1
|
Carter SKR, Tipton RK, Forman CM, Mosher AP, Ostrowski D, Ostrowski TD. Glutamate stress in the caudal nucleus tractus solitarii (nTS): Impact on respiratory function and synaptic signaling in an Alzheimer's disease model. Exp Neurol 2025; 387:115190. [PMID: 39986554 PMCID: PMC11921524 DOI: 10.1016/j.expneurol.2025.115190] [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: 12/13/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Respiratory dysfunction is a prevalent comorbidity in Alzheimer's disease (AD), yet its underlying mechanisms are poorly understood. Using the Streptozotocin (STZ) -induced rat model of AD, which replicates respiratory dysfunction and brain pathologies observed in human AD, we analyzed how these impairments relate to central neurological integration within the peripheral chemoreflex. Our focus was on glutamatergic signaling at the synapse between peripheral chemoafferents and second-order neurons in the nucleus tractus solitarii (nTS), a critical brainstem center for respiratory control. Activating the peripheral chemoreflex with potassium cyanide (KCN) increased respiratory frequency. Response magnitudes to repeated KCN injections typically decreased over time, which was exacerbated in the STZ-AD group. Similarly, repeated glutamate nanoinjections into the caudal/commissural nTS caused a pronounced reduction of respiratory frequency responses in STZ-AD. Electrophysiological analysis of nTS neurons within the peripheral chemoreflex revealed increased network activity, enhanced excitatory postsynaptic currents evoked by solitary tract stimulation (TS-EPSCs), and elevated asynchronous glutamate release following high-frequency stimulation (aEPSCs). These data were consistent with molecular evidence for astrogliosis (elevated GFAP expression), reduced astrocytic glutamate uptake (decreased EAAT2 expression), and presynaptic calcium dysregulation (increased TRPV1 expression). Additionally, presynaptic metabotropic glutamate receptors (mGluR3) were downregulated, while postsynaptic ionotropic receptor expression remained unchanged. With high-frequency solitary tract stimulation, STZ-AD rats had greater frequency-dependent TS-EPSC depression than controls, mirroring the diminished respiratory responses to KCN and glutamate nanoinjections. Our findings connect neurophysiological and molecular changes at the first nTS synapse of the peripheral chemoreflex with impaired respiratory responses to hypoxia in the STZ-AD model.
Collapse
Affiliation(s)
- Sarah K R Carter
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Robert K Tipton
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Cassadi M Forman
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Adison P Mosher
- Department of Biology, Truman State University, Kirksville, MO, USA
| | - Daniela Ostrowski
- Department of Pharmacology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Tim D Ostrowski
- Department of Physiology, Kirksville College of Osteopathic Medicine, A.T. Still University of Health Sciences, Kirksville, MO, USA.
| |
Collapse
|
2
|
Tzekaki E, Bekiari C, Pantazaki A, Tsantarliotou M, Tsolaki M, Lavrentiadou SN. A new protocol for the development of organoids based on molecular mechanisms in the developing newborn rat brain: Prospective applications in the study of Alzheimer's disease. J Neurosci Methods 2025; 417:110404. [PMID: 39978482 DOI: 10.1016/j.jneumeth.2025.110404] [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: 11/16/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Brain organoids have emerged as powerful models for studying brain development and neurological disorders COMPARISON WITH EXISTING METHODS: Current models rely on stem cell isolation and differentiation using different growth factors. Thus, their composition varies according to the protocol followed. NEW METHOD We developed a simple protocol to generate organoids from newborn rat whole brain. It is a one-step procedure that yields organoids of consistent composition. The whole brains from 3-day old pups were digested enzymatically. All isolated cells were seeded in culture plates using a basement membrane extract (BME) matrix as a scaffold and cultured in the presence of the appropriate medium. RESULTS Hematoxylin-eosin staining of 28-day-old cultured domes revealed their structural integrity, while immunohistochemistry confirmed the presence of neurons, astrocytes, microglia, and progenitor stem cells in the structures. To assess whether these organoids can serve as a model to study brain physiopathology, and in particular neurodegenerative diseases such as Alzheimer's disease (AD), we determined how these organoids respond upon their exposure to lipopolysaccharides (LPS), a potent neuroinflammatory factor. LPS-induced amyloid precursor protein (APP), tau protein and glial fibrillary acidic protein (GFAP) expression. Moreover, the intracellular levels of IL-1β and the extracellular levels of amyloid-beta (Aβ) were also elevated. CONCLUSIONS Therefore, this simple protocol results in the generation of functional brain organoids with a consistent structure, that requires no use of varying factors that may affect the structure and function of the produced organoids, thus providing a valuable tool for the study of the physiopathology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Eleni Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Chryssa Bekiari
- Laboratory of Anatomy and Histology School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Anastasia Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Maria Tsantarliotou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Magda Tsolaki
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Sophia N Lavrentiadou
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece; Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
3
|
Ahmad S, Ahmad L, Adil M, Sharma R, Khan S, Hasan N, Aqil M. Emerging nano-derived therapy for the treatment of dementia: a comprehensive review. Drug Deliv Transl Res 2025:10.1007/s13346-025-01863-3. [PMID: 40268841 DOI: 10.1007/s13346-025-01863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Dementia includes a variety of neurodegenerative diseases that affect and target the brain's fundamental cognitive functions. It is undoubtedly one of the diseases that affects people globally. The ameliorating the disease is still not known; the symptoms, however, can be prevented to an extent. Dementia encompasses Alzheimer's disease, Parkinson's disease, Huntington's disease, Lewy body dementia, mixed dementia, and various other diseases. The aggregation of β-amyloid protein plaques and the formation of neurofibrillary tangles have been concluded as the foremost cause for the onset of the disease. As the cases climb, new neuroprotective methods are being developed in the form of new drug delivery systems that provide targeted delivery. Herbal drugs like Ashwagandha, Brahmi, and Cannabis have shown satisfactory results by not only treating the symptoms but have also been shown to reduce and ameliorate the formation of amyloid plaque formation. This article explores the intricate possibilities of drug delivery and the absolute use of herbal drugs to target neurodegenerative diseases. The various possibilities of nanotechnology currently available with new emerging techniques are also discussed.
Collapse
Affiliation(s)
- Shadaan Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Lubna Ahmad
- Department of Conservative Dentistry and Endodontics, Manav Rachna Dental College, Manav Rachna International Institute of Research and Studies, Faridabad, India
| | - Mohammad Adil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ritu Sharma
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saara Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
4
|
Sharma V, Verma R, Singh TG. Targeting hypoxia-related pathobiology in Alzheimer's disease: strategies for prevention and treatment. Mol Biol Rep 2025; 52:416. [PMID: 40266407 DOI: 10.1007/s11033-025-10520-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Alzheimer's Disease (AD) is a neurodegenerative condition characterised by cognitive decline and memory impairment. Recent research highlights the important role of hypoxia, a state of insufficient oxygen availability, in exacerbating AD pathogenesis. MATERIALS AND METHODS Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the role of hypoxia mediated pathobiology in AD. Only peerreviewed articles published in reputable journals in English language were included. Conversely, non-peer-reviewed articles, conference abstracts, and editorials were excluded, along with studies lacking experimental or clinical relevance or those unavailable in full text. CONCLUSION Hypoxia exacerbates core pathological features such as oxidative stress, neuroinflammation, mitochondrial dysfunction, amyloid-beta (Aβ) dysregulation, and hyperphosphorylation of tau protein. These interlinked mechanisms establish a self-perpetuating cycle of neuronal damage, accelerating disease progression. Addressing hypoxia as a modifiable risk factor offers potential for both prevention and treatment of AD. Exploring hypoxia and the HIF signalling pathway may help counteract the neuropathological and symptomatic effects of neurodegeneration.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Reet Verma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
5
|
Bhuiyan P, Zhang W, Liang G, Jiang B, Vera R, Chae R, Kim K, Louis LS, Wang Y, Liu J, Chuang DM, Wei H. Intranasal Delivery of Lithium Salt Suppresses Inflammatory Pyroptosis in the Brain and Ameliorates Memory Loss and Depression-like Behavior in 5XFAD Mice. J Neuroimmune Pharmacol 2025; 20:26. [PMID: 40095208 PMCID: PMC11914297 DOI: 10.1007/s11481-025-10185-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a devastating neurodegenerative disease (AD) and has no treatment that can cure or halt the disease progression. This study explored the therapeutic potential of lithium salt dissolved in Ryanodex formulation vehicle (RFV) and delivered to the brain by intranasal application. We first compared lithium concentrations in the brain and blood of wild-type mice following intranasal or oral administration of lithium chloride (LiCl) dissolved in either RFV or water. The beneficial and side effects of intranasal versus oral LiCl in RFV in these mice were assessed and potential mechanisms underlying the efficacy of anti-inflammation and anti-pyroptosis in the brains were also investigated in both wild-type and 5XFAD Alzheimer's Disease (AD) mice brains. METHODS For the study of brain versus blood lithium concentrations, wild-type (WT) B6SJLF1/J mice at 2 months of age were treated with intranasal or oral LiCl (3 mmol/kg) dissolved in RFV or in water. Brain and blood lithium concentrations were measured at various times after drugs administration. Brain/blood lithium concentration ratios were then determined. For studying therapeutic efficacy versus side effects and their underlying mechanisms, 5XFAD and WT B6SJLF1/J mice were treated with intranasal LiCl (3 mmol/kg) daily, Monday to Friday each week, in RFV beginning at 2 or 9 months of age with a 12-week treatment duration. Animal behaviors were assessed for depression (tail suspension), cognition (fear conditioning and Y maze), olfaction (buried food test), and motor functions (rotarod) at the age of 5 and 12 months. Blood and brain tissue were harvested from these mice at 13 months. Blood biomarkers for the functions of thyroid (thyroid stimulating hormone, TSH) and kidney (creatinine) were measured using ELISA. Changes in protein expression levels of the endoplasmic reticulum Ca2+ release channels type 1 InsP3 receptors (InsP3R-1), malondialdehyde (MDA)-modified proteins and 4-hydroxy-2-nonenal (4-HNE), pyroptosis regulatory proteins (NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, N-terminal of Gasdermin D (GSDMD)), cytotoxic (IL-1β, IL-18, IL-6, TNF-α) and cytoprotective (IL-10) cytokines and synapse proteins (PSD-95, synapsin-1) were determined using immunoblotting. Mouse body weights were monitored regularly. RESULTS Compared to oral LiCl in RFV nanoparticles, intranasal treatment of WT mice with LiCl in RFV markedly decreased blood concentrations at the time range of 30-120 min. The ratio of brain/blood lithium concentration after intranasal lithium chloride in RFV significantly increased, in comparison to those after oral administration lithium chloride in RFV or intranasal administration of lithium chloride in water. Intranasal lithium chloride in RFV inhibited both memory loss and depressive behavior in adult and aged 5XFAD mice. Additionally intranasal treatment of aged 5XFAD mice with LiCl in RFV effectively suppressed the increases in InsP3R-1, intracellular oxidative stress markers (4-HNE-bound and MDA-modified proteins), pyroptosis activation proteins (NLRP3, cleaved caspase-1, N-terminal GSDMD) and cytotoxic cytokines (IL-1β, IL-6, TNF-α), but reversed the down-regulation of cytoprotective cytokine IL-10. Intranasal LiCl in RFV also alleviated the loss of the postsynaptic synapse proteins PSD-95, but not synapsin-1, in aged 5XFAD mice. Blood level of the kidney function marker creatinine was significantly increased in 5XFAD than in WT mice in an age-dependent manner and this elevation was abolished by intranasal delivery of LiCl in RFV. Intranasal LiCl in RFV for 12 weeks in both WT or 5XFAD mice did not affect blood biomarkers for thyroid function, nor did it affect smell or muscle function or body weight. CONCLUSION Intranasal administration of LiCl in RFV significantly decreased lithium blood concentrations and increased brain/blood lithium concentration ratio, in comparison to its oral administration. Intranasal administration of LiCl in RFV robustly protected against both memory loss and depressive-like behavior, while had no side effects concerning thyroid and kidney toxicity in 5XFAD mice. These lithium-induced beneficial effects were strongly associated with lithium's suppression of InsP3R-1 Ca2+ channel receptor increase, pathological neuroinflammation and activation of the pyroptosis pathway, as well as the loss of the synaptic protein PSD-95. Intranasal delivery of lithium salt in RFV could become an effective and potent inhibitor of pathological inflammation/pyroptosis in the CNS and serve as a new treatment for both AD-associated dementia and depression with minimal unwanted side effects including peripheral organ toxicity.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Wenjia Zhang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Bailin Jiang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| | - Robert Vera
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Rebecca Chae
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Kyulee Kim
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Lauren St Louis
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
| | - Ying Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 26600, People's Republic of China
| | - De-Maw Chuang
- Scientist Emeritus, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, 305 John Morgan Building 3620 Hamilton Walk, Philadelphia, PA, 19104, U.S.A..
| |
Collapse
|
6
|
Arndt H, Bachurski M, Yuanxiang P, Franke K, Wessjohann LA, Kreutz MR, Grochowska KM. A Screen of Plant-Based Natural Products Revealed That Quercetin Prevents Pyroglutamylated Amyloid-β (Aβ3(pE)-42) Uptake in Astrocytes As Well As Resulting Astrogliosis and Synaptic Dysfunction. Mol Neurobiol 2025; 62:3730-3745. [PMID: 39317890 PMCID: PMC11790700 DOI: 10.1007/s12035-024-04509-6] [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: 03/23/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Two connected histopathological hallmarks of Alzheimer's disease (AD) are chronic neuroinflammation and synaptic dysfunction. The accumulation of the most prevalent posttranslationally modified form of Aβ1-42, pyroglutamylated amyloid-β (Aβ3(pE)-42) in astrocytes is directly linked to glial activation and the release of proinflammatory cytokines that in turn contribute to early synaptic dysfunction in AD. At present, the mechanisms of Aβ3(pE)-42 uptake to astrocytes are unknown and pharmacological interventions that interfere with this process are not available. Here we developed a simple screening assay to identify substances from a plant extract library that prevent astroglial Aβ3(pE)-42 uptake. We first show that this approach yields valid and reproducible results. Second, we show endocytosis of Aβ3(pE)-42 oligomers by astrocytes and that quercetin, a plant flavonol, is effective to specifically block astrocytic buildup of oligomeric Aβ3(pE)-42. Importantly, quercetin does not induce a general impairment of endocytosis. However, it efficiently protects against early synaptic dysfunction following exogenous Aβ3(pE)-42 application.
Collapse
Affiliation(s)
- Helene Arndt
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Mark Bachurski
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - PingAn Yuanxiang
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Katrin Franke
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06108, Halle, Germany
- Institute of Biology/Geobotany and Botanical Garden, Martin Luther University Halle-Wittenberg, 06108, Halle, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103, Leipzig, Germany
| | - Ludger A Wessjohann
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, 06108, Halle, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, 04103, Leipzig, Germany
- Institut Für Chemie, Chair of Natural Products Chemistry, Martin-Luther-University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, 39120, Magdeburg, Germany.
| | - Katarzyna M Grochowska
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
7
|
Santos DE, Silva Lima SA, Moreira LS, Lima Costa S, de Sampaio Schitine C. New perspectives on heterogeneity in astrocyte reactivity in neuroinflammation. Brain Behav Immun Health 2025; 44:100948. [PMID: 40028234 PMCID: PMC11871470 DOI: 10.1016/j.bbih.2025.100948] [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: 11/30/2023] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
The inflammatory response is a fundamental aspect of all insults to the central nervous system (CNS), which includes acute trauma, infections, and chronic neurodegenerative conditions. As methods for investigating astrocytes have progressed, recent findings indicate that astrocytes can react to a diverse spectrum of insults affecting the central nervous system. Astrocytes respond to external and internal stimuli from the nervous system in a process called glial reactivity. Astrocyte reactivity, previously considered uniform and functionally inactive, is currently a very diverse event in different inflammatory processes. These differences can occur due to the nature, the intensity of the stimulus, the brain region involved and can range from subtle changes in astrocytic morphology to protein expression alteration, gene transcription profile shifts, and variations in the secretory pattern of molecules. The elucidation of the diverse roles of astrocytes in both normal and pathological conditions has led to increased interest in the notion that various astrocyte subtypes may exist, each contributing with distinct functions. Our study will prioritize the characterization of astrocytic response patterns in the context of the development and progression of neurodegenerative diseases, particularly Alzheimer's and Parkinson's. In addition, we will investigate the astrocyte's response during bacterial and viral infections, given the potential to enhance specific therapeutic interventions based on the reactivity profiles of astrocytes.
Collapse
Affiliation(s)
| | | | - Leticia Santos Moreira
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| | - Clarissa de Sampaio Schitine
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| |
Collapse
|
8
|
Al-Abbas NS, Shaer NA. Gut microbiome synthesizes important core metabolites to prevent cognitive decline and mitigate onset and progression of Alzheimer's disease. J Alzheimers Dis Rep 2024; 8:1705-1721. [PMID: 40034366 PMCID: PMC11863740 DOI: 10.1177/25424823241309024] [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: 09/13/2024] [Accepted: 11/24/2024] [Indexed: 03/05/2025] Open
Abstract
Background This study explores how gut metabolites, produced through bacterial metabolism in the gut, influence neurological conditions like Alzheimer's disease (AD). Key metabolites such as succinate and short-chain fatty acids signal through the autonomic nervous system and can cross the blood-brain barrier, impacting central nervous system functions. Objective The aim is to examine the role of the gut microbiota in compensating for metabolic deficiencies in AD. By analyzing wild-type (WT) and APP/PS1 mice, the study investigates how the microbiome affects key metabolic processes and whether it can slow AD progression. Methods High-throughput sequencing data from the gut microbiomes of APP/PS1 transgenic AD model mice and age-matched WT C57BL/6 male mice were analyzed for microbial and metabolite profiles. Results Alpha and beta diversity analyses showed differences in microbial composition between groups. Partial least squares discriminant analysis and Anosim confirmed distinct microbiome profiles in WT and APP/PS1 mice. At the genus level, Vescimonas was more abundant in WT mice, while Odoribacter, Lacrimispora, Helicobacter, Bacteroides, and Alloprevotella were more prevalent in APP/PS1 mice. Conclusions While taxonomic differences did not directly link specific microorganisms to AD, functional analysis identified key metabolites-acetyl-CoA, glucose, succinate, lipids, choline, and acetylcholine-that may alleviate energy deficits and synaptic dysfunction. This study suggests that the microbiome may help compensate for AD-related impairments, opening avenues for microbiome-based therapies.
Collapse
Affiliation(s)
- Nouf S Al-Abbas
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nehad A Shaer
- Department of Chemistry, Al Lieth University College, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
9
|
Bhuiyan P, Zhang W, Liang G, Jiang B, Vera R, Chae R, Kim K, Louis LS, Wang Y, Liu J, Chuang DM, Wei H. Intranasal Delivery of Lithium Salt Suppresses Inflammatory Pyroptosis in the brain and Ameliorates Memory Loss and Depression-like Behavior in 5XFAD mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613794. [PMID: 39345574 PMCID: PMC11430220 DOI: 10.1101/2024.09.18.613794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background Alzheimer's disease (AD) is a devastating neurodegenerative disease (AD) and has no treatment that can cure or halt the disease progression. This study explored the therapeutic potential of lithium salt dissolved in Ryanodex formulation vehicle (RFV) and delivered to the brain by intranasal application. We first compared lithium concentrations in the brain and blood of wild-type mice following intranasal or oral administration of lithium chloride (LiCl) dissolved in either RFV or water. The beneficial and side effects of intranasal versus oral LiCl in RFV in these mice were assessed and potential mechanisms underlying the efficacy of anti-inflammation and anti-pyroptosis in the brains were also investigated in both wild-type (WT) and 5XFAD Alzheimer's Disease (AD) mice brains. Methods For the study of brain versus blood lithium concentrations, WT B6SJLF1/J mice at 2 months of age were treated with intranasal or oral LiCl (3 mmol/kg) dissolved in RFV or in water. Brain and blood lithium concentrations were measured at various times after drugs administration. Brain/blood lithium concentration ratios were then determined. For studying therapeutic efficacy versus side effects and their underlying mechanisms, 5XFAD and WT B6SJLF1/J mice were treated with intranasal LiCl (3 mmol/kg) daily, Monday to Friday each week, in RFV beginning at 2 or 9 months of age with a 12-week treatment duration. Animal behaviors were assessed for depression (tail suspension), cognition (fear conditioning and Y maze), olfaction (buried food test), and motor functions (rotarod) at the age of 5 and 12 months. Blood and brain tissue were harvested from these mice at 13 months. Blood biomarkers for the functions of thyroid (thyroid stimulating hormone, TSH) and kidney (creatinine) were measured using ELISA. Changes in protein expression levels of the endoplasmic reticulum Ca2+ release channels type 1 InsP3 receptors (InsP3R-1), malondialdehyde (MDA)-modified proteins and 4-hydroxy-2-nonenal (4-HNE), pyroptosis regulatory proteins (NLR family pyrin domain containing 3 (NLRP3), cleaved caspase-1, N-terminal of Gasdermin D (GSDMD)), cytotoxic (IL-1β, IL-18, IL-6, TNF-α) and cytoprotective (IL-10) cytokines and synapse proteins (PSD-95, synapsin-1) were determined using immunoblotting. Mouse body weights were monitored regularly. Results Compared to oral LiCl in RFV nanoparticles, intranasal treatment of WT mice with LiCl in RFV markedly decreased blood concentrations at the time frame of 30-120 minutes. The ratio of brain/blood lithium concentration after Intranasal lithium chloride in RFV significantly increased, in comparison to those after oral administration lithium chloride in RFV or intranasal administration of lithium chloride in water. Intranasal lithium chloride in RFV inhibited both memory loss and depressive behavior in adult and aged 5XFAD mice. Additionally intranasal treatment of aged 5XFAD mice with LiCl in RFV effectively suppressed the increases in InsP3R-1, intracellular oxidative stress markers (4-HNE-bound and MDA-modified proteins), pyroptosis activation proteins (NLRP3, cleaved caspase-1, N-terminal GSDMD) and cytotoxic cytokines (IL-1β, IL-6, TNF-α), but reversed the down-regulation of cytoprotective cytokine IL-10. Intranasal LiCl in RFV also alleviated the loss of the postsynaptic synapse protein PSD-95, but not synapsin-1, in aged 5XFAD mice. Blood level of the kidney function marker creatinine was significantly increased in 5XFAD than in WT mice in an age-dependent manner and this elevation was abolished by intranasal delivery of LiCl in RFV. Intranasal LiCl in RFV for 12 weeks in both WT or 5XFAD mice did not affect blood biomarkers for thyroid function, nor did it affect smell or muscle function or body weight. Conclusion Intranasal administration of LiCl in RFV significantly decreased lithium blood concentrations and increased brain/blood lithium concentration ratio, in comparison to its oral administration. Intranasal administration of LiCl in RFV robustly protected against both memory loss and depressive-like behavior, while had no side effects concerning thyroid and kidney toxicity in 5XFAD mice. These lithium-induced beneficial effects were strongly associated with lithium's suppression of InsP3R-1 Ca2+ channel receptor increase, pathological neuroinflammation and activation of the pyroptosis pathway, as well as the loss of some synaptic proteins. Intranasal delivery of lithium salt in RFV could become an effective and potent inhibitor of pathological inflammation/pyroptosis in the CNS and serve as a new treatment for both AD-associated dementia and depression with minimal unwanted side effects including peripheral organ toxicity.
Collapse
Affiliation(s)
- Piplu Bhuiyan
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Wenjia Zhang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Bailin Jiang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
- Department of Anesthesiology, Peking University People’s Hospital, Beijing, China
| | - Robert Vera
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Rebecca Chae
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Kyulee Kim
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Lauren St. Louis
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| | - Ying Wang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Jia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 26600, P. R. China
| | - De-Maw Chuang
- Scientist Emeritus, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, U.S.A
| |
Collapse
|
10
|
Peretti DE, Boccalini C, Ribaldi F, Scheffler M, Marizzoni M, Ashton NJ, Zetterberg H, Blennow K, Frisoni GB, Garibotto V. Association of glial fibrillary acid protein, Alzheimer's disease pathology and cognitive decline. Brain 2024; 147:4094-4104. [PMID: 38940331 PMCID: PMC11629700 DOI: 10.1093/brain/awae211] [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: 03/01/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Increasing evidence shows that neuroinflammation is a possible modulator of tau spread effects on cognitive impairment in Alzheimer's disease. In this context, plasma levels of the glial fibrillary acidic protein (GFAP) have been suggested to have a robust association with Alzheimer's disease pathophysiology. This study aims to assess the correlation between plasma GFAP and Alzheimer's disease pathology, and their synergistic effect on cognitive performance and decline. A cohort of 122 memory clinic subjects with amyloid and tau PET, MRI scans, plasma GFAP and Mini-Mental State Examination (MMSE) was included in the study. A subsample of 94 subjects had a follow-up MMSE score at ≥1 year after baseline. Regional and voxel-based correlations between Alzheimer's disease biomarkers and plasma GFAP were assessed. Mediation analyses were performed to evaluate the effects of plasma GFAP on the association between amyloid and tau PET and between tau PET and cognitive impairment and decline. GFAP was associated with increased tau PET ligand uptake in the lateral temporal and inferior temporal lobes in a strong left-sided pattern independently of age, sex, education, amyloid and APOE status (β = 0.001, P < 0.01). The annual rate of MMSE change was significantly and independently correlated with both GFAP (β = 0.006, P < 0.01) and global tau standardized uptake value ratio (β = 4.33, P < 0.01), but not with amyloid burden. Partial mediation effects of GFAP were found on the association between amyloid and tau pathology (13.7%) and between tau pathology and cognitive decline (17.4%), but not on global cognition at baseline. Neuroinflammation measured by circulating GFAP is independently associated with tau Alzheimer's disease pathology and with cognitive decline, suggesting neuroinflammation as a potential target for future disease-modifying trials targeting tau pathology.
Collapse
Grants
- Private Foundation of Geneva University Hospitals
- Association Suisse pour la Recherche sur la Maladie d'Alzheimer, Genève
- Fondation Segré, Genève
- Race Against Dementia Foundation, London, UK
- Fondation Child Care, Genève
- Fondation Edmond J. Safra, Genève
- Fondation Minkoff, Genève
- Fondazione Agusta, Lugano
- McCall Macbain Foundation, Canada
- Nicole et René Keller, Genève
- Fondation AETAS, Genève
- Association Suisse pour la Recherche sur la Maladie d’Alzheimer, Genève
Collapse
Affiliation(s)
- Débora E Peretti
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocentre and Faculty of Medicine, University of Geneva, Geneva 1205, Switzerland
| | - Cecilia Boccalini
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocentre and Faculty of Medicine, University of Geneva, Geneva 1205, Switzerland
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva 1205, Switzerland
- Geneva Memory Centre, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva 1205, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva 1205, Switzerland
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia 25125, Italy
| | - Nicholas J Ashton
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger 4011, Norway
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal 413 90, Sweden
- King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RX, UK
- Mental Health & Biomedical Research Unit for Dementia, Maudsley NIHR Biomedical Research Centre, London SE5 8AF, UK
| | - Henrik Zetterberg
- Mental Health & Biomedical Research Unit for Dementia, Maudsley NIHR Biomedical Research Centre, London SE5 8AF, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London WC1N 3BG, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 413 45, Sweden
- Hong Kong Centre for Neurodegenerative Diseases, Clear Water Bay, Units 1501–1502, Hong Kong 1512–1518, China
- Wisconsin Alzheimer’s Disease Research Centre, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal 413 90, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 413 45, Sweden
- Paris Brain Institute, ICM, Pitié Salpêtrière Hospital, Sorbonne University, Paris 75013, France
- Neurodegenerative Disorder Research Centre, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei 230001, China
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva 1205, Switzerland
- Geneva Memory Centre, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva 1205, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocentre and Faculty of Medicine, University of Geneva, Geneva 1205, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals, Geneva 1205, Switzerland
- Centre for Biomedical Imaging, University of Geneva, Geneva 1205, Switzerland
| |
Collapse
|
11
|
McComish SF, O'Sullivan J, Copas AMM, Imiolek M, Boyle NT, Crompton LA, Lane JD, Caldwell MA. Reactive astrocytes generated from human iPSC are pro-inflammatory and display altered metabolism. Exp Neurol 2024; 382:114979. [PMID: 39357593 DOI: 10.1016/j.expneurol.2024.114979] [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: 06/21/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Astrocytes are the most abundant type of glial cell in the central nervous system and they play pivotal roles in both normal health and disease. Their dysfunction is detrimental to many brain related pathologies. Under pathological conditions, such as Alzheimer's disease, astrocytes adopt an activated reactive phenotype which can contribute to disease progression. A prominent risk factor for many neurodegenerative diseases is neuroinflammation which is the purview of glial cells, such as astrocytes and microglia. Human in vitro models have the potential to reveal relevant disease specific mechanisms, through the study of individual cell types such as astrocytes or the addition of specific factors, such as those secreted by microglia. The aim of this study was to generate human cortical astrocytes, in order to assess their protein and gene expression, examine their reactivity profile in response to exposure to the microglial secreted factors IL-1α, TNFα and C1q and assess their functionality in terms of calcium signalling and metabolism. The successfully differentiated and stimulated reactive astrocytes display increased IL-6, RANTES and GM-CSF secretion, and increased expression of genes associated with reactivity including, IL-6, ICAM1, LCN2, C3 and SERPINA3. Functional assessment of these reactive astrocytes showed a delayed and sustained calcium response to ATP and a concomitant decrease in the expression of connexin-43. Furthermore, it was demonstrated these astrocytes had an increased glycolytic capacity with no effect on oxidative phosphorylation. These findings not only increase our understanding of astrocyte reactivity but also provides a functional platform for drug discovery.
Collapse
Affiliation(s)
- Sarah F McComish
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Julia O'Sullivan
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Adina Mac Mahon Copas
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Magdalena Imiolek
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Noreen T Boyle
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lucy A Crompton
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK; Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Maeve A Caldwell
- Discipline of Physiology & School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
12
|
Zhang Y, Wang J, Zhang H, Tan Z, Zheng Y, Ping J, Zhang J, Luo J, Li L, Lu L, Liu X. Elevated circulating levels of GFAP associated with reduced volumes in hippocampal subregions linked to mild cognitive impairment among community-dwelling elderly individuals. Front Aging Neurosci 2024; 16:1461556. [PMID: 39534430 PMCID: PMC11554497 DOI: 10.3389/fnagi.2024.1461556] [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: 07/14/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Objective Cerebrospinal fluid biomarkers are challenging to use for diagnosing mild cognitive impairment (MCI) in large populations, and there is an urgent need for new blood biomarkers. The aim of this study is to investigate whether astrocyte activation is correlated with hippocampal atrophy, and to assess the potential of glial fibrillary acidic protein (GFAP) as a biomarker for diagnosing MCI among community-dwelling older individuals. Methods This cross-sectional study included 107 older adults. The levels of GFAP in serum were measured, and the volumetric assessment of gray matter within hippocampal subregions was conducted using Voxel-Based Morphometry (VBM). The relationship between hippocampal subregion volume and blood biomarkers were analyzed using partial correlation. The effectiveness of blood biomarkers in differentiating MCI was assessed using a receiver operating characteristic (ROC) curve. Results We found that serum GFAP levels were significantly elevated in the MCI group compared to the cognitively normal (CN) group. Additionally, individuals with MCI exhibited a reduction gray matter volume in specific hippocampal subregions. Notably, the right dentate gyrus (DG) and right cornu ammonis (CA) subregions were found to be effective for distinguishing MCI patients from CN individuals. Serum levels of GFAP demonstrate a sensitivity of 65.9% and a specificity of 75.6% in differentiating patients with MCI from CN individuals. Conclusion Specific atrophy within hippocampal subregions has been observed in the brains of community-dwelling elderly individuals. Elevated levels of circulating GFAP may serve as a sensitive peripheral biomarker indicative of hippocampal-specific cognitive alterations in patients with MCI.
Collapse
Affiliation(s)
- Ying Zhang
- School of Public Health, GuangDong Pharmaceutical University, Guangzhou, Guangdong, China
- Research Laboratory, The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Jun Wang
- Department of Radiology, Zhongshan Torch Development Zone People's Hospital, Zhongshan, Guangdong, China
| | - Haibo Zhang
- Department of Imaging, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| | - Zhenkun Tan
- School of Public Health, GuangDong Pharmaceutical University, Guangzhou, Guangdong, China
- Research Laboratory, The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
| | - Yingyan Zheng
- Zhongshan Torch High-Tech Industrial Development Zone of Community Health Service Center, Zhongshan, Guangdong, China
| | - Junjiao Ping
- Research Laboratory, The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| | - Jie Zhang
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| | - Jiali Luo
- Research Laboratory, The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| | - Linsen Li
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| | - Liming Lu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xinxia Liu
- School of Public Health, GuangDong Pharmaceutical University, Guangzhou, Guangdong, China
- Research Laboratory, The Third People's Hospital of Zhongshan, Zhongshan, Guangdong, China
- Department of Psychiatry, The Third People's Hospital of Zhongshan City, Zhongshan, Guangdong, China
| |
Collapse
|
13
|
Cerasuolo M, Di Meo I, Auriemma MC, Paolisso G, Papa M, Rizzo MR. Exploring the Dynamic Changes of Brain Lipids, Lipid Rafts, and Lipid Droplets in Aging and Alzheimer's Disease. Biomolecules 2024; 14:1362. [PMID: 39595539 PMCID: PMC11591903 DOI: 10.3390/biom14111362] [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: 09/06/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Aging induces complex changes in the lipid profiles across different areas of the brain. These changes can affect the function of brain cells and may contribute to neurodegenerative diseases such as Alzheimer's disease. Research shows that while the overall lipid profile in the human brain remains quite steady throughout adulthood, specific changes occur with age, especially after the age of 50. These changes include a slow decline in total lipid content and shifts in the composition of fatty acids, particularly in glycerophospholipids and cholesterol levels, which can vary depending on the brain region. Lipid rafts play a crucial role in maintaining membrane integrity and facilitating cellular signaling. In the context of Alzheimer's disease, changes in the composition of lipid rafts have been associated with the development of the disease. For example, alterations in lipid raft composition can lead to increased accumulation of amyloid β (Aβ) peptides, contributing to neurotoxic effects. Lipid droplets store neutral lipids and are key for cellular energy metabolism. As organisms age, the dynamics of lipid droplets in the brain change, with evidence suggesting a decline in metabolic activity over time. This reduced activity may lead to an imbalance in lipid synthesis and mobilization, contributing to neurodegenerative processes. In model organisms like Drosophila, studies have shown that lipid metabolism in the brain can be influenced by diet and insulin signaling pathways, crucial for maintaining metabolic balance. The interplay between lipid metabolism, oxidative stress, and inflammation is critical in the context of aging and Alzheimer's disease. Lipid peroxidation, a consequence of oxidative stress, can lead to the formation of reactive aldehydes that further damage neurons. Inflammatory processes can also disrupt lipid metabolism, contributing to the pathology of AD. Consequently, the accumulation of oxidized lipids can affect lipid raft integrity, influencing signaling pathways involved in neuronal survival and function.
Collapse
Affiliation(s)
- Michele Cerasuolo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Irene Di Meo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Maria Chiara Auriemma
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| | - Michele Papa
- Laboratory of Neuronal Networks Morphology and System Biology, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.C.); (I.D.M.); (M.C.A.); (G.P.)
| |
Collapse
|
14
|
Zhang R, Ohshima M, Brodin D, Wang Y, Morancé A, Schultzberg M, Chen G, Johansson J. Intravenous chaperone treatment of late-stage Alzheimer´s disease (AD) mouse model affects amyloid plaque load, reactive gliosis and AD-related genes. Transl Psychiatry 2024; 14:453. [PMID: 39448576 PMCID: PMC11502864 DOI: 10.1038/s41398-024-03161-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
Treatment strategies that are efficient against established Alzheimer's disease (AD) are needed. BRICHOS is a molecular chaperone domain that prevents amyloid fibril formation and associated cellular toxicity. In this study, we treated an AD mouse model seven months after pathology onset, using intravenous administration of recombinant human (rh) Bri2 BRICHOS R221E. Two injections of rh Bri2 BRICHOS R221E per week for three months in AD mice reduced amyloid β (Aβ) burden, and mitigated astro- and microgliosis, as determined by glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule 1 (Iba1) immunohistochemistry. Sequencing of RNA from cortical microglia cells showed that BRICHOS treatment normalized the expression of identified plaque-induced genes in mice and humans, including clusterin and GFAP. Rh Bri2 BRICHOS R221E passed the blood-brain barrier (BBB) in age-matched wild-type mice as efficiently as in the AD mice, but then had no effect on measures of AD-like pathology, and mainly affected the expression of genes that affect cellular shape and movement. These results indicate a potential of rh Bri2 BRICHOS against advanced AD and underscore the ability of BRICHOS to target amyloid-induced pathology.
Collapse
Affiliation(s)
- Ruixin Zhang
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Makiko Ohshima
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden
| | - David Brodin
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Yu Wang
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Antonin Morancé
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden
- Department of Neuroscience, University of Mons (UMONS), Mons, Belgium
| | - Marianne Schultzberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences & Society, Karolinska Institutet, Solna, Sweden.
| | - Gefei Chen
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden.
| | - Jan Johansson
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
15
|
Pellitteri R, La Cognata V, Russo C, Patti A, Sanfilippo C. Protective Role of Eicosapentaenoic and Docosahexaenoic and Their N-Ethanolamide Derivatives in Olfactory Glial Cells Affected by Lipopolysaccharide-Induced Neuroinflammation. Molecules 2024; 29:4821. [PMID: 39459191 PMCID: PMC11510059 DOI: 10.3390/molecules29204821] [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: 09/03/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Neuroinflammation is a symptom of different neurodegenerative diseases, and growing interest is directed towards active drug development for the reduction of its negative effects. The anti-inflammatory activity of polyunsaturated fatty acids, eicosapentaenoic (EPA), docosahexaenoic (DHA), and their amide derivatives was largely investigated on some neural cells. Herein, we aimed to elucidate the protective role of both EPA and DHA and the corresponding N-ethanolamides EPA-EA and DHA-EA on neonatal mouse Olfactory Ensheathing Cells (OECs) after exposition to lipopolysaccharide (LPS)-induced neuroinflammation. To verify their anti-inflammatory effect and cell morphological features on OECs, the expression of IL-10 cytokine, and cytoskeletal proteins (vimentin and GFAP) was evaluated by immunocytochemical procedures. In addition, MTT assays, TUNEL, and mitochondrial health tests were carried out to assess their protective effects on OEC viability. Our results highlight a reduction in GFAP and vimentin expression in OECs exposed to LPS and treated with EPA or DHA or EPA-EA or DHA-EA in comparison with OECs exposed to LPS alone. We observed a protective role of EPA and DHA on cell morphology, while the amides EPA-EA and DHA-EA mainly exerted a superior anti-inflammatory effect compared to free acids.
Collapse
Affiliation(s)
- Rosalia Pellitteri
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, I-95126 Catania, Italy;
| | - Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council, Via P. Gaifami 18, I-95126 Catania, Italy;
| | - Cristina Russo
- Department of Biomedical and Biotechnological Sciences, Section of Pathology, University of Catania, Via Santa Sofia 97, I-95123 Catania, Italy;
| | - Angela Patti
- Institute of Biomolecular Chemistry, National Research Council, Via P. Gaifami 18, I-95126 Catania, Italy;
| | - Claudia Sanfilippo
- Institute of Biomolecular Chemistry, National Research Council, Via P. Gaifami 18, I-95126 Catania, Italy;
| |
Collapse
|
16
|
Lanni I, Chiacchierini G, Papagno C, Santangelo V, Campolongo P. Treating Alzheimer's disease with brain stimulation: From preclinical models to non-invasive stimulation in humans. Neurosci Biobehav Rev 2024; 165:105831. [PMID: 39074672 DOI: 10.1016/j.neubiorev.2024.105831] [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: 05/20/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Alzheimer's disease (AD) is a severe and progressive neurodegenerative condition that exerts detrimental effects on brain function. As of now, there is no effective treatment for AD patients. This review explores two distinct avenues of research. The first revolves around the use of animal studies and preclinical models to gain insights into AD's underlying mechanisms and potential treatment strategies. Specifically, it delves into the effectiveness of interventions such as Optogenetics and Chemogenetics, shedding light on their implications for understanding pathophysiological mechanisms and potential therapeutic applications. The second avenue focuses on non-invasive brain stimulation (NiBS) techniques in the context of AD. Evidence suggests that NiBS can successfully modulate cognitive functions associated with various neurological and neuropsychiatric disorders, including AD, as demonstrated by promising findings. Here, we critically assessed recent findings in AD research belonging to these lines of research and discuss their potential impact on the clinical horizon of AD treatment. These multifaceted approaches offer hope for advancing our comprehension of AD pathology and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Ilenia Lanni
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Giulia Chiacchierini
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Costanza Papagno
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Valerio Santangelo
- Functional Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy; Department of Philosophy, Social Sciences & Education, University of Perugia, Perugia, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Behavioral Neuropharmacology Unit, IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
17
|
Qin H, Zhou L, Haque FT, Martin-Jimenez C, Trang A, Benveniste EN, Wang Q. Diverse signaling mechanisms and heterogeneity of astrocyte reactivity in Alzheimer's disease. J Neurochem 2024; 168:3536-3557. [PMID: 37932959 PMCID: PMC11839148 DOI: 10.1111/jnc.16002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
Alzheimer's disease (AD) affects various brain cell types, including astrocytes, which are the most abundant cell types in the central nervous system (CNS). Astrocytes not only provide homeostatic support to neurons but also actively regulate synaptic signaling and functions and become reactive in response to CNS insults through diverse signaling pathways including the JAK/STAT, NF-κB, and GPCR-elicited pathways. The advent of new technology for transcriptomic profiling at the single-cell level has led to increasing recognition of the highly versatile nature of reactive astrocytes and the context-dependent specificity of astrocyte reactivity. In AD, reactive astrocytes have long been observed in senile plaques and have recently been suggested to play a role in AD pathogenesis and progression. However, the precise contributions of reactive astrocytes to AD remain elusive, and targeting this complex cell population for AD treatment poses significant challenges. In this review, we summarize the current understanding of astrocyte reactivity and its role in AD, with a particular focus on the signaling pathways that promote astrocyte reactivity and the heterogeneity of reactive astrocytes. Furthermore, we explore potential implications for the development of therapeutics for AD. Our objective is to shed light on the complex involvement of astrocytes in AD and offer insights into potential therapeutic targets and strategies for treating and managing this devastating neurodegenerative disorder.
Collapse
Affiliation(s)
- Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA 35294
| | - Lianna Zhou
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA 35294
| | - Faris T. Haque
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA 35294
| | - Cynthia Martin-Jimenez
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA 30912
| | - Amy Trang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA 30912
| | - Etty N. Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA 35294
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA 30912
| |
Collapse
|
18
|
Ozcelikay-Akyildiz G, Karadurmus L, Cetinkaya A, Uludag İ, Ozcan B, Unal MA, Sezginturk MK, Ozkan SA. The Evaluation of Clinical Applications for the Detection of the Alzheimer's Disease Biomarker GFAP. Crit Rev Anal Chem 2024:1-12. [PMID: 39178137 DOI: 10.1080/10408347.2024.2393874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
One of the most prevalent neurodegenerative diseases is Alzheimer's disease (AD). The hallmarks of AD include the accumulation of amyloid plaques and neurofibrillary tangles, which cause related secondary diseases, progressive neurodegeneration, and ultimately death. The most prevalent cell type in the human central nervous system, astrocytes, are crucial for controlling neuronal function. Glial fibrillary acidic protein (GFAP) is released from tissue into the bloodstream due to astrocyte breakdown in neurological diseases. Increased levels of GFAP in the serum can function as blood markers and be an effective prognostic indicator to help diagnose neurological conditions early on, from stroke to neurodegenerative diseases. The human central nervous system (CNS) is greatly affected by diseases associated with blood GFAP levels. These include multiple sclerosis, intracerebral hemorrhage, glioblastoma multiforme, traumatic brain injuries, and neuromyelitis optica. GFAP demonstrates a strong diagnostic capacity for projecting outcomes following an injury. Furthermore, the increased ability to identify GFAP protein fragments helps facilitate treatment, as it allows continuous screening of CNS injuries and early identification of potential recurrences. GFAP has recently gained attention due to data showing that the plasma biomarker is effective in identifying AD pathology. AD accounts for 60-70% of the approximately 50 million people with dementia worldwide. It is critical to develop molecular markers for AD, whose number is expected to increase to about 3 times and affect humans by 2050, and to investigate possible targets to confirm their effectiveness in the early diagnosis of AD. In addition, most diagnostic methods currently used are image-based and do not detect early disease, i.e. before symptoms appear; thus, treatment options and outcomes are limited. Therefore, recently developed methods such as point-of-care (POC), on-site applications, and enzyme-linked immunosorbent assay-polymerase chain reaction (ELISA-PCR) that provide both faster and more accurate results are gaining importance. This systematic review summarizes published studies with different approaches such as immunosensor, lateral flow, POC, ELISA-PCR, and molecularly imprinted polymer using GFAP, a potential blood biomarker to detect neurological disorders. Here, we also provide an overview of current approaches, analysis methods, and different future detection strategies for GFAP, the most popular biosensing field.
Collapse
Affiliation(s)
| | - Leyla Karadurmus
- Department of Analytical Chemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman, Türkiye
| | - Ahmet Cetinkaya
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
| | - İnci Uludag
- Bioengineering Department, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Burcu Ozcan
- Bioengineering Department, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | | | - Mustafa Kemal Sezginturk
- Bioengineering Department, Faculty of Engineering, Çanakkale Onsekiz Mart University, Çanakkale, Türkiye
| | - Sibel A Ozkan
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
| |
Collapse
|
19
|
Thomas SD, Abdalla S, Eissa N, Akour A, Jha NK, Ojha S, Sadek B. Targeting Microglia in Neuroinflammation: H3 Receptor Antagonists as a Novel Therapeutic Approach for Alzheimer's Disease, Parkinson's Disease, and Autism Spectrum Disorder. Pharmaceuticals (Basel) 2024; 17:831. [PMID: 39065682 PMCID: PMC11279978 DOI: 10.3390/ph17070831] [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: 05/15/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Histamine performs dual roles as an immune regulator and a neurotransmitter in the mammalian brain. The histaminergic system plays a vital role in the regulation of wakefulness, cognition, neuroinflammation, and neurogenesis that are substantially disrupted in various neurodegenerative and neurodevelopmental disorders. Histamine H3 receptor (H3R) antagonists and inverse agonists potentiate the endogenous release of brain histamine and have been shown to enhance cognitive abilities in animal models of several brain disorders. Microglial activation and subsequent neuroinflammation are implicated in impacting embryonic and adult neurogenesis, contributing to the development of Alzheimer's disease (AD), Parkinson's disease (PD), and autism spectrum disorder (ASD). Acknowledging the importance of microglia in both neuroinflammation and neurodevelopment, as well as their regulation by histamine, offers an intriguing therapeutic target for these disorders. The inhibition of brain H3Rs has been found to facilitate a shift from a proinflammatory M1 state to an anti-inflammatory M2 state, leading to a reduction in the activity of microglial cells. Also, pharmacological studies have demonstrated that H3R antagonists showed positive effects by reducing the proinflammatory biomarkers, suggesting their potential role in simultaneously modulating crucial brain neurotransmissions and signaling cascades such as the PI3K/AKT/GSK-3β pathway. In this review, we highlight the potential therapeutic role of the H3R antagonists in addressing the pathology and cognitive decline in brain disorders, e.g., AD, PD, and ASD, with an inflammatory component.
Collapse
Affiliation(s)
- Shilu Deepa Thomas
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.D.T.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 1551, United Arab Emirates
| | - Sabna Abdalla
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.D.T.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 1551, United Arab Emirates
| | - Nermin Eissa
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.D.T.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 1551, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
| | - Shreesh Ojha
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.D.T.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 1551, United Arab Emirates
| | - Bassem Sadek
- Department of Pharmacology & Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.D.T.); (S.A.)
- Zayed Center for Health Sciences, United Arab Emirates University, Al-Ain P.O. Box 1551, United Arab Emirates
| |
Collapse
|
20
|
Lombardi AM, Wong H, Bower ME, Milstead R, Borski C, Schmitt E, Griffioen M, LaPlante L, Ehringer MA, Stitzel J, Hoeffer CA. AKT2 modulates astrocytic nicotine responses in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596856. [PMID: 38854016 PMCID: PMC11160815 DOI: 10.1101/2024.05.31.596856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
A better understanding of nicotine neurobiology is needed to reduce or prevent chronic addiction, ameliorate the detrimental effects of nicotine withdrawal, and increase successful cessation of use. Nicotine binds and activates two astrocyte-expressed nicotinic acetylcholine receptors (nAChRs), α4β2 and α7. We recently found that Protein kinase B-β (Pkb-β or Akt2) expression is restricted to astrocytes in mice and humans. To determine if AKT2 plays a role in astrocytic nicotinic responses, we generated astrocyte-specific Akt2 conditional knockout (cKO) and full Akt2 KO mice for in vivo and in vitro experiments. For in vivo studies, we examined mice exposed to chronic nicotine for two weeks in drinking water (200 μg/mL) and following acute nicotine challenge (0.09, 0.2 mg/kg) after 24 hrs. Our in vitro studies used cultured mouse astrocytes to measure nicotine-dependent astrocytic responses. We validated our approaches using lipopolysaccharide (LPS) exposure inducing astrogliosis. Sholl analysis was used to measure glial fibrillary acidic protein responses in astrocytes. Our data show that wild-type (WT) mice exhibit increased astrocyte morphological complexity during acute nicotine exposure, with decreasing complexity during chronic nicotine use, whereas Akt2 cKO mice showed increased astrocyte morphology complexity. In culture, we found that 100μM nicotine was sufficient for morphological changes and blocking α7 or α4β2 nAChRs prevented observed morphologic changes. Finally, we performed conditioned place preference (CPP) in Akt2 cKO mice and found that astrocytic AKT2 deficiency reduced nicotine preference compared to controls. These findings show the importance of nAChRs and Akt2 signaling in the astrocytic response to nicotine.
Collapse
Affiliation(s)
- Andrew M. Lombardi
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Helen Wong
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Myra E. Bower
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Ryan Milstead
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Emily Schmitt
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Mina Griffioen
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Lauren LaPlante
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Marissa A. Ehringer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Jerry Stitzel
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Charles A. Hoeffer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
- Linda Crnic Institute, Anschutz Medical Center, Aurora, CO 80045
| |
Collapse
|
21
|
Zeng X, Chen Y, Sehrawat A, Lee J, Lafferty TK, Kofler J, Berman SB, Sweet RA, Tudorascu DL, Klunk WE, Ikonomovic MD, Pfister A, Zetterberg H, Snitz BE, Cohen AD, Villemagne VL, Pascoal TA, Kamboh ML, Lopez OI, Blennow K, Karikari TK. Alzheimer blood biomarkers: practical guidelines for study design, sample collection, processing, biobanking, measurement and result reporting. Mol Neurodegener 2024; 19:40. [PMID: 38750570 PMCID: PMC11095038 DOI: 10.1186/s13024-024-00711-1] [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: 07/25/2023] [Accepted: 02/13/2024] [Indexed: 05/19/2024] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, remains challenging to understand and treat despite decades of research and clinical investigation. This might be partly due to a lack of widely available and cost-effective modalities for diagnosis and prognosis. Recently, the blood-based AD biomarker field has seen significant progress driven by technological advances, mainly improved analytical sensitivity and precision of the assays and measurement platforms. Several blood-based biomarkers have shown high potential for accurately detecting AD pathophysiology. As a result, there has been considerable interest in applying these biomarkers for diagnosis and prognosis, as surrogate metrics to investigate the impact of various covariates on AD pathophysiology and to accelerate AD therapeutic trials and monitor treatment effects. However, the lack of standardization of how blood samples and collected, processed, stored analyzed and reported can affect the reproducibility of these biomarker measurements, potentially hindering progress toward their widespread use in clinical and research settings. To help address these issues, we provide fundamental guidelines developed according to recent research findings on the impact of sample handling on blood biomarker measurements. These guidelines cover important considerations including study design, blood collection, blood processing, biobanking, biomarker measurement, and result reporting. Furthermore, the proposed guidelines include best practices for appropriate blood handling procedures for genetic and ribonucleic acid analyses. While we focus on the key blood-based AD biomarkers for the AT(N) criteria (e.g., amyloid-beta [Aβ]40, Aβ42, Aβ42/40 ratio, total-tau, phosphorylated-tau, neurofilament light chain, brain-derived tau and glial fibrillary acidic protein), we anticipate that these guidelines will generally be applicable to other types of blood biomarkers. We also anticipate that these guidelines will assist investigators in planning and executing biomarker research, enabling harmonization of sample handling to improve comparability across studies.
Collapse
Affiliation(s)
- Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Yijun Chen
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Jihui Lee
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Tara K Lafferty
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Julia Kofler
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sarah B Berman
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Robert A Sweet
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dana L Tudorascu
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - William E Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Milos D Ikonomovic
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Anna Pfister
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Beth E Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anne D Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Victor L Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - M. llyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Oscar I Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA.
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden.
| |
Collapse
|
22
|
Zakeri Z, Heiderzadeh M, Kocaarslan A, Metin E, Hosseini Karimi SN, Saghati S, Vural A, Akyoldaş G, Baysal K, Yağcı Y, Gürsoy-Özdemir Y, Taşoğlu S, Rahbarghazi R, Sokullu E. Exosomes encapsulated in hydrogels for effective central nervous system drug delivery. Biomater Sci 2024; 12:2561-2578. [PMID: 38602364 DOI: 10.1039/d3bm01055d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
The targeted delivery of pharmacologically active molecules, metabolites, and growth factors to the brain parenchyma has become one of the major challenges following the onset of neurodegeneration and pathological conditions. The therapeutic effect of active biomolecules is significantly impaired after systemic administration in the central nervous system (CNS) because of the blood-brain barrier (BBB). Therefore, the development of novel therapeutic approaches capable of overcoming these limitations is under discussion. Exosomes (Exo) are nano-sized vesicles of endosomal origin that have a high distribution rate in biofluids. Recent advances have introduced Exo as naturally suitable bio-shuttles for the delivery of neurotrophic factors to the brain parenchyma. In recent years, many researchers have attempted to regulate the delivery of Exo to target sites while reducing their removal from circulation. The encapsulation of Exo in natural and synthetic hydrogels offers a valuable strategy to address the limitations of Exo, maintaining their integrity and controlling their release at a desired site. Herein, we highlight the current and novel approaches related to the application of hydrogels for the encapsulation of Exo in the field of CNS tissue engineering.
Collapse
Affiliation(s)
- Ziba Zakeri
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Morteza Heiderzadeh
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | - Azra Kocaarslan
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Ecem Metin
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
| | | | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atay Vural
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Göktuğ Akyoldaş
- Department of Neurosurgery, Koç University Hospital, Istanbul 34450, Turkey
| | - Kemal Baysal
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Biochemistry, School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Yusuf Yağcı
- Chemistry Department, Faculty of Science, İstanbul Technical University, İstanbul, Turkey
| | - Yasemin Gürsoy-Özdemir
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Department of Neurology, School of Medicine, KoÒ« University, Istanbul 34450, Turkey
| | - Savaş Taşoğlu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Mechanical Engineering Department, School of Engineering, Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emel Sokullu
- Research Center for Translational Medicine (KUTTAM), Koç University, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey.
- Biophysics Department, Koç University School of Medicine, Rumeli Feneri, 34450, Istanbul, Sariyer, Turkey
| |
Collapse
|
23
|
Cao Y, Liu P, Bian H, Jin S, Liu J, Yu N, Cui H, Sun F, Qian X, Qiu W, Ma C. Reduced neurogenesis in human hippocampus with Alzheimer's disease. Brain Pathol 2024; 34:e13225. [PMID: 38012054 PMCID: PMC11007046 DOI: 10.1111/bpa.13225] [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: 06/14/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Adult hippocampal neurogenesis (AHN), essential for the plasticity of hippocampal structure and function, may be disrupted in Alzheimer's disease (AD). However, the relationship between the changes in AHN and AD-related pathology in humans remains uncertain. By utilizing advanced immunostaining techniques, we could identify multiple biomarkers representing different stages of AHN in postmortem human hippocampal tissue that exhibited various AD-related neuropathological changes. In this study, we observed a significant presence of neurogenic cells in the hippocampus's dentate gyrus (DG) region in 30 individuals, including 14 individuals diagnosed with AD-related neuropathological changes and the remaining 16 individuals without any neurological diseases. Further investigation revealed that patients with AD exhibited pronounced astrogliosis and reduced neurogenesis. Specifically, the number of neuroblasts, immature and early mature granule cells decreased significantly as AD advanced. Although the number of neural stem cells (NSCs) remained unchanged in AD patients compared with mentally healthy individuals, they tended to be more quiescent state regulated by Notch and bone morphogenetic protein (BMP) signaling pathways. These abnormalities were strongly associated with the neuropathological alterations in AD patients. These research findings provide potential insights into the underlying mechanisms that underpin the pathogenesis of AD.
Collapse
Affiliation(s)
- Yan Cao
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Pan Liu
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Department of Radiation and Medical Oncology, Zhongnan HospitalWuhan UniversityWuhanChina
| | - Hongfei Bian
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Sixuan Jin
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Jiaqi Liu
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Ning Yu
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Huan Cui
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Fengrun Sun
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Xiaojing Qian
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Wenying Qiu
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
| | - Chao Ma
- National Human Brain Bank for Development and Function, Department of Human Anatomy, Histology and Embryology, Neuroscience CenterInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| |
Collapse
|
24
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Chong JR, Chai YL, Yam ATY, Hilal S, Vrooman H, Venketasubramanian N, Blennow K, Zetterberg H, Ashton NJ, Chen CP, Lai MKP. Association of plasma GFAP with elevated brain amyloid is dependent on severity of white matter lesions in an Asian cognitively impaired cohort. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12576. [PMID: 38605996 PMCID: PMC11007806 DOI: 10.1002/dad2.12576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/25/2024] [Accepted: 03/01/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION While elevated blood glial fibrillary acidic protein (GFAP) has been associated with brain amyloid pathology, whether this association occurs in populations with high cerebral small vessel disease (CSVD) concomitance remains unclear. METHODS Using a Singapore-based cohort of cognitively impaired subjects, we assessed associations between plasma GFAP and neuroimaging measures of brain amyloid and CSVD, including white matter hyperintensities (WMH). We also examined the diagnostic performance of plasma GFAP in detecting brain amyloid beta positivity (Aβ+). RESULTS When stratified by WMH status, elevated brain amyloid was associated with higher plasma GFAP only in the WMH- group (β = 0.383; P < 0.001). The diagnostic performance of plasma GFAP in identifying Aβ+ was significantly higher in the WMH- group (area under the curve [AUC] = 0.896) than in the WMH+ group (AUC = 0.712, P = 0.008). DISCUSSION The biomarker utility of plasma GFAP in detecting brain amyloid pathology is dependent on the severity of concomitant WMH. Highlight Glial fibrillary acidic protein (GFAP)'s association with brain amyloid is unclear in populations with high cerebral small vessel disease (CSVD).Plasma GFAP was measured in a cohort with CSVD and brain amyloid.Plasma GFAP was better in detecting amyloid in patients with low CSVD versus high CSVD.Biomarker utility of GFAP in detecting brain amyloid depends on the severity of CSVD.
Collapse
Affiliation(s)
- Joyce R. Chong
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Yuek Ling Chai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Amelia T. Y. Yam
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Saima Hilal
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemKent RidgeSingapore
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Henri Vrooman
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
- Department of Neurodegenerative DiseaseThe UCL Queen Square Institute of NeurologyLondonUK
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGöteborgSweden
| | - Christopher P. Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| |
Collapse
|
26
|
DeSimone JC, Wang W, Loewenstein DA, Duara R, Smith GE, McFarland KN, Armstrong MJ, Weber DM, Barker W, Coombes SA, Vaillancourt DE. Diffusion MRI relates to plasma Aβ42/40 in PET negative participants without dementia. Alzheimers Dement 2024; 20:2830-2842. [PMID: 38441274 PMCID: PMC11032550 DOI: 10.1002/alz.13693] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 03/10/2024]
Abstract
INTRODUCTION Magnetic resonance imaging (MRI) biomarkers are needed for indexing early biological stages of Alzheimer's disease (AD), such as plasma amyloid-β (Aβ42/40) positivity in Aβ positron emission tomography (PET) negative individuals. METHODS Diffusion free-water (FW) MRI was acquired in individuals with normal cognition (NC) and mild cognitive impairment (MCI) with Aβ plasma-/PET- (NC = 22, MCI = 60), plasma+/PET- (NC = 5, MCI = 20), and plasma+/PET+ (AD dementia = 21) biomarker status. Gray and white matter FW and fractional anisotropy (FAt) were compared cross-sectionally and the relationships between imaging, plasma and PET biomarkers were assessed. RESULTS Plasma+/PET- demonstrated increased FW (24 regions) and decreased FAt (66 regions) compared to plasma-/PET-. FW (16 regions) and FAt (51 regions) were increased in plasma+/PET+ compared to plasma+/PET-. Composite brain FW correlated with plasma Aβ42/40 and p-tau181. DISCUSSION FW imaging changes distinguish plasma Aβ42/40 positive and negative groups, independent of group differences in cognitive status, Aβ PET status, and other plasma biomarkers (i.e., t-tau, p-tau181, glial fibrillary acidic protein, neurofilament light). HIGHLIGHTS Plasma Aβ42/40 positivity is associated with brain microstructure decline. Plasma+/PET- demonstrated increased FW in 24 total GM and WM regions. Plasma+/PET- demonstrated decreased FAt in 66 total GM and WM regions. Whole-brain FW correlated with plasma Aβ42/40 and p-tau181 measures. Plasma+/PET- demonstrated decreased cortical volume and thickness.
Collapse
Affiliation(s)
- Jesse C. DeSimone
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
| | - Wei‐en Wang
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
| | - David A. Loewenstein
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Center for Cognitive Neuroscience and AgingUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Ranjan Duara
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai Medical CenterMiami BeachFloridaUSA
| | - Glenn E. Smith
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Department of Clinical and Health PsychologyUniversity of FloridaGainesvilleFloridaUSA
| | - Karen N. McFarland
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Department of NeurologyUniversity of FloridaGainesvilleFloridaUSA
| | - Melissa J. Armstrong
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Department of NeurologyUniversity of FloridaGainesvilleFloridaUSA
- Norman Fixel Institute for Neurological DiseasesUniversity of FloridaGainesvilleFloridaUSA
| | - Darren M. Weber
- Quest Diagnostics Nichols InstituteSan Juan CapistranoCaliforniaUSA
| | - Warren Barker
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Wien Center for Alzheimer's Disease and Memory DisordersMount Sinai Medical CenterMiami BeachFloridaUSA
| | - Stephen A. Coombes
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of FloridaGainesvilleFloridaUSA
| | - David E. Vaillancourt
- Department of Applied Physiology and KinesiologyUniversity of FloridaGainesvilleFloridaUSA
- 1Florida Alzheimer's Disease Research CenterGainesvilleFloridaUSA
- Department of NeurologyUniversity of FloridaGainesvilleFloridaUSA
- Norman Fixel Institute for Neurological DiseasesUniversity of FloridaGainesvilleFloridaUSA
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
27
|
Griñán‐Ferré C, Jarné‐Ferrer J, Bellver‐Sanchís A, Codony S, Puigoriol‐Illamola D, Sanfeliu C, Oh Y, Lee S, Vázquez S, Pallàs M. Novel molecular mechanism driving neuroprotection after soluble epoxide hydrolase inhibition: Insights for Alzheimer's disease therapeutics. CNS Neurosci Ther 2024; 30:e14511. [PMID: 37905690 PMCID: PMC11017401 DOI: 10.1111/cns.14511] [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: 02/16/2023] [Revised: 09/21/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Neuroinflammation is widely recognized as a significant hallmark of Alzheimer's disease (AD). To combat neuroinflammation, the inhibition of the soluble epoxide hydrolase (sEH) enzyme has been demonstrated crucial. Importantly, sEH inhibition could be related to other neuroprotective pathways described in AD. AIMS The aim of the study was to unveil new molecular pathways driving neuroprotection through sEH, we used an optimized, potent, and selective sEH inhibitor (sEHi, UB-SCG-51). MATERIALS AND METHODS UB-SCG-51 was tested in neuroblastoma cell line, SH-SY5Y, in primary mouse and human astrocytes cultures challenged with proinflammatory insults and in microglia cultures treated with amyloid oligomers, as well as in mice AD model (5XFAD). RESULTS UB-SCG-51 (10 and 30 μM) prevented neurotoxic reactive-astrocyte conversion in primary mouse astrocytes challenged with TNF-α, IL-1α, and C1q (T/I/C) combination for 24 h. Moreover, in microglial cultures, sEHi reduced inflammation and glial activity. In addition, UB-SCG-51 rescued 5XFAD cognitive impairment, reducing the number of Amyloid-β plaques and Tau hyperphosphorylation accompanied by a reduction in neuroinflammation and apoptotic markers. Notably, a transcriptional profile analysis revealed a new pathway modulated by sEHi treatment. Specifically, the eIF2α/CHOP pathway, which promoted the endoplasmic reticulum response, was increased in the 5XFAD-treated group. These findings were confirmed in human primary astrocytes by combining sEHi and eIF2α inhibitor (eIF2αi) treatment. Besides, combining both treatments resulted in increased in C3 gene expression after T/I/C compared with the group treated with sEHi alone in cultures. DISCUSSION Therefore, sEHi rescued cognitive impairment and neurodegeneration in AD mice model, based on the reduction of inflammation and eIF2α/CHOP signaling pathway. CONCLUSIONS In whole, our results support the concept that targeting neuroinflammation through sEH inhibition is a promising therapeutic strategy to fight against Alzheimer's disease with additive and/or synergistic activities targeting neuroinflammation and cell stress.
Collapse
Affiliation(s)
- Christian Griñán‐Ferré
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos IIIMadridSpain
| | - Júlia Jarné‐Ferrer
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Aina Bellver‐Sanchís
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Sandra Codony
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB)University of Barcelona (UB)BarcelonaSpain
| | - Dolors Puigoriol‐Illamola
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC)BarcelonaSpain
| | - Yumin Oh
- Neuraly Inc.MarylandGaithersburgUSA
| | | | - Santiago Vázquez
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB)University of Barcelona (UB)BarcelonaSpain
| | - Mercè Pallàs
- Department of Pharmacology and Therapeutic ChemistryInstitut de Neurociències‐Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
28
|
Hossain MK, Kim HR, Chae HJ. Aging phenotype in AD brain organoids: Track to success and challenges. Ageing Res Rev 2024; 96:102256. [PMID: 38460555 DOI: 10.1016/j.arr.2024.102256] [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: 10/01/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/11/2024]
Abstract
Alzheimer's disease (AD) poses a complex challenge, with abnormal protein accumulation in the brain causing memory loss and cognitive decline. Traditional models fall short in AD research, prompting interest in 3D brain organoids (BOs) from human stem cells. These findings hold promise for unveiling the mechanisms of AD, especially in relation to aging. However, an understanding of the aging impact of AD remains elusive. BOs offer insight but face challenges. This review delves into the role of BOs in deciphering aging-related AD and acknowledges limitations. Strategies to enhance BOs for accurate aging modeling in AD brains are suggested. Strengthened by molecular advancements, BOs have the potential to uncover the aging phenotype, advancing AD research.
Collapse
Affiliation(s)
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| | - Han Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| |
Collapse
|
29
|
Mazzeo S, Ingannato A, Giacomucci G, Bagnoli S, Cavaliere A, Moschini V, Balestrini J, Morinelli C, Galdo G, Emiliani F, Piazzesi D, Crucitti C, Frigerio D, Polito C, Berti V, Padiglioni S, Sorbi S, Nacmias B, Bessi V. The role of plasma neurofilament light chain and glial fibrillary acidic protein in subjective cognitive decline and mild cognitive impairment. Neurol Sci 2024; 45:1031-1039. [PMID: 37723371 PMCID: PMC10857957 DOI: 10.1007/s10072-023-07065-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023]
Abstract
INTRODUCTION AND AIM NfL and GFAP are promising blood-based biomarkers for Alzheimer's disease. However, few studies have explored plasma GFAP in the prodromal and preclinical stages of AD. In our cross-sectional study, our aim is to investigate the role of these biomarkers in the earliest stages of AD. MATERIALS AND METHODS We enrolled 40 patients (11 SCD, 21 MCI, 8 AD dementia). All patients underwent neurological and neuropsychological examinations, analysis of CSF biomarkers (Aβ42, Aβ42/Aβ40, p-tau, t-tau), Apolipoprotein E (APOE) genotype analysis and measurement of plasma GFAP and NfL concentrations. Patients were categorized according to the ATN system as follows: normal AD biomarkers (NB), carriers of non-Alzheimer's pathology (non-AD), prodromal AD, or AD with dementia (AD-D). RESULTS GFAP was lower in NB compared to prodromal AD (p = 0.003, d = 1.463) and AD-D (p = 0.002, d = 1.695). NfL was lower in NB patients than in AD-D (p = 0.011, d = 1.474). NfL demonstrated fair accuracy (AUC = 0.718) in differentiating between NB and prodromal AD, with a cut-off value of 11.65 pg/mL. GFAP showed excellent accuracy in differentiating NB from prodromal AD (AUC = 0.901) with a cut-off level of 198.13 pg/mL. CONCLUSIONS GFAP exhibited excellent accuracy in distinguishing patients with normal CSF biomarkers from those with prodromal AD. Our results support the use of this peripheral biomarker for detecting AD in patients with subjective and objective cognitive decline.
Collapse
Affiliation(s)
- Salvatore Mazzeo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giulia Giacomucci
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Arianna Cavaliere
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Moschini
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Juri Balestrini
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Carmen Morinelli
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giulia Galdo
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Filippo Emiliani
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Diletta Piazzesi
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Chiara Crucitti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Daniele Frigerio
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | - Valentina Berti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Sonia Padiglioni
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
- Regional Referral Centre for Relational Criticalities- 50139, Tuscany Region, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy.
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy.
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| |
Collapse
|
30
|
Dias-Carvalho A, Sá SI, Carvalho F, Fernandes E, Costa VM. Inflammation as common link to progressive neurological diseases. Arch Toxicol 2024; 98:95-119. [PMID: 37964100 PMCID: PMC10761431 DOI: 10.1007/s00204-023-03628-8] [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: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Life expectancy has increased immensely over the past decades, bringing new challenges to the health systems as advanced age increases the predisposition for many diseases. One of those is the burden of neurologic disorders. While many hypotheses have been placed to explain aging mechanisms, it has been widely accepted that the increasing pro-inflammatory status with advanced age or "inflammaging" is a main determinant of biological aging. Furthermore, inflammaging is at the cornerstone of many age-related diseases and its involvement in neurologic disorders is an exciting hypothesis. Indeed, aging and neurologic disorders development in the elderly seem to share some basic pathways that fundamentally converge on inflammation. Peripheral inflammation significantly influences brain function and contributes to the development of neurological disorders, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Understanding the role of inflammation in the pathogenesis of progressive neurological diseases is of crucial importance for developing effective treatments and interventions that can slow down or prevent disease progression, therefore, decreasing its social and economic burden.
Collapse
Affiliation(s)
- Ana Dias-Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Susana Isabel Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO- Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
31
|
Trieu C, van Harten AC, Leeuwis AE, Exalto LG, Hooghiemstra AM, Verberk IMW, Allaart CP, Brunner-La Rocca HP, Kappelle LJ, van Oostenbrugge RJ, Biessels GJ, Teunissen CE, van der Flier WM. Alzheimer's Disease and Cognitive Decline in Patients with Cardiovascular Diseases Along the Heart-Brain Axis. J Alzheimers Dis 2024; 98:987-1000. [PMID: 38489178 DOI: 10.3233/jad-231096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background We hypothesize that Alzheimer's disease (AD)-related pathology may accelerate cognitive decline in patients with cardiovascular diseases. Objective To investigate the association between blood-based biomarkers of AD, astrocyte activation, and neurodegeneration and cognitive decline. Methods From the multi-center Heart-Brain study, we included 412 patients with heart failure, carotid occlusive disease or vascular cognitive impairment (age:68.6±9.0) and 128 reference participants (65.7±7.5). Baseline amyloid-β42/40 (Aβ42/40), phosphorylated-tau181 (pTau181), glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) were determined using SiMoA (Quanterix). Memory, attention, language, and executive functioning were evaluated (follow-up:2.1±0.3 years). We applied linear mixed models with terms for biomarker, time and biomarker*time interactions, adjusted for age, sex, education, and site, to assess associations between biomarkers and cognitive decline. Results Among patients, Aβ42/40 was not associated with cognitive performance at baseline. However, lower Aβ42/40 was associated with steeper decline in global cognition (β±SE:0.04±0.02). Higher pTau181 was associated with worse baseline performance on global cognition (-0.14±0.04) and memory (-0.31±0.09) and with steeper decline in global cognition (-0.07±0.02), memory (-0.09±0.04), attention (-0.05±0.02), and language (-0.10±0.03). Higher GFAP was associated with worse baseline performance on global cognition (-0.22±0.05), memory (-0.43±0.10), attention (-0.14±0.06), language (-0.15±0.05), and executive functioning (-0.15±0.05) and steeper decline in global cognition (-0.05±0.01). Higher NfL was associated with worse baseline performance on global cognition (-0.16±0.04), memory (-0.28±0.09), attention (-0.20±0.06), and executive functioning (-0.10±0.04), but was not associated with performance over time. In reference participants, no associations were found. Conclusions Our findings suggest that blood-based biomarkers of AD-related pathology predict cognitive decline in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Calvin Trieu
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam Neuroscience, Program Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Argonde C van Harten
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Anna E Leeuwis
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Lieza G Exalto
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
- Brain Research Center, Zwolle, The Netherlands
- Julius Clinical, Zeist, The Netherlands
| | - Astrid M Hooghiemstra
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam Neuroscience, Program Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Cor P Allaart
- Department of Cardiology, Institute for Cardiovascular Research, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | | | - L Jaap Kappelle
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Geert-Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Charlotte E Teunissen
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam Neuroscience, Program Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center (Amsterdam UMC), Amsterdam, The Netherlands
- Amsterdam Neuroscience, Program Neurodegeneration, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Rivera J, Sharma B, Torres MM, Kumar S. Factors affecting the GABAergic synapse function in Alzheimer's disease: Focus on microRNAs. Ageing Res Rev 2023; 92:102123. [PMID: 37967653 DOI: 10.1016/j.arr.2023.102123] [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: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurological disease characterized by the loss of cognitive function, confusion, and memory deficit. Accumulation of abnormal proteins, amyloid beta (Aß), and phosphorylated Tau (p-tau) forms plaques and tangles that deteriorate synapse function, resulting in neurodegeneration and cognitive decline in AD. The human brain is composed of different types of neurons and/or synapses that are functionally defective in AD. The GABAergic synapse, the most abundant inhibitory neuron in the human brain was found to be dysfunctional in AD and contributes to disrupting neurological function. This study explored the types of GABA receptors associated with neurological dysfunction and various biological and environmental factors that cause GABAergic neuron dysfunction in AD, such as Aβ, p-tau, aging, sex, astrocytes, microglia, APOE, mental disorder, diet, physical activity, and sleep. Furthermore, we explored the role of microRNAs (miRNAs) in the regulation of GABAergic synapse function in neurological disorders and AD states. We also discuss the molecular mechanisms underlying GABAergic synapse dysfunction with a focus on miR-27b, miR-30a, miR-190a/b, miR-33, miR-51, miR-129-5p, miR-376-3p, miR-376c, miR-30b and miR-502-3p. The purpose of our article is to highlight the recent research on miRNAs affecting the regulation of GABAergic synapse function and factors that contribute to the progression of AD.
Collapse
Affiliation(s)
- Jazmin Rivera
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Bhupender Sharma
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Melissa M Torres
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Subodh Kumar
- Center of Emphasis in Neuroscience, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA; L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA.
| |
Collapse
|
33
|
Yang Z, Sreenivasan K, Toledano Strom EN, Osse AML, Pasia LG, Cosme CG, Mugosa MRN, Chevalier EL, Ritter A, Miller JB, Cordes D, Cummings JL, Kinney JW. Clinical and biological relevance of glial fibrillary acidic protein in Alzheimer's disease. Alzheimers Res Ther 2023; 15:190. [PMID: 37924152 PMCID: PMC10623866 DOI: 10.1186/s13195-023-01340-4] [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: 05/06/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023]
Abstract
INTRODUCTION There is a tremendous need for identifying reliable blood-based biomarkers for Alzheimer's disease (AD) that are tied to the biological ATN (amyloid, tau and neurodegeneration) framework as well as clinical assessment and progression. METHODS One hundred forty-four elderly participants underwent 18F-AV45 positron emission tomography (PET) scan, structural magnetic resonance imaging (MRI) scan, and blood sample collection. The composite standardized uptake value ratio (SUVR) was derived from 18F-AV45 PET to assess brain amyloid burden, and the hippocampal volume was determined from structural MRI scans. Plasma glial fibrillary acidic protein (GFAP), phosphorylated tau-181 (ptau-181), and neurofilament light (NfL) measured by single molecular array (SIMOA) technology were assessed with respect to ATN framework, genetic risk factor, age, clinical assessment, and future functional decline among the participants. RESULTS Among the three plasma markers, GFAP best discriminated participants stratified by clinical diagnosis and brain amyloid status. Age was strongly associated with NfL, followed by GFAP and ptau-181 at much weaker extent. Brain amyloid was strongly associated with plasma GFAP and ptau-181 and to a lesser extent with plasma NfL. Moderate association was observed between plasma markers. Hippocampal volume was weakly associated with all three markers. Elevated GFAP and ptau-181 were associated with worse cognition, and plasma GFAP was the most predictive of future functional decline. Combining GFAP and ptau-181 together was the best model to predict brain amyloid status across all participants (AUC = 0.86) or within cognitively impaired participants (AUC = 0.93); adding NfL as an additional predictor only had a marginal improvement. CONCLUSION Our findings indicate that GFAP is of potential clinical utility in screening amyloid pathology and predicting future cognitive decline. GFAP, NfL, and ptau-181 were moderately associated with each other, with discrepant relevance to age, sex, and AD genetic risk, suggesting their relevant but differential roles for AD assessment. The combination of GFAP with ptau-181 provides an accurate model to predict brain amyloid status, with the superior performance of GFAP over ptau-181 when the prediction is limited to cognitively impaired participants.
Collapse
Affiliation(s)
- Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA.
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | - Karthik Sreenivasan
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | | | | | | | - Celica Glenn Cosme
- Kirk Kerkorian School of Medicine, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Maya Rae N Mugosa
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Emma Léa Chevalier
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Aaron Ritter
- Hoag's Pickup Family Neurosciences Institute, Newport Beach, CA, USA
| | - Justin B Miller
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, 80309, USA
| | - Jeffrey L Cummings
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Jefferson W Kinney
- Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, University of Nevada Las Vegas, Las Vegas, NV, USA
| |
Collapse
|
34
|
Verkhratsky A, Butt A, Li B, Illes P, Zorec R, Semyanov A, Tang Y, Sofroniew MV. Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct Target Ther 2023; 8:396. [PMID: 37828019 PMCID: PMC10570367 DOI: 10.1038/s41392-023-01628-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/14/2023] Open
Abstract
Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS). Astroglia contribute to the pathophysiology of all neurological and neuropsychiatric disorders in ways that can be either beneficial or detrimental to disorder outcome. Pathophysiological changes in astroglia can be primary or secondary and can result in gain or loss of functions. Astroglia respond to external, non-cell autonomous signals associated with any form of CNS pathology by undergoing complex and variable changes in their structure, molecular expression, and function. In addition, internally driven, cell autonomous changes of astroglial innate properties can lead to CNS pathologies. Astroglial pathophysiology is complex, with different pathophysiological cell states and cell phenotypes that are context-specific and vary with disorder, disorder-stage, comorbidities, age, and sex. Here, we classify astroglial pathophysiology into (i) reactive astrogliosis, (ii) astroglial atrophy with loss of function, (iii) astroglial degeneration and death, and (iv) astrocytopathies characterised by aberrant forms that drive disease. We review astroglial pathophysiology across the spectrum of human CNS diseases and disorders, including neurotrauma, stroke, neuroinfection, autoimmune attack and epilepsy, as well as neurodevelopmental, neurodegenerative, metabolic and neuropsychiatric disorders. Characterising cellular and molecular mechanisms of astroglial pathophysiology represents a new frontier to identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| | - Arthur Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany
| | - Robert Zorec
- Celica Biomedical, Lab Cell Engineering, Technology Park, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alexey Semyanov
- Department of Physiology, Jiaxing University College of Medicine, 314033, Jiaxing, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Lia A, Di Spiezio A, Vitalini L, Tore M, Puja G, Losi G. Ion Channels and Ionotropic Receptors in Astrocytes: Physiological Functions and Alterations in Alzheimer's Disease and Glioblastoma. Life (Basel) 2023; 13:2038. [PMID: 37895420 PMCID: PMC10608464 DOI: 10.3390/life13102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The human brain is composed of nearly one hundred billion neurons and an equal number of glial cells, including macroglia, i.e., astrocytes and oligodendrocytes, and microglia, the resident immune cells of the brain. In the last few decades, compelling evidence has revealed that glial cells are far more active and complex than previously thought. In particular, astrocytes, the most abundant glial cell population, not only take part in brain development, metabolism, and defense against pathogens and insults, but they also affect sensory, motor, and cognitive functions by constantly modulating synaptic activity. Not surprisingly, astrocytes are actively involved in neurodegenerative diseases (NDs) and other neurological disorders like brain tumors, in which they rapidly become reactive and mediate neuroinflammation. Reactive astrocytes acquire or lose specific functions that differently modulate disease progression and symptoms, including cognitive impairments. Astrocytes express several types of ion channels, including K+, Na+, and Ca2+ channels, transient receptor potential channels (TRP), aquaporins, mechanoreceptors, and anion channels, whose properties and functions are only partially understood, particularly in small processes that contact synapses. In addition, astrocytes express ionotropic receptors for several neurotransmitters. Here, we provide an extensive and up-to-date review of the roles of ion channels and ionotropic receptors in astrocyte physiology and pathology. As examples of two different brain pathologies, we focus on Alzheimer's disease (AD), one of the most diffuse neurodegenerative disorders, and glioblastoma (GBM), the most common brain tumor. Understanding how ion channels and ionotropic receptors in astrocytes participate in NDs and tumors is necessary for developing new therapeutic tools for these increasingly common neurological conditions.
Collapse
Affiliation(s)
- Annamaria Lia
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
| | - Alessandro Di Spiezio
- Department Biomedical Science, University of Padova, 35131 Padova, Italy; (A.L.); (A.D.S.)
- Neuroscience Institute (CNR-IN), Padova Section, 35131 Padova, Italy
| | - Lorenzo Vitalini
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Manuela Tore
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulia Puja
- Department Life Science, University of Modena and Reggio Emilia, 41125 Modena, Italy; (L.V.); (G.P.)
| | - Gabriele Losi
- Institute of Nanoscience (CNR-NANO), Modena Section, 41125 Modena, Italy;
- Department Biomedical Science, Metabolic and Neuroscience, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
36
|
Xue S, Zhou X, Yang ZH, Si XK, Sun X. Stroke-induced damage on the blood-brain barrier. Front Neurol 2023; 14:1248970. [PMID: 37840921 PMCID: PMC10569696 DOI: 10.3389/fneur.2023.1248970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 10/17/2023] Open
Abstract
The blood-brain barrier (BBB) is a functional phenotype exhibited by the neurovascular unit (NVU). It is maintained and regulated by the interaction between cellular and non-cellular matrix components of the NVU. The BBB plays a vital role in maintaining the dynamic stability of the intracerebral microenvironment as a barrier layer at the critical interface between the blood and neural tissues. The large contact area (approximately 20 m2/1.3 kg brain) and short diffusion distance between neurons and capillaries allow endothelial cells to dominate the regulatory role. The NVU is a structural component of the BBB. Individual cells and components of the NVU work together to maintain BBB stability. One of the hallmarks of acute ischemic stroke is the disruption of the BBB, including impaired function of the tight junction and other molecules, as well as increased BBB permeability, leading to brain edema and a range of clinical symptoms. This review summarizes the cellular composition of the BBB and describes the protein composition of the barrier functional junction complex and the mechanisms regulating acute ischemic stroke-induced BBB disruption.
Collapse
Affiliation(s)
| | | | | | | | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Cisternas P, Gherardelli C, Gutierrez J, Salazar P, Mendez-Orellana C, Wong GW, Inestrosa NC. Adiponectin and resistin modulate the progression of Alzheimer´s disease in a metabolic syndrome model. Front Endocrinol (Lausanne) 2023; 14:1237796. [PMID: 37732123 PMCID: PMC10507329 DOI: 10.3389/fendo.2023.1237796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
Metabolic syndrome (MetS), a cluster of metabolic conditions that include obesity, hyperlipidemia, and insulin resistance, increases the risk of several aging-related brain diseases, including Alzheimer's disease (AD). However, the underlying mechanism explaining the link between MetS and brain function is poorly understood. Among the possible mediators are several adipose-derived secreted molecules called adipokines, including adiponectin (ApN) and resistin, which have been shown to regulate brain function by modulating several metabolic processes. To investigate the impact of adipokines on MetS, we employed a diet-induced model to induce the various complications associated with MetS. For this purpose, we administered a high-fat diet (HFD) to both WT and APP/PSN1 mice at a pre-symptomatic disease stage. Our data showed that MetS causes a fast decline in cognitive performance and stimulates Aβ42 production in the brain. Interestingly, ApN treatment restored glucose metabolism and improved cognitive functions by 50% while decreasing the Aβ42/40 ratio by approximately 65%. In contrast, resistin exacerbated Aβ pathology, increased oxidative stress, and strongly reduced glucose metabolism. Together, our data demonstrate that ApN and resistin alterations could further contribute to AD pathology.
Collapse
Affiliation(s)
- Pedro Cisternas
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua, Chile
| | - Camila Gherardelli
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Joel Gutierrez
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paulina Salazar
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Mendez-Orellana
- Carrera de Fonoaudiología, Departamento Ciencias de la Salud, facultad Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - G. William Wong
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Center for Metabolism and Obesity Research, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nibaldo C. Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
38
|
Swanson MJ, Lewis KN, Carpenter R, Whetzel A, Bae NS. The human RAP1 and GFAPɛ proteins increase γ-secretase activity in a yeast model system. G3 (BETHESDA, MD.) 2023; 13:jkad057. [PMID: 36929840 PMCID: PMC10411568 DOI: 10.1093/g3journal/jkad057] [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: 12/09/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023]
Abstract
Alzheimer's disease (AD) is an age-related disorder that results in progressive cognitive impairment and memory loss. Deposition of amyloid β (Aβ) peptides in senile plaques is a hallmark of AD. γ-secretase produces Aβ peptides, mostly as the soluble Aβ40 with fewer insoluble Aβ42 peptides. Rare, early-onset AD (EOAD) occurs in individuals under 60 years of age. Most EOAD cases are due to unknown genetic causes, but a subset is due to mutations in the genes encoding the amyloid precursor protein that is processed into Aβ peptides or the presenilins (PS1 and PS2) that process APP. PS1 interacts with the epsilon isoform of glial fibrillary acidic protein (GFAPɛ), a protein found in the subventricular zone of the brain. We have found that GFAPɛ interacts with the telomere protection factor RAP1 (TERF2IP). RAP1 can also interact with PS1 alone or with GFAPɛ in vitro. Our data show that the nuclear protein RAP1 has an extratelomeric role in the cytoplasm through its interactions with GFAPɛ and PS1. GFAPɛ coprecipitated with RAP1 from human cell extracts. RAP1, GFAPɛ, and PS1 all colocalized in human SH-SY5Y cells. Using a genetic model of the γ-secretase complex in Saccharomyces cerevisiae, RAP1 increased γ-secretase activity, and this was potentiated by GFAPɛ. Our studies are the first to connect RAP1 with an age-related disorder.
Collapse
Affiliation(s)
- Mark J Swanson
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ 85308, USA
| | - Kelsey N Lewis
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ 85308, USA
| | - Robert Carpenter
- Department of Biomedical Sciences, College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Alexis Whetzel
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ 85308, USA
| | - Nancy S Bae
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
39
|
Pacheco-Sánchez B, Tovar R, Ben Rabaa M, Sánchez-Salido L, Vargas A, Suárez J, Rodríguez de Fonseca F, Rivera P. Sex-Dependent Altered Expression of Cannabinoid Signaling in Hippocampal Astrocytes of the Triple Transgenic Mouse Model of Alzheimer's Disease: Implications for Controlling Astroglial Activity. Int J Mol Sci 2023; 24:12598. [PMID: 37628778 PMCID: PMC10454447 DOI: 10.3390/ijms241612598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease. In AD-associated neuroinflammation, astrocytes play a key role, finding glial activation both in patients and in animal models. The endocannabinoid system (ECS) is a neurolipid signaling system with anti-inflammatory and neuroprotective properties implicated in AD. Astrocytes respond to external cannabinoid signals and also have their own cannabinoid signaling. Our main objective is to describe the cannabinoid signaling machinery present in hippocampal astrocytes from 3×Tg-AD mice to determine if they are actively involved in the neurodegenerative process. Primary cultures of astrocytes from the hippocampus of 3×Tg-AD and non-Tg offspring were carried out. We analyzed the gene expression of astrogliosis markers, the main components of the ECS and Ca2+ signaling. 3×Tg-AD hippocampal astrocytes show low inflammatory activity (Il1b, Il6, and Gls) and Ca2+ flow (P2rx5 and Mcu), associated with low cannabinoid signaling (Cnr1 and Cnr2). These results were more evident in females. Our study corroborates glial involvement in AD pathology, in which cannabinoid signaling plays an important role. 3×Tg-AD mice born with hippocampal astrocytes with differential gene expression of the ECS associated with an innate attenuation of their activity. In addition, we show that there are sex differences from birth in this AD animal, which should be considered when investigating the pathogenesis of the disease.
Collapse
Affiliation(s)
- Beatriz Pacheco-Sánchez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| | - Rubén Tovar
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| | - Meriem Ben Rabaa
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
- Molecular Biotechnology, FH Campus Wien, University for Applied Sciences, Favoritenstraße 222, 1100 Vienna, Austria
| | - Lourdes Sánchez-Salido
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| | - Antonio Vargas
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| | - Patricia Rivera
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Regional de Málaga, 29010 Málaga, Spain; (B.P.-S.); (R.T.); (M.B.R.); (L.S.-S.); (A.V.); (J.S.)
| |
Collapse
|
40
|
Pereira MJ, Ayana R, Holt MG, Arckens L. Chemogenetic manipulation of astrocyte activity at the synapse- a gateway to manage brain disease. Front Cell Dev Biol 2023; 11:1193130. [PMID: 37534103 PMCID: PMC10393042 DOI: 10.3389/fcell.2023.1193130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/14/2023] [Indexed: 08/04/2023] Open
Abstract
Astrocytes are the major glial cell type in the central nervous system (CNS). Initially regarded as supportive cells, it is now recognized that this highly heterogeneous cell population is an indispensable modulator of brain development and function. Astrocytes secrete neuroactive molecules that regulate synapse formation and maturation. They also express hundreds of G protein-coupled receptors (GPCRs) that, once activated by neurotransmitters, trigger intracellular signalling pathways that can trigger the release of gliotransmitters which, in turn, modulate synaptic transmission and neuroplasticity. Considering this, it is not surprising that astrocytic dysfunction, leading to synaptic impairment, is consistently described as a factor in brain diseases, whether they emerge early or late in life due to genetic or environmental factors. Here, we provide an overview of the literature showing that activation of genetically engineered GPCRs, known as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), to specifically modulate astrocyte activity partially mimics endogenous signalling pathways in astrocytes and improves neuronal function and behavior in normal animals and disease models. Therefore, we propose that expressing these genetically engineered GPCRs in astrocytes could be a promising strategy to explore (new) signalling pathways which can be used to manage brain disorders. The precise molecular, functional and behavioral effects of this type of manipulation, however, differ depending on the DREADD receptor used, targeted brain region and timing of the intervention, between healthy and disease conditions. This is likely a reflection of regional and disease/disease progression-associated astrocyte heterogeneity. Therefore, a thorough investigation of the effects of such astrocyte manipulation(s) must be conducted considering the specific cellular and molecular environment characteristic of each disease and disease stage before this has therapeutic applicability.
Collapse
Affiliation(s)
- Maria João Pereira
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Rajagopal Ayana
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| | - Matthew G. Holt
- Instituto de Investigação e Inovação em Saúde (i3S), Laboratory of Synapse Biology, Universidade do Porto, Porto, Portugal
| | - Lutgarde Arckens
- Department of Biology, Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, Leuven, Belgium
- KU Leuven Brain Institute, Leuven, Belgium
| |
Collapse
|
41
|
Mantik KEK, Kim S, Gu B, Moon S, Kwak HB, Park DH, Kang JH. Repositioning of Anti-Diabetic Drugs against Dementia: Insight from Molecular Perspectives to Clinical Trials. Int J Mol Sci 2023; 24:11450. [PMID: 37511207 PMCID: PMC10380685 DOI: 10.3390/ijms241411450] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin resistance as a hallmark of type 2 DM (T2DM) plays a role in dementia by promoting pathological lesions or enhancing the vulnerability of the brain. Numerous studies related to insulin/insulin-like growth factor 1 (IGF-1) signaling are linked with various types of dementia. Brain insulin resistance in dementia is linked to disturbances in Aβ production and clearance, Tau hyperphosphorylation, microglial activation causing increased neuroinflammation, and the breakdown of tight junctions in the blood-brain barrier (BBB). These mechanisms have been studied primarily in Alzheimer's disease (AD), but research on other forms of dementia like vascular dementia (VaD), Lewy body dementia (LBD), and frontotemporal dementia (FTD) has also explored overlapping mechanisms. Researchers are currently trying to repurpose anti-diabetic drugs to treat dementia, which are dominated by insulin sensitizers and insulin substrates. Although it seems promising and feasible, none of the trials have succeeded in ameliorating cognitive decline in late-onset dementia. We highlight the possibility of repositioning anti-diabetic drugs as a strategy for dementia therapy by reflecting on current and previous clinical trials. We also describe the molecular perspectives of various types of dementia through the insulin/IGF-1 signaling pathway.
Collapse
Affiliation(s)
- Keren Esther Kristina Mantik
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Sujin Kim
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Bonsang Gu
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, College of Arts and Sports, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
- Department of Kinesiology, College of Arts and Sports, Inha University, Incheon 22212, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology, Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
42
|
Andrade MK, Souza LC, Azevedo EM, Bail EL, Zanata SM, Andreatini R, Vital MABF. Melatonin reduces β-amyloid accumulation and improves short-term memory in streptozotocin-induced sporadic Alzheimer's disease model. IBRO Neurosci Rep 2023; 14:264-272. [PMID: 36926592 PMCID: PMC10011440 DOI: 10.1016/j.ibneur.2023.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Melatonin is a hormone secreted by the pineal gland, it can be associated with circadian rhythms, aging and neuroprotection. Melatonin levels are decreased in sporadic Alzheimer's disease (sAD) patients, which suggests a relationship between the melatonergic system and sAD. Melatonin may reduce inflammation, oxidative stress, TAU protein hyperphosphorylation, and the formation of β-amyloid (Aβ) aggregates. Therefore, the objective of this work was to investigate the impact of treatment with 10 mg/kg of melatonin (i.p) in the animal model of sAD induced by the intracerebroventricular (ICV) infusion of 3 mg/kg of streptozotocin (STZ). ICV-STZ causes changes in the brain of rats similar to those found in patients with sAD. These changes include; progressive memory decline, the formation of neurofibrillary tangles, senile plaques, disturbances in glucose metabolism, insulin resistance and even reactive astrogliosis characterized by the upregulation of glucose levels and glial fibrillary acidic protein (GFAP). The results show that ICV-STZ caused short-term spatial memory impairment in rats after 30 days of STZ infusion without locomotor impairment which was evaluated on day 27 post-injury. Furthermore, we observed that a prolonged 30-day treatment with melatonin can improve the cognitive impairment of animals in the Y-maze test, but not in the object location test. Finally, we demonstrated that animals receiving ICV-STZ have high levels of Aβ and GFAP in the hippocampus and that treatment with melatonin reduces Aβ levels but does not reduce GFAP levels, concluding that melatonin may be useful to control the progression of amyloid pathology in the brain.
Collapse
Key Words
- AD, Alzheimer Disease
- APP, Amyloid precursor protein
- Alzheimer's disease
- Aβ, β-amyloid
- GFAP
- GFAP, Glial fibrillary acidic protein
- ICV-STZ, Intracerebroventricular injection of streptozotocin
- MEL, Melatonin
- MT1, Melatonin Receptor 1
- MT2, Melatonin Receptor 2
- Melatonin
- OLT, Object location test
- STZ, Streptozotocin
- Short-term memory
- Streptozotocin
- TNF-α, Tumor Necrosis factor alpha
- Y maze
- sAD, Sporadic Alzheimer disease
- β-amyloid
Collapse
Affiliation(s)
- Marcos K Andrade
- Department of Pharmacology, Federal University of Paraná, PR, Brazil
| | - Leonardo C Souza
- Department of Pharmacology, Federal University of Paraná, PR, Brazil
| | - Evellyn M Azevedo
- Department of Physiology, Federal University of Paraná, PR, Brazil.,Department of Basic Pathology, Federal University of Paraná, PR, Brazil
| | - Ellen L Bail
- Department of Physiology, Federal University of Paraná, PR, Brazil.,Department of Basic Pathology, Federal University of Paraná, PR, Brazil
| | - Silvio M Zanata
- Department of Basic Pathology, Federal University of Paraná, PR, Brazil
| | | | - Maria A B F Vital
- Department of Pharmacology, Federal University of Paraná, PR, Brazil
| |
Collapse
|
43
|
Furukawa M, Tada H, Raju R, Wang J, Yokoi H, Ikuyo Y, Yamada M, Shikama Y, Matsushita K. Long-Term Capsaicin Administration Ameliorates the Dysfunction and Astrogliosis of the Brain in Aged Mice with Missing Maxillary Molars. Nutrients 2023; 15:nu15112471. [PMID: 37299434 DOI: 10.3390/nu15112471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Tooth loss and decreased masticatory function reportedly affect cognitive function; tooth loss allegedly induces astrogliosis and aging of astrocytes in the hippocampus and hypothalamus, which is a response specific to the central nervous system owing to homeostasis in different brain regions. Capsaicin, a component of red peppers, has positive effects on brain disorders in mice. Decreased expression of transient receptor potential vanilloid 1, a receptor of capsaicin, is associated with the development of dementia. In this study, we investigated the effect of capsaicin administration in aged mice (C57BL/6N mice) with reduced masticatory function owing to the extraction of maxillary molars to investigate preventive/therapeutic methods for cognitive decline attributed to age-related masticatory function loss. The results demonstrated that mice with impaired masticatory function showed decreased motor and cognitive function at the behavioral level. At the genetic level, neuroinflammation, microglial activity, and astrogliosis, such as increased glial fibrillary acidic protein levels, were observed in the mouse brain. The mice with extracted molars fed on a diet containing capsaicin for 3 months demonstrated improved behavioral levels and astrogliosis, which suggest that capsaicin is useful in maintaining brain function in cases of poor oral function and prosthetic difficulties.
Collapse
Affiliation(s)
- Masae Furukawa
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Hirobumi Tada
- Department of Nutrition, Faculty of Wellness, Shigakkan University, Obu 474-8651, Japan
- Department of Integrative Physiology, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Resmi Raju
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Jingshu Wang
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Haruna Yokoi
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Yoriko Ikuyo
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Mitsuyoshi Yamada
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
- Department of Operative Dentistry, School of Dentistry, Aichi Gakuin University, Nagoya 464-8651, Japan
| | - Yosuke Shikama
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, Geroscience Research Center, National Center for Geriatrics and Gerontology, Obu 474-8511, Japan
| |
Collapse
|
44
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
45
|
Saleh BM, Pourmostafa A, Patrawalla NY, Kishore V. Xeno-Free Biomimetic ECM Model for Investigation of Matrix Composition and Stiffness on Astrocyte Cell Response. J Funct Biomater 2023; 14:jfb14050256. [PMID: 37233366 DOI: 10.3390/jfb14050256] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/27/2023] Open
Abstract
Astrocytes, highly specialized glial cells, play a critical role in neuronal function. Variations in brain extracellular matrix (ECM) during development and disease can significantly alter astrocyte cell function. Age-related changes in ECM properties have been linked to neurodegenerative diseases such as Alzheimer's disease. The goal of this study was to develop hydrogel-based biomimetic ECM models with varying stiffness and evaluate the effects of ECM composition and stiffness on astrocyte cell response. Xeno-free ECM models were synthesized by combining varying ratios of human collagen and thiolated hyaluronic acid (HA) crosslinked with polyethylene glycol diacrylate. Results showed that modulating ECM composition yielded hydrogels with varying stiffnesses that match the stiffness of the native brain ECM. Collagen-rich hydrogels swell more and exhibit greater stability. Higher metabolic activity and greater cell spreading was observed in hydrogels with lower HA. Soft hydrogels trigger astrocyte activation indicated by greater cell spreading, high GFAP expression and low ALDH1L1 expression. This work presents a baseline ECM model to investigate the synergistic effects of ECM composition and stiffness on astrocytes, which could be further developed to identify key ECM biomarkers and formulate new therapies to alleviate the impact of ECM changes on the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Bayan M Saleh
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL 32901, USA
| | - Ayda Pourmostafa
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL 32901, USA
| | - Nashaita Y Patrawalla
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL 32901, USA
| | - Vipuil Kishore
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL 32901, USA
| |
Collapse
|
46
|
Mao S, Teng X, Li Z, Zu J, Zhang T, Xu C, Cui G. Association of serum neurofilament light chain and glial fibrillary acidic protein levels with cognitive decline in Parkinson's disease. Brain Res 2023; 1805:148271. [PMID: 36754139 DOI: 10.1016/j.brainres.2023.148271] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVES To investigate whether serum neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) levels are associated with motor and cognitive function in Parkinson's disease (PD). METHODS This cross-sectional study recruited 140 participants, including 103 PD patients and 37 healthy controls (HC). Serum NfL and GFAP levels were measured using the ultrasensitive single-molecule array (Simoa) technique. Motor and cognitive function were evaluated using the Movement Disorder Society Unified Parkinson's Disease Rating Scale Part III (MDS-UPDRS III) and Beijing version of the Montreal Cognitive Assessment (MoCA). Spearman's correlation analyses were used to determine the correlation between serum NfL and GFAP levels and clinical features in PD patients. Binary logistic regression analysis was used to assess the association between serum biomarkers and cognitive impairment in PD patients. RESULTS We observed significantly higher serum NfL and GFAP levels in PD patients than in HC (p < 0.001). Serum NfL and GFAP levels were negatively correlated with MoCA scores (NfL: r = - 0.472, p < 0.001; r = 0.395, p < 0.001) and multiple cognitive domains and showed no correlation with motor symptom severity after adjusting for age and sex. Binary logistic regression analysis showed that the serum NfL and GFAP levels were independent contributors to PD with dementia (p < 0.05). CONCLUSIONS Both serum NfL and GFAP levels correlated with cognitive impairment, but not motor symptoms, in PD patients. Serum NfL and GFAP levels can serve as biomarkers for PD patients at risk of cognitive decline.
Collapse
Affiliation(s)
- Shuai Mao
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Xing Teng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Zhen Li
- Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China
| | - Tao Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China
| | - Chuanying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China.
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu Province 221000, China; Department of Neurology, The First Clinical College, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu Province 221000, China.
| |
Collapse
|
47
|
Tang Y, Yan Y, Mao J, Ni J, Qing H. The hippocampus associated GABAergic neural network impairment in early-stage of Alzheimer's disease. Ageing Res Rev 2023; 86:101865. [PMID: 36716975 DOI: 10.1016/j.arr.2023.101865] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is the commonest neurodegenerative disease with slow progression. Pieces of evidence suggest that the GABAergic system is impaired in the early stage of AD, leading to hippocampal neuron over-activity and further leading to memory and cognitive impairment in patients with AD. However, the precise impairment mechanism of the GABAergic system on the pathogenesis of AD is still unclear. The impairment of neural networks associated with the GABAergic system is tightly associated with AD. Therefore, we describe the roles played by hippocampus-related GABAergic circuits and their impairments in AD neuropathology. In addition, we give our understand on the process from GABAergic circuit impairment to cognitive and memory impairment, since recent studies on astrocyte in AD plays an important role behind cognition dysfunction caused by GABAergic circuit impairment, which helps better understand the GABAergic system and could open up innovative AD therapy.
Collapse
Affiliation(s)
- Yuanhong Tang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jian Mao
- Zhengzhou Tobacco Institute of China National Tobacco Company, Zhengzhou 450001, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Department of Biology, Shenzhen MSU-BIT University, Shenzhen 518172, China.
| |
Collapse
|
48
|
Huang Y, Zhang W, Guo X, Zhang Y, Wu J, Zu H. Cellular cholesterol loss by DHCR24 knockdown leads to Aβ production by changing APP intracellular localization. J Lipid Res 2023; 64:100367. [PMID: 37011864 PMCID: PMC10173783 DOI: 10.1016/j.jlr.2023.100367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
For the past 20 years, the majority of cell culture studies reported that increasing cholesterol level increases amyloid-β (Aβ) production. Conversely, other studies and genetic evidences support that cellular cholesterol loss leads to Aβ generation. As a highly controversial issue in Alzheimer's disease (AD) pathogenesis,the apparent contradiction prompted us to again explore the role of cellular cholesterol in Aβ production. Here, we adopted new neuronal and astrocytic cell models induced by 3β-hydroxysterol-Δ24 reductase (DHCR24), which obviously differ from the widely used cell models with overexpressing amyloid precursor protein (APP) in the majority of previous studies. In neuronal and astrocytic cell model, we found that deficiency of cellular cholesterol by DHCR24 knock-down obviously increased intracellular and extracellular Aβ generation. Importantly, in cell models with overexpressing APP, we found that APP overexpression could disrupt cellular cholesterol homeostasis and affect function of cells, coupled with the increase of APP β-cleavage product, 99-residue transmembrane C-terminal domain (C99). Therefore, we suppose the results derived from the APP knock-in models will need to be re-evaluated. One rational explanation for the discrepancy between our outcomes and the previous studies could be attributed to the two different cell models. Mechanistically, we showed that cellular cholesterol loss obviously altered APP intracellular localization by affecting cholesterol-related trafficking protein of APP. Therefore, our outcomes strongly support cellular cholesterol loss by DHCR24 knockdown leads to Aβ production.
Collapse
|
49
|
Purnell BS, Alves M, Boison D. Astrocyte-neuron circuits in epilepsy. Neurobiol Dis 2023; 179:106058. [PMID: 36868484 DOI: 10.1016/j.nbd.2023.106058] [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: 12/19/2022] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
The epilepsies are a diverse spectrum of disease states characterized by spontaneous seizures and associated comorbidities. Neuron-focused perspectives have yielded an array of widely used anti-seizure medications and are able to explain some, but not all, of the imbalance of excitation and inhibition which manifests itself as spontaneous seizures. Furthermore, the rate of pharmacoresistant epilepsy remains high despite the regular approval of novel anti-seizure medications. Gaining a more complete understanding of the processes that turn a healthy brain into an epileptic brain (epileptogenesis) as well as the processes which generate individual seizures (ictogenesis) may necessitate broadening our focus to other cell types. As will be detailed in this review, astrocytes augment neuronal activity at the level of individual neurons in the form of gliotransmission and the tripartite synapse. Under normal conditions, astrocytes are essential to the maintenance of blood-brain barrier integrity and remediation of inflammation and oxidative stress, but in epilepsy these functions are impaired. Epilepsy results in disruptions in the way astrocytes relate to each other by gap junctions which has important implications for ion and water homeostasis. In their activated state, astrocytes contribute to imbalances in neuronal excitability due to their decreased capacity to take up and metabolize glutamate and an increased capacity to metabolize adenosine. Furthermore, due to their increased adenosine metabolism, activated astrocytes may contribute to DNA hypermethylation and other epigenetic changes that underly epileptogenesis. Lastly, we will explore the potential explanatory power of these changes in astrocyte function in detail in the specific context of the comorbid occurrence of epilepsy and Alzheimer's disease and the disruption in sleep-wake regulation associated with both conditions.
Collapse
Affiliation(s)
- Benton S Purnell
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America
| | - Mariana Alves
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America; Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States of America; Brain Health Institute, Rutgers University, Piscataway, NJ, United States of America.
| |
Collapse
|
50
|
Cunha Feio Leal MD, Amaral Junior FLD, Silva Arruda BFD, Kurosawa JAA, Vieira AA, Maia JCC, Scalfoni VVB, Silveira Junior AMD, Feijó MO, Albuquerque FBAD, Marta MHM, Normando MPN, Silva AGOCD, Trindade FCPD, Siqueira Mendes FDCCD, Sosthenes MCK. The Masticatory Activity Interference in Quantitative Estimation of CA1, CA3 and Dentate Gyrus Hippocampal Astrocytes of Aged Murine Models and under Environmental Stimulation. Int J Mol Sci 2023; 24:ijms24076529. [PMID: 37047502 PMCID: PMC10095286 DOI: 10.3390/ijms24076529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023] Open
Abstract
Studies indicating the influence of masticatory dysfunction, due to a soft diet or lack of molars, on impairing spatial memory and learning have led to research about neuronal connections between areas and cell populations possibly affected. In this sense, with scarce detailed data on the subfields of hippocampus in dementia neurodegeneration, there is no information about astrocytic responses in its different layers. Thus, considering this context, the present study evaluated the effects of deprivation and rehabilitation of masticatory activity, aging, and environmental enrichment on the stereological quantification of hippocampal astrocytes from layers CA1, CA3, and DG. For this purpose, we examined mature (6-month-old; 6M), and aged (18-month-old; 18M) mice, subjected to distinct masticatory regimens and environments. Three different regimens of masticatory activity were applied: continuous normal mastication with hard pellets (HD); normal mastication followed by deprived mastication with equal periods of pellets followed by soft powder (HD/SD); or rehabilitated masticatory activity with equal periods of HD, followed by powder, followed by pellets (HD/SD/HD). Under each specific regimen, half of the animals were raised in standard cages (impoverished environment (IE)) and the other half in enriched cages (enriched environment (EE)), mimicking sedentary or active lifestyles. Microscopic stereological, systematic, and random sampling approaches with an optical dissector of GFAP-immunolabeled astrocytes were done, allowing for an astrocyte numerical estimate. Stratum moleculare and hilus, from the dentate gyrus (DG) and Strata Lacunosum-Moleculare, Oriens, and Radiatum, similarly to the dentate gyrus, showed no significant change in any of the investigated variables (age, diet, or environment) in these layers. However, in Stratum radiatum, it was possible to observe significant differences associated with diet regimens and age. Therefore, diet-related differences were found when the HD 18M IE group was compared to the HD/SD/HD 18-month-old group in the same environment (IE) (p = 0.007). In the present study, we present modulatory factors (masticatory function, environmental enrichment, and aging) for the differentiated quantitative laminar response in the hippocampal regions, suggesting other studies to read the plasticity and responsiveness of astrocytes, including the molecular background.
Collapse
Affiliation(s)
- Marília da Cunha Feio Leal
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fabio Leite do Amaral Junior
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Bernardo Freire da Silva Arruda
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | | | - Amanda Almeida Vieira
- Curso de Medicina, Centro Universitário do Estado do Pará, Belém 66613-903, PA, Brazil
| | | | | | - Antonio Morais da Silveira Junior
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Matheus Oliveira Feijó
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fernanda Beatriz Araújo de Albuquerque
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | | | | | - Alana Gabriele Oliveira Cabeça da Silva
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fernanda Catharina Pires da Trindade
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| | - Fabíola de Carvalho Chaves de Siqueira Mendes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
- Curso de Medicina, Centro Universitário do Estado do Pará, Belém 66613-903, PA, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, PA, Brazil
| |
Collapse
|