dose of 75 mg/kg body weight followed by neurobehavioral, biochemical, molecular and neuronal count analysis to delineate the mode of action of EA. RESULT The ICV injection of STZ in mice significantly increased the expression of AD biomarkers in addition to enhanced oxidative stress. A decline in the discrimination index in Novel Object Recognition Test was observed indicating the compromise of recognition memory in AD. Studies on the expression of genes involved in synaptic plasticity reveal the dysregulation of the α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) of the glutamate and its scaffolding proteins in the postsynaptic density and thereby synaptic plasticity in AD. ICV-STZ led to significant upregulation of apoptotic markers which led to decrease in neuronal density of the cerebral cortex. EA significantly reversed the above and improved anomalies of sAD. CONCLUSION EA was observed to profoundly modulate the genes involved in AD pathophysiology, restored antioxidant enzymes activity, reduced lipid peroxidation and neuronal loss in the sAD brain. Further, EA was observed to effectively modulate the genes involved in apoptosis and synaptic plasticity. Therefore, EA possesses promising anti-AD properties, which may improve AD-associated anomalies by modulating synaptic plasticity via AMPAR signaling.
Collapse
Affiliation(s)
- Nidhi Anand K Singh
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi, 211005, Uttar Pradesh, India
| | - S Prasad
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study in Zoology, Institute of Science, Banaras Hindu University, Varanasi, 211005, Uttar Pradesh, India.
| |
Collapse
|
18
|
Yu CC, Wang XF, Wang J, Li C, Xiao J, Wang XS, Han R, Wang SQ, Lin YF, Kong LH, Du YJ. Electroacupuncture Alleviates Memory Deficits in APP/PS1 Mice by Targeting Serotonergic Neurons in Dorsal Raphe Nucleus. Curr Med Sci 2024; 44:987-1000. [PMID: 38990450 DOI: 10.1007/s11596-024-2908-9] [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: 05/01/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE Alzheimer's disease (AD) has become a significant global concern, but effective drugs able to slow down AD progression is still lacked. Electroacupuncture (EA) has been demonstrated to ameliorate cognitive impairment in individuals with AD. However, the underlying mechanisms remains poorly understood. This study aimed at examining the neuroprotective properties of EA and its potential mechanism of action against AD. METHODS APP/PS1 transgenic mice were employed to evaluate the protective effects of EA on Shenshu (BL 23) and Baihui (GV 20). Chemogenetic manipulation was used to activate or inhibit serotonergic neurons within the dorsal raphe nucleus (DRN). Learning and memory abilities were assessed by the novel object recognition and Morris water maze tests. Golgi staining, western blot, and immunostaining were utilized to determine EA-induced neuroprotection. RESULTS EA at Shenshu (BL 23) and Baihui (GV 20) effectively ameliorated learning and memory impairments in APP/PS1 mice. EA attenuated dendritic spine loss, increased the expression levels of PSD95, synaptophysin, and brain-derived neurotrophic factor in hippocampus. Activation of serotonergic neurons within the DRN can ameliorate cognitive deficits in AD by activating glutamatergic neurons mediated by 5-HT1B. Chemogenetic inhibition of serotonergic neurons in the DRN reversed the effects of EA on synaptic plasticity and memory. CONCLUSION EA can alleviate cognitive dysfunction in APP/PS1 mice by activating serotonergic neurons in the DRN. Further study is necessary to better understand how the serotonergic neurons-related neural circuits involves in EA-induced memory improvement in AD.
Collapse
Affiliation(s)
- Chao-Chao Yu
- Department of Rehabilitation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Xiao-Fei Wang
- Department of Rehabilitation, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430074, China
| | - Jia Wang
- Department of Acupuncture and Moxibustion, Wuhan Hospital of Integrated Traditional Chinese and Western Medicine, Wuhan, 430030, China
| | - Chu Li
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Juan Xiao
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xue-Song Wang
- College of Acupuncture-Moxibustion and Tuina, Hebei University of Chinese Medicine, Shijiazhuang, 050299, China
| | - Rui Han
- Department of Child Rehabilitation Medicine, Qujing Hospital of Maternity and Childcare, Qujing, 655002, China
| | - Shu-Qin Wang
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuan-Fang Lin
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China
- The 4th Clinical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Li-Hong Kong
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yan-Jun Du
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
19
|
Sinha P, Turchyna Y, Mitchell SPC, Sadek M, Armagan G, Perrin F, Maesako M, Berezovska O. Glutamate Transporter 1 as a Novel Negative Regulator of Amyloid β. Cells 2024; 13:1600. [PMID: 39404364 PMCID: PMC11475981 DOI: 10.3390/cells13191600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Glutamate transporter-1 (GLT-1) dynamics are implicated in excitotoxicity and Alzheimer's disease (AD) progression. Early stages of AD are often marked by hyperactivity and increased epileptiform activity preceding cognitive decline. Previously, we identified a direct interaction between GLT-1 and Presenilin 1 (PS1) in the brain, highlighting GLT-1 as a promising target in AD research. This study reports the significance of this interaction and uncovers a novel role of GLT-1 in modulating amyloid-beta (Aβ) production. Overexpression of GLT-1 in cells reduces the levels of Aβ40 and Aβ42 by decreasing γ-secretase activity pertinent to APP processing and induces a more "open" PS1 conformation, resulting in decreased Aβ42/40 ratio. Inhibition of the GLT-1/PS1 interaction using cell-permeable peptides produced an opposing effect on Aβ, highlighting the pivotal role of this interaction in regulating Aβ levels. These findings emphasize the potential of targeting the GLT-1/PS1 interaction as a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Oksana Berezovska
- Alzheimer Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, 114, 16th Street, Charlestown, MA 02129, USA
| |
Collapse
|
20
|
Talukder MEK, Akhter S, Ahammad F, Aktar A, Islam MS, Laboni AA, Afroze M, Khan M, Uddin MJ, Rahman MM. Multi-modal neuroprotection of Argemone mexicana L. against Alzheimer's disease: In vitro and in silico study. Heliyon 2024; 10:e37178. [PMID: 39286063 PMCID: PMC11402773 DOI: 10.1016/j.heliyon.2024.e37178] [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: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
Argemone mexicana L. is a medicinal plant, but its impact on Alzheimer's disease (AD) is right now undetermined. We intended to investigate the in-vitro anti-AD potential of leaves and flowers of A. mexicana methanol, ethanol, and ethyl extracts and to identify multi-modal anti-AD phytochemicals by computational approaches. Molecular docking of 196 phytochemicals identified three hit phytochemicals (protoberberine, protopine, and codeine) with higher binding affinity and multi-targeting ability toward AChE, BChE, BACE-1, and GSK-3β. Further MM-GBSA assays confirmed the integrity of these phytochemicals as the hit phytochemicals. However, these phytochemicals demonstrated favorable pharmacokinetics (PK) and drugable properties having no toxicity. Molecular dynamics simulations confirmed the binding strength of the hit phytoconstituents in the active pockets of AChE, BChE, BACE-1, and GSK-3β with multi-targeting inhibitory activities. All the extracts exhibited dose-dependent antioxidant and anti-cholinesterase activities supporting the in silico results in the context of oxidative stress and cholinergic pathways. Our results offer scientific validation of the anti-AD properties of Argemone mexicana L. and identified protoberberine, protopine, and codeine that could be used for the development of multi-modal inhibitors of AChE, BChE, BACE-1, and GSK-3β to combat AD. Additional in vivo validation is recommended to ensure a thorough assessment in the present research.
Collapse
Affiliation(s)
- Md Enamul Kabir Talukder
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Shahina Akhter
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Foy's Lake, Chittagong, 4202, Bangladesh
| | - Foysal Ahammad
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Asmim Aktar
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Saidul Islam
- Korea Institute of Radiological & Medical Sciences, 75, Nowon-ro, Nowon-gu, Seoul, South Korea
| | - Aysha Akter Laboni
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mirola Afroze
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mala Khan
- Bangladesh Reference Institute for Chemical Measurements (BRICM), Bangladesh Council of Scientific and Industrial Research, Dr Qudrat-i-Khuda Road, Dhanmondi, Dhaka, 1205, Bangladesh
| | - Mohammad Jashim Uddin
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Md Mashiar Rahman
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| |
Collapse
|
21
|
Zhang H, Cao D, Xu T, Chen E, Li G, Chen Y, Payne P, Province M, Li F. mosGraphFlow: a novel integrative graph AI model mining disease targets from multi-omic data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606219. [PMID: 39282361 PMCID: PMC11398418 DOI: 10.1101/2024.08.01.606219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Multi-omic data can better characterize complex cellular signaling pathways from multiple views compared to individual omic data. However, integrative multi-omic data analysis to rank key disease biomarkers and infer core signaling pathways remains an open problem. In this study, our novel contributions are that we developed a novel graph AI model, mosGraphFlow, for analyzing multi-omic signaling graphs (mosGraphs), 2) analyzed multi-omic mosGraph datasets of AD, and 3) identified, visualized and evaluated a set of AD associated signaling biomarkers and network. The comparison results show that the proposed model not only achieves the best classification accuracy but also identifies important AD disease biomarkers and signaling interactions. Moreover, the signaling sources are highlighted at specific omic levels to facilitate the understanding of the pathogenesis of AD. The proposed model can also be applied and expanded for other studies using multi-omic data. Model code is accessible via GitHub: https://github.com/FuhaiLiAiLab/mosGraphFlow.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Dekang Cao
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Computer Science and Engineering, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Tim Xu
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Computer Science and Engineering, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Emily Chen
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- School of Arts and Sciences, University of Rochester, Rochester, NY, 14627, USA
| | - Guangfu Li
- Department of Surgery, School of Medicine, University of Connecticut, CT, 06032, USA
| | - Yixin Chen
- Department of Computer Science and Engineering, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Philip Payne
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
22
|
Wiesman AI, Gallego‐Rudolf J, Villeneuve S, Baillet S, Wilson TW. Neurochemical organization of cortical proteinopathy and neurophysiology along the Alzheimer's disease continuum. Alzheimers Dement 2024; 20:6316-6331. [PMID: 39001629 PMCID: PMC11497661 DOI: 10.1002/alz.14110] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Despite parallel research indicating amyloid-β accumulation, alterations in cortical neurophysiological signaling, and multi-system neurotransmitter disruptions in Alzheimer's disease (AD), the relationships between these phenomena remains unclear. METHODS Using magnetoencephalography, positron emission tomography, and an atlas of 19 neurotransmitters, we studied the alignment between neurophysiological alterations, amyloid-β deposition, and the neurochemical gradients of the cortex. RESULTS In patients with mild cognitive impairment and AD, changes in cortical rhythms were topographically aligned with cholinergic, serotonergic, and dopaminergic systems. These alignments correlated with the severity of clinical impairments. Additionally, cortical amyloid-β plaques were preferentially deposited along neurochemical boundaries, influencing how neurophysiological alterations align with muscarinic acetylcholine receptors. Most of the amyloid-β-neurochemical and alpha-band neuro-physio-chemical alignments replicated in an independent dataset of individuals with asymptomatic amyloid-β accumulation. DISCUSSION Our findings demonstrate that AD pathology aligns topographically with the cortical distribution of chemical neuromodulator systems and scales with clinical severity, with implications for potential pharmacotherapeutic pathways. HIGHLIGHTS Changes in cortical rhythms in Alzheimer's are organized along neurochemical boundaries. The strength of these alignments is related to clinical symptom severity. Deposition of amyloid-β (Aβ) is aligned with similar neurotransmitter systems. Aβ deposition mediates the alignment of beta rhythms with cholinergic systems. Most alignments replicate in participants with pre-clinical Alzheimer's pathology.
Collapse
Affiliation(s)
- Alex I. Wiesman
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Department of Biomedical Physiology & KinesiologySimon Fraser UniversityBurnabyBritish ColumbiaCanada
| | - Jonathan Gallego‐Rudolf
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | - Sylvain Baillet
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Tony W. Wilson
- Institute for Human NeuroscienceBoys Town National Research HospitalOmahaNebraskaUSA
- Department of Pharmacology & NeuroscienceCreighton UniversityOmahaNebraskaUSA
| | | |
Collapse
|
23
|
Tyagi S, Thakur AK. Effect of Capsaicin on 3-NP-Induced Neurotoxicity: A Pre-Clinical Study. Neurochem Res 2024; 49:2038-2059. [PMID: 38814358 DOI: 10.1007/s11064-024-04158-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
The study objectives are to investigate the ability of capsaicin to revert the toxic effects in glutamate and lipopolysaccharide (LPS)-induced neurotoxicity in Neuro2a (N2a) cells as well as thwarting cognitive impairments, mitochondrial deficits, and oxidative insults induced by 3-nitropropanoic acid (3-NP) in a rodent model of Huntington's disease. In-vitro study with N2a cells was performed through MTT and LDH assay and their biochemical examinations were also performed. 3-NP-administered mice (n = 6) were treated with capsaicin (5, 10, and 20 mg/kg) through the per-oral (p.o.) route for 7 consecutive days. Physiological and behavioral studies were performed in drug-treated mice. After behavioral studies, biochemical parameters were performed for cytokines levels, various oxidative stress parameters, and mitochondrial enzyme complex activities with mitochondrial permeability. N2a cells treated with capsaicin demonstrated neuroprotective effects and reduced neurotoxicity. Based on experimental observation, in an in-vitro study, the effective dose of CAP was 50 µM. Moreover, a 100 µM dose of capsaicin had toxic effects on neuronal cells (N2a cells). On the other hand, the effective dose of 3-NP was 20 mg/kg, (p.o.) in animals (in-vivo). All tested doses of capsaicin upturned the cognitive impairment and motor in-coordination effects induced by 3-NP. 3-NP-injected mice demonstrated substantially increased pro-inflammatory cytokine concentrations, defective mitochondrial complex activity, and augmented oxidative insult. However, capsaicin at different doses reduced oxidative damage and cytokines levels and improved mitochondrial complex activity along with mitochondrial permeability. Furthermore, capsaicin (10 and 20 mg/kg) improved the TNF-α concentration. These findings suggested because of the anti-inflammatory and antioxidant effect, capsaicin can be considered a novel treatment for the management of neurodegenerative disorders by reverting the antioxidant enzyme activity, pro-inflammatory cytokines concentration, and mitochondrial functions.
Collapse
Affiliation(s)
- Sakshi Tyagi
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110 017, India
| | - Ajit Kumar Thakur
- Neuropharmacology Research Laboratory, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110 017, India.
| |
Collapse
|
24
|
Gao Y, Xu SM, Cheng Y, Takenaka K, Lindner G, Janitz M. Investigation of the Circular Transcriptome in Alzheimer's Disease Brain. J Mol Neurosci 2024; 74:64. [PMID: 38981928 PMCID: PMC11233389 DOI: 10.1007/s12031-024-02236-0] [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: 03/15/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024]
Abstract
Circular RNAs (circRNAs) are a subclass of non-coding RNAs which have demonstrated potential as biomarkers for Alzheimer's disease (AD). In this study, we conducted a comprehensive exploration of the circRNA transcriptome within AD brain tissues. Specifically, we assessed circRNA expression patterns in the dorsolateral prefrontal cortex collected from nine AD-afflicted individuals and eight healthy controls. Utilising two circRNA detection tools, CIRI2 and CIRCexplorer2, we detected thousands of circRNAs and performed a differential expression analysis. CircRNAs which exhibited statistically significantly differential expression were identified as AD-specific differentially expressed circRNAs. Notably, our investigation revealed 120 circRNAs with significant upregulation and 1325 circRNAs displaying significant downregulation in AD brains when compared to healthy brain tissue. Additionally, we explored the expression profiles of the linear RNA counterparts corresponding to differentially expressed circRNAs in AD-afflicted brains and discovered that the linear RNA counterparts exhibited no significant changes in the levels of expression. We used CRAFT tool to predict that circUBE4B had potential to target miRNA named as hsa-miR-325-5p, ultimately regulated CD44 gene. This study provides a comprehensive overview of differentially expressed circRNAs in the context of AD brains, underscoring their potential as molecular biomarkers for AD. These findings significantly enhance our comprehension of AD's underlying pathophysiological mechanisms, offering promising avenues for future diagnostic and therapeutic developments.
Collapse
Affiliation(s)
- Yulan Gao
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Si-Mei Xu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Yuning Cheng
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Konii Takenaka
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Grace Lindner
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
25
|
Leong VS, Yu J, Castor K, Al-Ezzi A, Arakaki X, Fonteh AN. Associations of Plasma Glutamatergic Metabolites with Alpha Desynchronization during Cognitive Interference and Working Memory Tasks in Asymptomatic Alzheimer's Disease. Cells 2024; 13:970. [PMID: 38891102 PMCID: PMC11171970 DOI: 10.3390/cells13110970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Electroencephalogram (EEG) studies have suggested compensatory brain overactivation in cognitively healthy (CH) older adults with pathological beta-amyloid(Aβ42)/tau ratios during working memory and interference processing. However, the association between glutamatergic metabolites and brain activation proxied by EEG signals has not been thoroughly investigated. We aim to determine the involvement of these metabolites in EEG signaling. We focused on CH older adults classified under (1) normal CSF Aβ42/tau ratios (CH-NATs) and (2) pathological Aβ42/tau ratios (CH-PATs). We measured plasma glutamine, glutamate, pyroglutamate, and γ-aminobutyric acid concentrations using tandem mass spectrometry and conducted a correlational analysis with alpha frequency event-related desynchronization (ERD). Under the N-back working memory paradigm, CH-NATs presented negative correlations (r = ~-0.74--0.96, p = 0.0001-0.0414) between pyroglutamate and alpha ERD but positive correlations (r = ~0.82-0.95, p = 0.0003-0.0119) between glutamine and alpha ERD. Under Stroop interference testing, CH-NATs generated negative correlations between glutamine and left temporal alpha ERD (r = -0.96, p = 0.037 and r = -0.97, p = 0.027). Our study demonstrated that glutamine and pyroglutamate levels were associated with EEG activity only in CH-NATs. These results suggest cognitively healthy adults with amyloid/tau pathology experience subtle metabolic dysfunction that may influence EEG signaling during cognitive challenge. A longitudinal follow-up study with a larger sample size is needed to validate these pilot studies.
Collapse
Affiliation(s)
- Vincent Sonny Leong
- Cognition and Brain Integration Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA (X.A.)
| | - Jiaquan Yu
- Biomarker and Neuro-Disease Mechanism Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA
| | - Katherine Castor
- Biomarker and Neuro-Disease Mechanism Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA
| | - Abdulhakim Al-Ezzi
- Cognition and Brain Integration Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA (X.A.)
| | - Xianghong Arakaki
- Cognition and Brain Integration Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA (X.A.)
| | - Alfred Nji Fonteh
- Biomarker and Neuro-Disease Mechanism Laboratory, Neurosciences Department, Huntington Medical Research Institutes, Pasadena, CA 91105, USA
| |
Collapse
|
26
|
Dos Santos HM, Bertollo AG, Mingoti MED, Grolli RE, Kreuz KM, Ignácio ZM. Dementia and depression: Biological connections with amyloid β protein. Basic Clin Pharmacol Toxicol 2024; 134:563-573. [PMID: 38459754 DOI: 10.1111/bcpt.13996] [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: 10/15/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024]
Abstract
Dementia is an umbrella term for a broad group of age-associated neurodegenerative diseases. It is estimated that dementia affects 50 million people worldwide and that Alzheimer's disease (AD) is responsible for up to 75% of cases. Small extracellular senile plaques composed of filamentous aggregates of amyloid β (Aβ) protein tend to bind to neuronal receptors, affecting cholinergic, serotonergic, dopaminergic and noradrenergic neurotransmission, leading to neuroinflammation, among other pathophysiologic processes and subsequent neuronal death, followed by dementia. The amyloid cascade hypothesis points to a pathological process in the cleavage of the amyloid precursor protein (APP), resulting in pathological Aβ. There is a close relationship between the pathologies that lead to dementia and depression. It is estimated that depression is prevalent in up to 90% of individuals diagnosed with Parkinson's disease, with varying severity, and in 20 to 30% of cases of Alzheimer's disease. The hypothalamic pituitary adrenal (HPA) axis is the great intermediary between the pathophysiological mechanisms in neurodegenerative diseases and depression. This review discusses the role of Aβ protein in the pathophysiological mechanisms of dementia and depression, considering the HPA axis, neuroinflammation, oxidative stress, signalling pathways and neurotransmission.
Collapse
Affiliation(s)
- Helamã Moraes Dos Santos
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Amanda Gollo Bertollo
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Roberta Eduarda Grolli
- Laboratory for research into care, patient safety, and technological innovation in nursing and health, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Kelli Maria Kreuz
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Zuleide Maria Ignácio
- Laboratory of Physiology, Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| |
Collapse
|
27
|
Fakih N, Fakhoury M. Alzheimer Disease-Link With Major Depressive Disorder and Efficacy of Antidepressants in Modifying its Trajectory. J Psychiatr Pract 2024; 30:181-191. [PMID: 38819242 DOI: 10.1097/pra.0000000000000779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
Alzheimer disease (AD) is a devastating neurodegenerative disorder that affects millions of individuals worldwide, with no effective cure. The main symptoms include learning and memory loss, and the inability to carry out the simplest tasks, significantly affecting patients' quality of life. Over the past few years, tremendous progress has been made in research demonstrating a link between AD and major depressive disorder (MDD). Evidence suggests that MDD is commonly associated with AD and that it can serve as a precipitating factor for this disease. Antidepressants such as selective serotonin reuptake inhibitors, which are the first line of treatment for MDD, have shown great promise in the treatment of depression in AD, although their effectiveness remains controversial. The goal of this review is to summarize current knowledge regarding the association between AD, MDD, and antidepressant treatment. It first provides an overview of the interaction between AD and MDD at the level of genes, brain regions, neurotransmitter systems, and neuroinflammatory markers. The review then presents current evidence regarding the effectiveness of various antidepressants for AD-related pathophysiology and then finally discusses current limitations, challenges, and future directions.
Collapse
Affiliation(s)
- Nour Fakih
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | | |
Collapse
|
28
|
Wiesman AI, Gallego-Rudolf J, Villeneuve S, Baillet S, Wilson TW. Alignments between cortical neurochemical systems, proteinopathy and neurophysiological alterations along the Alzheimer's disease continuum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.13.24305551. [PMID: 38645027 PMCID: PMC11030470 DOI: 10.1101/2024.04.13.24305551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Two neuropathological hallmarks of Alzheimer's disease (AD) are the accumulation of amyloid-β (Aβ) proteins and alterations in cortical neurophysiological signaling. Despite parallel research indicating disruption of multiple neurotransmitter systems in AD, it has been unclear whether these two phenomena are related to the neurochemical organization of the cortex. We leveraged task-free magnetoencephalography and positron emission tomography, with a cortical atlas of 19 neurotransmitters to study the alignment and interactions between alterations of neurophysiological signaling, Aβ deposition, and the neurochemical gradients of the human cortex. In patients with amnestic mild cognitive impairment (N = 18) and probable AD (N = 20), we found that changes in rhythmic, but not arrhythmic, cortical neurophysiological signaling relative to healthy controls (N = 20) are topographically aligned with cholinergic, serotonergic, and dopaminergic neurochemical systems. These neuro-physio-chemical alignments are related to the severity of cognitive and behavioral impairments. We also found that cortical Aβ plaques are preferentially deposited along neurochemical boundaries, and mediate how beta-band rhythmic cortical activity maps align with muscarinic acetylcholine receptors. Finally, we show in an independent dataset that many of these alignments manifest in the asymptomatic stages of cortical Aβ accumulation (N = 33; N = 71 healthy controls), particularly the Aβ-neurochemical alignments (57.1%) and neuro-physio-chemical alignments in the alpha frequency band (62.5%). Overall, the present study demonstrates that the expression of pathology in pre-clinical and clinical AD aligns topographically with the cortical distribution of chemical neuromodulator systems, scaling with clinical severity and with implications for potential pharmacotherapeutic pathways.
Collapse
Affiliation(s)
- Alex I. Wiesman
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jonathan Gallego-Rudolf
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Sylvia Villeneuve
- Montreal Neurological Institute, McGill University, Montreal, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Sylvain Baillet
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Tony W. Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Omaha, NE, USA
- Department of Pharmacology & Neuroscience, Creighton University, Omaha, NE, USA
| | | |
Collapse
|
29
|
Eteleeb AM, Novotny BC, Tarraga CS, Sohn C, Dhungel E, Brase L, Nallapu A, Buss J, Farias F, Bergmann K, Bradley J, Norton J, Gentsch J, Wang F, Davis AA, Morris JC, Karch CM, Perrin RJ, Benitez BA, Harari O. Brain high-throughput multi-omics data reveal molecular heterogeneity in Alzheimer's disease. PLoS Biol 2024; 22:e3002607. [PMID: 38687811 PMCID: PMC11086901 DOI: 10.1371/journal.pbio.3002607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 05/10/2024] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Unbiased data-driven omic approaches are revealing the molecular heterogeneity of Alzheimer disease. Here, we used machine learning approaches to integrate high-throughput transcriptomic, proteomic, metabolomic, and lipidomic profiles with clinical and neuropathological data from multiple human AD cohorts. We discovered 4 unique multimodal molecular profiles, one of them showing signs of poor cognitive function, a faster pace of disease progression, shorter survival with the disease, severe neurodegeneration and astrogliosis, and reduced levels of metabolomic profiles. We found this molecular profile to be present in multiple affected cortical regions associated with higher Braak tau scores and significant dysregulation of synapse-related genes, endocytosis, phagosome, and mTOR signaling pathways altered in AD early and late stages. AD cross-omics data integration with transcriptomic data from an SNCA mouse model revealed an overlapping signature. Furthermore, we leveraged single-nuclei RNA-seq data to identify distinct cell-types that most likely mediate molecular profiles. Lastly, we identified that the multimodal clusters uncovered cerebrospinal fluid biomarkers poised to monitor AD progression and possibly cognition. Our cross-omics analyses provide novel critical molecular insights into AD.
Collapse
Affiliation(s)
- Abdallah M. Eteleeb
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
| | - Brenna C. Novotny
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Carolina Soriano Tarraga
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Christopher Sohn
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Eliza Dhungel
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Logan Brase
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Aasritha Nallapu
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Jared Buss
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
| | - Fabiana Farias
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Kristy Bergmann
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Joseph Bradley
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Joanne Norton
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Jen Gentsch
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Fengxian Wang
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
| | - Albert A. Davis
- Department of Neurology, Washington University, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, United States of America
| | - John C. Morris
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
- Department of Neurology, Washington University, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, United States of America
| | - Celeste M. Karch
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
- NeuroGenomics and Informatics Center, Washington University, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, United States of America
| | - Richard J. Perrin
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
- Department of Neurology, Washington University, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri, United States of America
| | - Bruno A. Benitez
- Department of Neurology and Neuroscience, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Oscar Harari
- Department of Psychiatry, Washington University, Saint Louis, St. Louis, Missouri, United States of America
- The Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University, St. Louis, Missouri, United States of America
| |
Collapse
|
30
|
Wrzesień A, Andrzejewski K, Jampolska M, Kaczyńska K. Respiratory Dysfunction in Alzheimer's Disease-Consequence or Underlying Cause? Applying Animal Models to the Study of Respiratory Malfunctions. Int J Mol Sci 2024; 25:2327. [PMID: 38397004 PMCID: PMC10888758 DOI: 10.3390/ijms25042327] [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: 01/17/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disease that is the most common cause of dementia among the elderly. In addition to dementia, which is the loss of cognitive function, including thinking, remembering, and reasoning, and behavioral abilities, AD patients also experience respiratory disturbances. The most common respiratory problems observed in AD patients are pneumonia, shortness of breath, respiratory muscle weakness, and obstructive sleep apnea (OSA). The latter is considered an outcome of Alzheimer's disease and is suggested to be a causative factor. While this narrative review addresses the bidirectional relationship between obstructive sleep apnea and Alzheimer's disease and reports on existing studies describing the most common respiratory disorders found in patients with Alzheimer's disease, its main purpose is to review all currently available studies using animal models of Alzheimer's disease to study respiratory impairments. These studies on animal models of AD are few in number but are crucial for establishing mechanisms, causation, implementing potential therapies for respiratory disorders, and ultimately applying these findings to clinical practice. This review summarizes what is already known in the context of research on respiratory disorders in animal models, while pointing out directions for future research.
Collapse
Affiliation(s)
| | | | | | - Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland; (A.W.); (K.A.); (M.J.)
| |
Collapse
|
31
|
Beckers CJ, Mrestani A, Komma F, Dannhäuser S. Versatile Endogenous Editing of GluRIIA in Drosophila melanogaster. Cells 2024; 13:323. [PMID: 38391936 PMCID: PMC10887371 DOI: 10.3390/cells13040323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Glutamate receptors at the postsynaptic side translate neurotransmitter release from presynapses into postsynaptic excitation. They play a role in many forms of synaptic plasticity, e.g., homeostatic scaling of the receptor field, activity-dependent synaptic plasticity and the induction of presynaptic homeostatic potentiation (PHP). The latter process has been extensively studied at Drosophila melanogaster neuromuscular junctions (NMJs). The genetic removal of the glutamate receptor subunit IIA (GluRIIA) leads to an induction of PHP at the synapse. So far, mostly imprecise knockouts of the GluRIIA gene have been utilized. Furthermore, mutated and tagged versions of GluRIIA have been examined in the past, but most of these constructs were not expressed under endogenous regulatory control or involved the mentioned imprecise GluRIIA knockouts. We performed CRISPR/Cas9-assisted gene editing at the endogenous locus of GluRIIA. This enabled the investigation of the endogenous expression pattern of GluRIIA using tagged constructs with an EGFP and an ALFA tag for super-resolution immunofluorescence imaging, including structured illumination microscopy (SIM) and direct stochastic optical reconstruction microscopy (dSTORM). All GluRIIA constructs exhibited full functionality and PHP could be induced by philanthotoxin at control levels. By applying hierarchical clustering algorithms to analyze the dSTORM data, we detected postsynaptic receptor cluster areas of ~0.15 µm2. Consequently, our constructs are suitable for ultrastructural analyses of GluRIIA.
Collapse
Affiliation(s)
- Constantin J. Beckers
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| | - Achmed Mrestani
- Department of Neurology, University of Leipzig Medical Center, D-04103 Leipzig, Germany;
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, D-04103 Leipzig, Germany
| | - Fabian Komma
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| | - Sven Dannhäuser
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, D-97070 Würzburg, Germany
| |
Collapse
|
32
|
Baek JH, Park H, Kang H, Kim R, Kang JS, Kim HJ. The Role of Glutamine Homeostasis in Emotional and Cognitive Functions. Int J Mol Sci 2024; 25:1302. [PMID: 38279303 PMCID: PMC10816396 DOI: 10.3390/ijms25021302] [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: 12/20/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Glutamine (Gln), a non-essential amino acid, is synthesized de novo by glutamine synthetase (GS) in various organs. In the brain, GS is exclusively expressed in astrocytes under normal physiological conditions, producing Gln that takes part in glutamatergic neurotransmission through the glutamate (Glu)-Gln cycle. Because the Glu-Gln cycle and glutamatergic neurotransmission play a pivotal role in normal brain activity, maintaining Gln homeostasis in the brain is crucial. Recent findings indicated that a neuronal Gln deficiency in the medial prefrontal cortex in rodents led to depressive behaviors and mild cognitive impairment along with lower glutamatergic neurotransmission. In addition, exogenous Gln supplementation has been tested for its ability to overcome neuronal Gln deficiency and reverse abnormal behaviors induced by chronic immobilization stress (CIS). Although evidence is accumulating as to how Gln supplementation contributes to normalizing glutamatergic neurotransmission and the Glu-Gln cycle, there are few reviews on this. In this review, we summarize recent evidence demonstrating that Gln supplementation ameliorates CIS-induced deleterious changes, including an imbalance of the Glu-Gln cycle, suggesting that Gln homeostasis is important for emotional and cognitive functions. This is the first review of detailed mechanistic studies on the effects of Gln supplementation on emotional and cognitive functions.
Collapse
Affiliation(s)
| | | | | | | | | | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences, College of Medicine, Institute of Medical Science, Tyrosine Peptide Multiuse Research Group, Anti-Aging Bio Cell Factory Regional Leading Research Center, Gyeongsang National University, 15 Jinju-daero 816 Beongil, Jinju 52727, Gyeongnam, Republic of Korea; (J.H.B.); (H.P.); (H.K.); (R.K.); (J.S.K.)
| |
Collapse
|
33
|
Milham LT, Morris GP, Konen LM, Rentsch P, Avgan N, Vissel B. Quantification of AMPA receptor subunits and RNA editing-related proteins in the J20 mouse model of Alzheimer's disease by capillary western blotting. Front Mol Neurosci 2024; 16:1338065. [PMID: 38299128 PMCID: PMC10828003 DOI: 10.3389/fnmol.2023.1338065] [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: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Accurate modelling of molecular changes in Alzheimer's disease (AD) dementia is crucial for understanding the mechanisms driving neuronal pathology and for developing treatments. Synaptic dysfunction has long been implicated as a mechanism underpinning memory dysfunction in AD and may result in part from changes in adenosine deaminase acting on RNA (ADAR) mediated RNA editing of the GluA2 subunit of AMPA receptors and changes in AMPA receptor function at the post synaptic cleft. However, few studies have investigated changes in proteins which influence RNA editing and notably, AD studies that focus on studying changes in protein expression, rather than changes in mRNA, often use traditional western blotting. Methods Here, we demonstrate the value of automated capillary western blotting to investigate the protein expression of AMPA receptor subunits (GluA1-4), the ADAR RNA editing proteins (ADAR1-3), and proteins known to regulate RNA editing (PIN1, WWP2, FXR1P, and CREB1), in the J20 AD mouse model. We describe extensive optimisation and validation of the automated capillary western blotting method, demonstrating the use of total protein to normalise protein load, in addition to characterising the optimal protein/antibody concentrations to ensure accurate protein quantification. Following this, we assessed changes in proteins of interest in the hippocampus of 44-week-old J20 AD mice. Results We observed an increase in the expression of ADAR1 p110 and GluA3 and a decrease in ADAR2 in the hippocampus of 44-week-old J20 mice. These changes signify a shift in the balance of proteins that play a critical role at the synapse. Regression analysis revealed unique J20-specific correlations between changes in AMPA receptor subunits, ADAR enzymes, and proteins that regulate ADAR stability in J20 mice, highlighting potential mechanisms mediating RNA-editing changes found in AD. Discussion Our findings in J20 mice generally reflect changes seen in the human AD brain. This study underlines the importance of novel techniques, like automated capillary western blotting, to assess protein expression in AD. It also provides further evidence to support the hypothesis that a dysregulation in RNA editing-related proteins may play a role in the initiation and/or progression of AD.
Collapse
Affiliation(s)
- Luke T. Milham
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Gary P. Morris
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Lyndsey M. Konen
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Peggy Rentsch
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Nesli Avgan
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
| | - Bryce Vissel
- Centre for Neuroscience and Regenerative Medicine, St Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital, Sydney, NSW, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
34
|
Singh S, Ahuja A, Pathak S. Potential Role of Oxidative Stress in the Pathophysiology of Neurodegenerative Disorders. Comb Chem High Throughput Screen 2024; 27:2043-2061. [PMID: 38243956 DOI: 10.2174/0113862073280680240101065732] [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: 09/21/2023] [Revised: 11/27/2023] [Accepted: 12/14/2023] [Indexed: 01/22/2024]
Abstract
Neurodegeneration causes premature death in the peripheral and central nervous system. Neurodegeneration leads to the accumulation of oxidative stress, inflammatory responses, and the generation of free radicals responsible for nervous disorders like amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, and Huntington's disorders. Therefore, focus must be diverted towards treating and managing these disorders, as it is very challenging. Furthermore, effective therapies are also lacking, so the growing interest of the global market must be inclined towards developing newer therapeutic approaches that can intercept the progression of neurodegeneration. Emerging evidences of research findings suggest that antioxidant therapy has significant potential in modulating disease phenotypes. This makes them promising candidates for further investigation. This review focuses on the role of oxidative stress and reactive oxygen species in the pathological mechanisms of various neurodegenerative diseases, amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, and Huntington's disorders and their neuroprotection. Additionally, it highlights the potential of antioxidant-based therapeutics in mitigating disease severity in humans and improving patient compliance. Ongoing extensive global research further sheds light on exploring new therapeutic targets for a deeper understanding of disease mechanisms in the field of medicine and biology targeting neurogenerative disorders.
Collapse
Affiliation(s)
- Sonia Singh
- Institute of Pharmaceutical Research, GLA University Mathura, U.P, 281406, India
| | - Ashima Ahuja
- Institute of Pharmaceutical Research, GLA University Mathura, U.P, 281406, India
| | - Shilpi Pathak
- Institute of Pharmaceutical Research, GLA University Mathura, U.P, 281406, India
| |
Collapse
|
35
|
Gautam D, Naik UP, Naik MU, Yadav SK, Chaurasia RN, Dash D. Glutamate Receptor Dysregulation and Platelet Glutamate Dynamics in Alzheimer's and Parkinson's Diseases: Insights into Current Medications. Biomolecules 2023; 13:1609. [PMID: 38002291 PMCID: PMC10669830 DOI: 10.3390/biom13111609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Two of the most prevalent neurodegenerative disorders (NDDs), Alzheimer's disease (AD) and Parkinson's disease (PD), present significant challenges to healthcare systems worldwide. While the etiologies of AD and PD differ, both diseases share commonalities in synaptic dysfunction, thereby focusing attention on the role of neurotransmitters. The possible functions that platelets may play in neurodegenerative illnesses including PD and AD are becoming more acknowledged. In AD, platelets have been investigated for their ability to generate amyloid-ß (Aß) peptides, contributing to the formation of neurotoxic plaques. Moreover, platelets are considered biomarkers for early AD diagnosis. In PD, platelets have been studied for their involvement in oxidative stress and mitochondrial dysfunction, which are key factors in the disease's pathogenesis. Emerging research shows that platelets, which release glutamate upon activation, also play a role in these disorders. Decreased glutamate uptake in platelets has been observed in Alzheimer's and Parkinson's patients, pointing to a systemic dysfunction in glutamate handling. This paper aims to elucidate the critical role that glutamate receptors play in the pathophysiology of both AD and PD. Utilizing data from clinical trials, animal models, and cellular studies, we reviewed how glutamate receptors dysfunction contributes to neurodegenerative (ND) processes such as excitotoxicity, synaptic loss, and cognitive impairment. The paper also reviews all current medications including glutamate receptor antagonists for AD and PD, highlighting their mode of action and limitations. A deeper understanding of glutamate receptor involvement including its systemic regulation by platelets could open new avenues for more effective treatments, potentially slowing disease progression and improving patient outcomes.
Collapse
Affiliation(s)
- Deepa Gautam
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Ulhas P. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Meghna U. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Santosh K. Yadav
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Rameshwar Nath Chaurasia
- The Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India;
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
36
|
Davis SE, Cirincione AB, Jimenez-Torres AC, Zhu J. The Impact of Neurotransmitters on the Neurobiology of Neurodegenerative Diseases. Int J Mol Sci 2023; 24:15340. [PMID: 37895020 PMCID: PMC10607327 DOI: 10.3390/ijms242015340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide. Neurodegenerative diseases result from progressive damage to nerve cells in the brain or peripheral nervous system connections that are essential for cognition, coordination, strength, sensation, and mobility. Dysfunction of these brain and nerve functions is associated with Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and motor neuron disease. In addition to these, 50% of people living with HIV develop a spectrum of cognitive, motor, and/or mood problems collectively referred to as HIV-Associated Neurocognitive Disorders (HAND) despite the widespread use of a combination of antiretroviral therapies. Neuroinflammation and neurotransmitter systems have a pathological correlation and play a critical role in developing neurodegenerative diseases. Each of these diseases has a unique pattern of dysregulation of the neurotransmitter system, which has been attributed to different forms of cell-specific neuronal loss. In this review, we will focus on a discussion of the regulation of dopaminergic and cholinergic systems, which are more commonly disturbed in neurodegenerative disorders. Additionally, we will provide evidence for the hypothesis that disturbances in neurotransmission contribute to the neuronal loss observed in neurodegenerative disorders. Further, we will highlight the critical role of dopamine as a mediator of neuronal injury and loss in the context of NeuroHIV. This review will highlight the need to further investigate neurotransmission systems for their role in the etiology of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA; (S.E.D.); (A.B.C.); (A.C.J.-T.)
| |
Collapse
|
37
|
Cervetto C, Amaroli A, Amato S, Gatta E, Diaspro A, Maura G, Signore A, Benedicenti S, Marcoli M. Photons Induce Vesicular Exocytotic Release of Glutamate in a Power-Dependent Way. Int J Mol Sci 2023; 24:10977. [PMID: 37446155 DOI: 10.3390/ijms241310977] [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: 04/30/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Increasing evidence indicates that photobiomodulation, based on tissue irradiation with photons in the red to near-infrared spectrum, may be an effective therapeutic approach to central nervous system disorders. Although nervous system functionality has been shown to be affected by photons in animal models, as well as in preliminary evidence in healthy subjects or in patients with neuropsychiatric disorders, the mechanisms involved in the photobiomodulation effects have not yet been clarified. We previously observed that photobiomodulation could stimulate glutamate release. Here, we investigate mechanisms potentially involved in the glutamate-releasing effect of photons from adult mouse cerebrocortical nerve terminals. We report evidence of photon ability to induce an exocytotic vesicular release of glutamate from the terminals of glutamatergic neurons in a power-dependent way. It can be hypothesized that photobiomodulation, depending on the potency, can release glutamate in a potentially neurotoxic or physiological range.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
| | - Andrea Amaroli
- Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy
| | - Sarah Amato
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Elena Gatta
- DIFILAB, Department of Physics, University of Genova, Via Dodecaneso 33, 16146 Genova, Italy
| | - Alberto Diaspro
- DIFILAB, Department of Physics, University of Genova, Via Dodecaneso 33, 16146 Genova, Italy
- Nanoscopy, Nanophysics, Istituto Italiano di Tecnologia-IIT, Via Morego 30, 16133 Genova, Italy
- Biophysics Institute, National Research Council-CNR, Via de Marini, 6, 16149 Genova, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Antonio Signore
- Therapeutic Dentistry Department, Institute of Dentistry, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, b. 2, 119992 Moskow, Russia
| | - Stefano Benedicenti
- Department of Surgical Sciences and Integrated Diagnostics, University of Genova, Viale Benedetto XV 6, 16132 Genova, Italy
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
- Interuniversity Center for the Promotion of the 3Rs Principles in Teaching and Research (Centro 3R), 56122 Pisa, Italy
- Center of Excellence for Biomedical Research, University of Genova, 16132 Genova, Italy
| |
Collapse
|
38
|
Reveglia P, Paolillo C, Angiolillo A, Ferretti G, Angelico R, Sirabella R, Corso G, Matrone C, Di Costanzo A. A Targeted Mass Spectrometry Approach to Identify Peripheral Changes in Metabolic Pathways of Patients with Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24119736. [PMID: 37298687 DOI: 10.3390/ijms24119736] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/20/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder, is the most common cause of dementia in the elderly population. Since its original description, there has been intense debate regarding the factors that trigger its pathology. It is becoming apparent that AD is more than a brain disease and harms the whole-body metabolism. We analyzed 630 polar and apolar metabolites in the blood of 20 patients with AD and 20 healthy individuals, to determine whether the composition of plasma metabolites could offer additional indicators to evaluate any alterations in the metabolic pathways related to the illness. Multivariate statistical analysis showed that there were at least 25 significantly dysregulated metabolites in patients with AD compared with the controls. Two membrane lipid components, glycerophospholipids and ceramide, were upregulated, whereas glutamic acid, other phospholipids, and sphingolipids were downregulated. The data were analyzed using metabolite set enrichment analysis and pathway analysis using the KEGG library. The results showed that at least five pathways involved in the metabolism of polar compounds were dysregulated in patients with AD. Conversely, the lipid pathways did not show significant alterations. These results support the possibility of using metabolome analysis to understand alterations in the metabolic pathways related to AD pathophysiology.
Collapse
Affiliation(s)
- Pierluigi Reveglia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Carmela Paolillo
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Antonella Angiolillo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy
| | - Gabriella Ferretti
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Ruggero Angelico
- Department of Agriculture, Environmental and Food Sciences, University of Molise, 86100 Campobasso, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Alfonso Di Costanzo
- Centre for Research and Training in Medicine of Aging, Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy
| |
Collapse
|
39
|
Saura CA, Deprada A, Capilla-López MD, Parra-Damas A. Revealing cell vulnerability in Alzheimer's disease by single-cell transcriptomics. Semin Cell Dev Biol 2023; 139:73-83. [PMID: 35623983 DOI: 10.1016/j.semcdb.2022.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that by affecting specific brain cell types and regions cause severe pathological and functional changes in memory neural circuits. A comprehensive knowledge of the pathogenic mechanisms underlying AD requires a deeper understanding of the cell-specific pathological responses through integrative molecular analyses. Recent application of high-throughput single-cell transcriptomics to postmortem tissue has proved powerful to unravel cell susceptibility and biological networks responding to amyloid and tau pathologies. Here, we review single-cell transcriptomic studies successfully applied to decipher cell-specific gene expression programs and pathways in the brain of AD patients. Transcriptional information reveals both specific and common gene signatures affecting the major cerebral cell types, including astrocytes, endothelial cells, microglia, neurons, and oligodendrocytes. Cell type-specific transcriptomes associated with AD pathology and clinical symptoms are related to common biological networks affecting, among others pathways, synaptic function, inflammation, proteostasis, cell death, oxidative stress, and myelination. The general picture that emerges from systems-level single-cell transcriptomics is a spatiotemporal pattern of cell diversity and biological pathways, and novel cell subpopulations affected in AD brain. We argue that broader implementation of cell transcriptomics in larger AD human cohorts using standardized protocols is fundamental for reliable assessment of temporal and regional cell-type gene profiling. The possibility of applying this methodology for personalized medicine in clinics is still challenging but opens new roads for future diagnosis and treatment in dementia.
Collapse
Affiliation(s)
- Carlos A Saura
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Angel Deprada
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Maria Dolores Capilla-López
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Arnaldo Parra-Damas
- Institut de Neurociències, Department de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona 08193, Spain; Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Spain
| |
Collapse
|
40
|
Ailioaie LM, Ailioaie C, Litscher G. Photobiomodulation in Alzheimer's Disease-A Complementary Method to State-of-the-Art Pharmaceutical Formulations and Nanomedicine? Pharmaceutics 2023; 15:916. [PMID: 36986776 PMCID: PMC10054386 DOI: 10.3390/pharmaceutics15030916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
Alzheimer's disease (AD), as a neurodegenerative disorder, usually develops slowly but gradually worsens. It accounts for approximately 70% of dementia cases worldwide, and is recognized by WHO as a public health priority. Being a multifactorial disease, the origins of AD are not satisfactorily understood. Despite huge medical expenditures and attempts to discover new pharmaceuticals or nanomedicines in recent years, there is no cure for AD and not many successful treatments are available. The current review supports introspection on the latest scientific results from the specialized literature regarding the molecular and cellular mechanisms of brain photobiomodulation, as a complementary method with implications in AD. State-of-the-art pharmaceutical formulations, development of new nanoscale materials, bionanoformulations in current applications and perspectives in AD are highlighted. Another goal of this review was to discover and to speed transition to completely new paradigms for the multi-target management of AD, to facilitate brain remodeling through new therapeutic models and high-tech medical applications with light or lasers in the integrative nanomedicine of the future. In conclusion, new insights from this interdisciplinary approach, including the latest results from photobiomodulation (PBM) applied in human clinical trials, combined with the latest nanoscale drug delivery systems to easily overcome protective brain barriers, could open new avenues to rejuvenate our central nervous system, the most fascinating and complex organ. Picosecond transcranial laser stimulation could be successfully used to cross the blood-brain barrier together with the latest nanotechnologies, nanomedicines and drug delivery systems in AD therapy. Original, smart and targeted multifunctional solutions and new nanodrugs may soon be developed to treat AD.
Collapse
Affiliation(s)
- Laura Marinela Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Constantin Ailioaie
- Department of Medical Physics, Alexandru Ioan Cuza University, 11 Carol I Boulevard, 700506 Iasi, Romania
| | - Gerhard Litscher
- President of ISLA (International Society for Medical Laser Applications), Research Unit of Biomedical Engineering in Anesthesia and Intensive Care Medicine, Research Unit for Complementary and Integrative Laser Medicine, Traditional Chinese Medicine (TCM) Research Center Graz, Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 39, 8036 Graz, Austria
| |
Collapse
|
41
|
Kodam P, Sai Swaroop R, Pradhan SS, Sivaramakrishnan V, Vadrevu R. Integrated multi-omics analysis of Alzheimer's disease shows molecular signatures associated with disease progression and potential therapeutic targets. Sci Rep 2023; 13:3695. [PMID: 36879094 PMCID: PMC9986671 DOI: 10.1038/s41598-023-30892-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the formation of amyloid plaques implicated in neuronal death. Genetics, age, and sex are the risk factors attributed to AD. Though omics studies have helped to identify pathways associated with AD, an integrated systems analysis with the available data could help to understand mechanisms, potential biomarkers, and therapeutic targets. Analysis of transcriptomic data sets from the GEO database, and proteomic and metabolomic data sets from literature was performed to identify deregulated pathways and commonality analysis identified overlapping pathways among the data sets. The deregulated pathways included those of neurotransmitter synapses, oxidative stress, inflammation, vitamins, complement, and coagulation pathways. Cell type analysis of GEO data sets showed microglia, endothelial, myeloid, and lymphoid cells are affected. Microglia are associated with inflammation and pruning of synapses with implications for memory and cognition. Analysis of the protein-cofactor network of B2, B6, and pantothenate shows metabolic pathways modulated by these vitamins which overlap with the deregulated pathways from the multi-omics analysis. Overall, the integrated analysis identified the molecular signature associated with AD. Treatment with anti-oxidants, B2, B6, and pantothenate in genetically susceptible individuals in the pre-symptomatic stage might help in better management of the disease.
Collapse
Affiliation(s)
- Pradeep Kodam
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India
| | - R Sai Swaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India.
| | - Ramakrishna Vadrevu
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
42
|
Ku T, Liu Y, Xie Y, Hu J, Hou Y, Tan X, Ning X, Li G, Sang N. Tebuconazole mediates cognitive impairment via the microbe-gut-brain axis (MGBA) in mice. ENVIRONMENT INTERNATIONAL 2023; 173:107821. [PMID: 36827814 DOI: 10.1016/j.envint.2023.107821] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/19/2023] [Accepted: 02/11/2023] [Indexed: 06/18/2023]
Abstract
Tebuconazole, one of the most widely used triazole fungicides, is reported to potentially pose a risk of inducing neurological disorders in human beings. Considering the increasing exposure, whether it could influence cognitive function remains to be elucidated. Herein, we used a mouse model to evaluate the potential cognitive risks and possible mechanisms from the continuous edible application of tebuconazole at low concentrations. Our study revealed that tebuconazole deteriorated spatial learning and memory and downregulated the expression of glutamate receptor subunits. Importantly, metagenomic analysis indicated that tebuconazole not only led to significant shifts in the composition and diversity of the gut microbiota but also changed intestinal homeostasis. Specifically, after exposure, tebuconazole circulated in the bloodstream and largely entered the gut-brain axis for disruption, including disturbing the Firmicutes/Bacteroidetes ratio, interrelated neurotransmitters and systemic immune factors. Moreover, pretreatment with probiotics improved immune factor expression and restored the deterioration of synaptic function and spatial learning and memory. The current study provides novel insights concerning perturbations of the gut microbiome and its functions as a potential new mechanism by which tebuconazole exposes cognitive function-related human health.
Collapse
Affiliation(s)
- Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Yutong Liu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Yuanyuan Xie
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Jindong Hu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Yanwen Hou
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xin Tan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Xia Ning
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
43
|
Chakraborty P, Dey A, Gopalakrishnan AV, Swati K, Ojha S, Prakash A, Kumar D, Ambasta RK, Jha NK, Jha SK, Dewanjee S. Glutamatergic neurotransmission: A potential pharmacotherapeutic target for the treatment of cognitive disorders. Ageing Res Rev 2023; 85:101838. [PMID: 36610558 DOI: 10.1016/j.arr.2022.101838] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023]
Abstract
In the mammalian brain, glutamate is regarded to be the primary excitatory neurotransmitter due to its widespread distribution and wide range of metabolic functions. Glutamate plays key roles in regulating neurogenesis, synaptogenesis, neurite outgrowth, and neuron survival in the brain. Ionotropic and metabotropic glutamate receptors, neurotransmitters, neurotensin, neurosteroids, and others co-ordinately formulate a complex glutamatergic network in the brain that maintains optimal excitatory neurotransmission. Cognitive activities are potentially synchronized by the glutamatergic activities in the brain via restoring synaptic plasticity. Dysfunctional glutamate receptors and other glutamatergic components are responsible for the aberrant glutamatergic activity in the brain that cause cognitive impairments, loss of synaptic plasticity, and neuronal damage. Thus, controlling the brain's glutamatergic transmission and modifying glutamate receptor function could be a potential therapeutic strategy for cognitive disorders. Certain drugs that regulate glutamate receptor activities have shown therapeutic promise in improving cognitive functions in preclinical and clinical studies. However, several issues regarding precise functional information of glutamatergic activity are yet to be comprehensively understood. The present article discusses the scope of developing glutamatergic systems as prospective pharmacotherapeutic targets to treat cognitive disorders. Special attention has been given to recent developments, challenges, and future prospects.
Collapse
Affiliation(s)
- Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Kumari Swati
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Anand Prakash
- Department of Biotechnology, School of Life Science, Mahatma Gandhi Central University, Motihari, Bihar, India
| | - Dhruv Kumar
- School of Health Sciences & Technology, UPES University, Dehradun, Uttarakhand 248007, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, UP, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India.
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India.
| |
Collapse
|
44
|
Vrabec R, Blunden G, Cahlíková L. Natural Alkaloids as Multi-Target Compounds towards Factors Implicated in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054399. [PMID: 36901826 PMCID: PMC10003045 DOI: 10.3390/ijms24054399] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly people; currently, there is no efficient treatment. Considering the increase in life expectancy worldwide AD rates are predicted to increase enormously, and thus the search for new AD drugs is urgently needed. A great amount of experimental and clinical evidence indicated that AD is a complex disorder characterized by widespread neurodegeneration of the CNS, with major involvement of the cholinergic system, causing progressive cognitive decline and dementia. The current treatment, based on the cholinergic hypothesis, is only symptomatic and mainly involves the restoration of acetylcholine (ACh) levels through the inhibition of acetylcholinesterase (AChE). Since the introduction of the Amaryllidaceae alkaloid galanthamine as an antidementia drug in 2001, alkaloids have been one of the most attractive groups for searching for new AD drugs. The present review aims to comprehensively summarize alkaloids of various origins as multi-target compounds for AD. From this point of view, the most promising compounds seem to be the β-carboline alkaloid harmine and several isoquinoline alkaloids since they can simultaneously inhibit several key enzymes of AD's pathophysiology. However, this topic remains open for further research on detailed mechanisms of action and the synthesis of potentially better semi-synthetic analogues.
Collapse
Affiliation(s)
- Rudolf Vrabec
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
| | - Gerald Blunden
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Lucie Cahlíková
- Secondary Metabolites of Plants as Potential Drugs Research Group, Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmacy, Charles University, Heyrovského 1203, 500 05 Hradec Králové, Czech Republic
- Correspondence:
| |
Collapse
|
45
|
Urdánoz-Casado A, Sánchez-Ruiz de Gordoa J, Robles M, Roldan M, Macías Conde M, Acha B, Blanco-Luquin I, Mendioroz M. circRNA from APP Gene Changes in Alzheimer's Disease Human Brain. Int J Mol Sci 2023; 24:ijms24054308. [PMID: 36901741 PMCID: PMC10002054 DOI: 10.3390/ijms24054308] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/09/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of age-related dementia. Amyloid precursor protein (APP) is the precursor of Aβ peptides, and its role in AD has been widely investigated. Recently, it has been reported that a circular RNA (circRNA) originated from APP gene can serve as a template for Aβ synthesis, postulating it as an alternative pathway for the Aβ biogenesis. Moreover, circRNAs play important roles in brain development and in neurological diseases. Therefore, our aim was to study the expression of a circAPP (hsa_circ_0007556) and its linear cognate in AD human entorhinal cortex, a brain region most vulnerable to AD pathology. First, we confirmed the presence of circAPP (hsa_circ_0007556) in human entorhinal cortex samples using RT-PCR and Sanger sequencing of PCR products. Next, a 0.49-fold decrease in circAPP (hsa_circ_0007556) levels was observed in entorhinal cortex of AD cases compared to controls (p-value < 0.05) by qPCR. In contrast, APP mRNA expression did not show changes in the entorhinal cortex between AD cases and controls (Fold-change = 1.06; p-value = 0.81). A negative correlation was found between Aβ deposits and circAPP (hsa_circ_0007556) and APP expression levels (Rho Spearman = -0.56, p-value < 0.001 and Rho Spearman = -0.44, p-values < 0.001, respectively). Finally, by using bioinformatics tools, 17 miRNAs were predicted to bind circAPP (hsa_circ_0007556), and the functional analysis predicted that they were involved in some pathways, such as the Wnt-signaling pathway (p = 3.32 × 10-6). Long-term potentiation (p = 2.86 × 10-5), among others, is known to be altered in AD. To sum up, we show that circAPP (hsa_circ_0007556) is deregulated in the entorhinal cortex of AD patients. These results add to the notion that circAPP (hsa_circ_0007556) could be playing a role in the pathogenesis of AD disease.
Collapse
Affiliation(s)
- Amaya Urdánoz-Casado
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
| | - Javier Sánchez-Ruiz de Gordoa
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
- Department of Neurology, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Maitane Robles
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
| | - Miren Roldan
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
| | - Mónica Macías Conde
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
- Department of Neurology, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
| | - Blanca Acha
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
| | - Idoia Blanco-Luquin
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
| | - Maite Mendioroz
- Neuroepigenetics Laboratory-Navarrabiomed, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Universidad Pública de Navarra (UPNA), Pamplona, 31008 Navarra, Spain
- Department of Neurology, Complejo Hospitalario de Navarra-IdiSNA (Navarra Institute for Health Research), Pamplona, 31008 Navarra, Spain
- Correspondence: ; Tel.: +34-848422677
| |
Collapse
|
46
|
Neurotransmitters in Prevention and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24043841. [PMID: 36835251 PMCID: PMC9966535 DOI: 10.3390/ijms24043841] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Alzheimer's disease (AD) is the most frequent cause of cognitive impairment in middle-aged and older populations. There is a lack of drugs that demonstrate significant efficacy in AD, so the study of the pathogenesis of AD is of great importance. More efficacious interventions are needed, as reflected by our population's fast aging. Synaptic plasticity is the capacity of neurons to adjust their connections, and it is strongly tied to learning and memory, cognitive function, and brain injury recovery. Changes in synaptic strength, such as long-term potentiation (LTP) or inhibition (LTD), are thought to represent the biological foundation of the early stages of learning and memory. The results of numerous studies confirm that neurotransmitters and their receptors play an important role in the regulation of synaptic plasticity. However, so far, there is no definite correlation between the function of neurotransmitters in aberrant neural oscillation and AD-related cognitive impairment. We summarized the AD process to understand the impact of neurotransmitters in the progression and pathogenesis of AD, including the current status of neurotransmitter target drugs, and the latest evidence of neurotransmitters' function and changes in the AD process.
Collapse
|
47
|
Brymer KJ, Hurley EP, Barron JC, Mukherjee B, Barnes JR, Nafar F, Parsons MP. Asymmetric dysregulation of glutamate dynamics across the synaptic cleft in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2023; 11:27. [PMID: 36788598 PMCID: PMC9926626 DOI: 10.1186/s40478-023-01524-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/28/2023] [Indexed: 02/16/2023] Open
Abstract
Most research on glutamate spillover focuses on the deleterious consequences of postsynaptic glutamate receptor overactivation. However, two decades ago, it was noted that the glial coverage of hippocampal synapses is asymmetric: astrocytic coverage of postsynaptic sites exceeds coverage of presynaptic sites by a factor of four. The fundamental relevance of this glial asymmetry remains poorly understood. Here, we used the glutamate biosensor iGluSnFR, and restricted its expression to either CA3 or CA1 neurons to visualize glutamate dynamics at pre- and postsynaptic microenvironments, respectively. We demonstrate that inhibition of the primarily astrocytic glutamate transporter-1 (GLT-1) slows glutamate clearance to a greater extent at presynaptic compared to postsynaptic membranes. GLT-1 expression was reduced early in a mouse model of AD, resulting in slower glutamate clearance rates at presynaptic but not postsynaptic membranes that opposed presynaptic short-term plasticity. Overall, our data demonstrate that the presynapse is particularly vulnerable to GLT-1 dysfunction and may have implications for presynaptic impairments in a variety of brain diseases.
Collapse
Affiliation(s)
- Kyle J. Brymer
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Emily P. Hurley
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Jessica C. Barron
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Bandhan Mukherjee
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Jocelyn R. Barnes
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Firoozeh Nafar
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| | - Matthew P. Parsons
- grid.25055.370000 0000 9130 6822Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, NL A1B 3V6 Canada
| |
Collapse
|
48
|
Wang F, Jia T, Wang Y, Hu H, Wang Y, Chang L, Shen X, Liu G. Polycyclic aromatic hydrocarbons exposure was associated with microRNA differential expression and neurotransmitter changes: a cross-sectional study in coal miners. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:14838-14848. [PMID: 36161575 DOI: 10.1007/s11356-022-23230-2] [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: 04/21/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Exposure to polycyclic aromatic hydrocarbons (PAHs) may cause neurobehavioral changes. This study aimed to explore the underlying mechanism of PAH neurotoxicity in coal miners. Urinary PAH metabolites, neurotransmitters, and oxidative stress biomarkers of 652 coal miners were examined. Subjects were divided into high and low-exposure groups based on the median of total urinary PAH metabolites. Differentially expressed miRNAs were screened from 5 samples in the low-exposure group (≤ 4.88 μmol/mol Cr) and 5 samples in the high-exposure group (> 4.88 μmol/mol Cr) using microarray technology, followed by bioinformatics analysis of the potential molecular functions of miRNA target genes. Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) was used to validate differentially expressed miRNAs. Restricted cubic splines (RCS) were applied to assess the possible dose-response relationships. Compared to the low PAH exposure group, the high-exposure group had higher levels of 5-hydroxytryptamine (5-HT), epinephrine (E), and acetylcholine (ACh), and lower levels of acetylcholinesterase (AChE). 1-OHP had a dose-response relationship with malondialdehyde (MDA), dopamine (DA), 5-HT, and AChE (P for overall associations < 0.05). There were 19 differentially expressed microRNAs in microarray analysis, significantly enriched in the cell membrane, molecular binding to regulate transcription, and several signaling pathways such as PI3K-Akt. And in the validation stage, miR-885-5p, miR-20a-5p, and let-7i-3p showed differences in the low and high-exposure groups (P < 0.05). Changes in neurotransmitters and microRNA expression levels among the coal miners were associated with PAH exposure. Their biological functions are mainly related to the transcriptional regulation of nervous system diseases or signaling pathways of disorders. These findings provide new insights for future research of PAH neurotoxicity.
Collapse
Affiliation(s)
- Fang Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China.
| | - Teng Jia
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China
| | - Yu Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China
| | - Haiyuan Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China
| | - Yuying Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China
| | - Li Chang
- Department of Epidemiology and Health Statistics, School of Public Health, Shanxi Medical University, No. 56, Xinjian South Road, Yingze District, Taiyuan, Shanxi, China
| | - Xiaojun Shen
- Xishan Coal and Electricity (Group) Co., Ltd. Occupational Disease Prevention and Control Center, Taiyuan, China
| | - Gaisheng Liu
- Xishan Coal and Electricity (Group) Co., Ltd. Occupational Disease Prevention and Control Center, Taiyuan, China
| |
Collapse
|
49
|
Cognitive Healthy Aging in Mice: Boosting Memory by an Ergothioneine-Rich Hericium erinaceus Primordium Extract. BIOLOGY 2023; 12:biology12020196. [PMID: 36829475 PMCID: PMC9953177 DOI: 10.3390/biology12020196] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023]
Abstract
Brain aging is a crucial risk factor for several neurodegenerative disorders and dementia. The most affected cognitive function is memory, worsening early during aging. Inflammation and oxidative stress are known to have a role in pathogenesis of cognitive impairments, and a link exists between aging/frailty and immunosenescence/inflammaging. Based on anti-aging properties, medicinal mushrooms represent a source to develop medicines and functional foods. In particular, Hericium erinaceus (He) displays several actions ranging from boosting the immune system to fighting senescence, due to its active ingredients/metabolites. Among these, Ergothioneine (ERGO) is known as the longevity vitamin. Currently, we demonstrated the efficacy of an ERGO-rich He primordium extract (He2) in preventing cognitive decline in a murine model of aging. We focused on recognition memory deterioration during aging, monitored through spontaneous behavioral tests assessing both memory components and frailty index. A parallel significant decrease in key markers of inflammation and oxidative stress, i.e., IL6, TGFβ1, GFAP, Nrf2, SOD1, COX2, NOS2, was revealed in the hippocampus by immunohistochemistry, accompanied by an enhancement of NMDAR1and mGluR2, crucially involved in glutamatergic neurotransmission. In summary, we disclosed a selective, preventive and neuroprotective effect of He2 on aged hippocampus, both on recognition memory as well on inflammation/oxidative stress/glutamate receptors expression.
Collapse
|
50
|
Liu Z, Zhang H, Liu S, Hou Y, Chi G. The Dual Role of Astrocyte-Derived Exosomes and Their Contents in the Process of Alzheimer's Disease. J Alzheimers Dis 2023; 91:33-42. [PMID: 36373321 DOI: 10.3233/jad-220698] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Millions of patients worldwide are affected by Alzheimer's disease (AD), and the number of patients with AD is increasing. However, current treatment can only improve symptoms but cannot cure the disease. Astrocytes, glial cells in the central nervous system, play important roles in support, nutrition, protection, and information transmission in the nervous system. Pathological changes in astrocytes are closely associated with the development and progression of AD. As carriers for material and information exchange between astrocytes and other neural cells, astrocyte-derived exosomes (ADEs) have been widely studied in recent years, and ADE secretion has been shown to be increased in patients with AD and animal models of AD. ADEs contain a variety of substances, including nucleic acids, proteins, and lipids. The contents of ADEs can effectively control oxidative stress and detoxification during the early development of AD, thereby playing positive and negative roles in the occurrence and development of AD. In this review, we elaborate on the functions of ADEs and their components in AD and discuss their applications in AD research and clinical practice.
Collapse
Affiliation(s)
- Ziyu Liu
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Haotian Zhang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Shiji Liu
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Yi Hou
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|