1
|
Baidya AT, Dante D, Das B, Wang L, Darreh-Shori T, Kumar R. Discovery and characterization of novel pyridone and furan substituted ligands of choline acetyltransferase. Eur J Pharmacol 2025; 998:177638. [PMID: 40252901 DOI: 10.1016/j.ejphar.2025.177638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
The key to the management of two devastating diseases, namely Alzheimer's Disease (AD) and Amyotrophic Lateral Sclerosis (ALS) lies in an early diagnosis, which is difficult due to its multifactorial nature. However, a common hallmark of AD and ALS is degeneration of cholinergic system. Choline acetyltransferase (ChAT) has been proposed as a potential target for development of cholinergic-specific biomarker. However, lack of selective, potent, brain permeable molecular probes of ChAT hinder development of ChAT biomarkers. In this study, we have successfully utilised structure-based virtual screening approach and identified two ChAT inhibitors from a database of 1.4 million compounds. The compounds were then subjected to rigorous in vitro characterization. Compound V6 showed Ki value of 11 μM and IC50 value of 21.73 μM, while V15 showed Ki and IC50 values of 4.5 and 9.42 μM, respectively for ChAT enzyme. V6 and V15 showed good solubility of 0.21 mg/mL and 0.17 mg/mL respectively and cytotoxicity analysis indicated no toxicity. We also performed a 200 ns molecular dynamics simulation, which revealed the intricate interaction dynamics for V6 and V15 with ChAT binding pocket. Moreover, the Tanimoto similarity analysis indicated the novelty and structural diversity of the hits. In conclusion, these validated hits provide a platform to develop potent, selective, blood-brain barrier permeable small molecules as chemical probes of ChAT or as Positron Emission Tomography tracer for early diagnosis and/or in vivo monitoring of the effect of new therapeutic candidates in spectrum of neurodegenerative disorders, in which cholinergic deficit is one of the hallmarks.
Collapse
Affiliation(s)
- Anurag Tk Baidya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India
| | - Davide Dante
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Bhanuranjan Das
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India
| | - Lisha Wang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Taher Darreh-Shori
- Division of Clinical Geriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 141 52, Stockholm, Sweden
| | - Rajnish Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.), Varanasi, 221005, U.P., India.
| |
Collapse
|
2
|
Neto DCF, Diz JSF, Guimarães SJA, Dos Santos EM, Nascimento MDDSB, de Azevedo-Santos APS, França TCC, LaPlante SR, do Nascimento CJ, Lima JA. Guanylhydrazone and semicarbazone derivatives as potential prototypes for the design of cholinesterase inhibitors against Alzheimer's disease: biological evaluation and molecular modeling studies. Chem Biol Interact 2025; 415:111515. [PMID: 40246050 DOI: 10.1016/j.cbi.2025.111515] [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: 09/26/2024] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Despite being present in many drugs, guanylhydrazones and semicarbazones are two functional groups that have been little investigated as potential therapeutic strategies for the treatment of Alzheimer's disease (AD). For this reason, we initiated the synthesis and evaluation of these compounds as potential anticholinesterase agents, aiming to offer new alternatives for drug development against AD. In the severe phase of AD butyrylcholinesterase (BChE) becomes the main enzyme responsible for the hydrolysis of acetylcholine (ACh). Therefore, in this project, we present the results of BChE inhibitory activity, enzyme kinetics, cytotoxicity, and molecular modeling studies for three guanylhydrazone and two semicarbazone derivatives that were previously synthesized and evaluated as acetylcholinesterase (AChE) inhibitors. Among the compounds tested, guanylhydrazones (1, 2, and 3) showed inhibitory activity against BChE, exhibiting a mixed non-competitive inhibition profile. Specifically, compound 2 (phenanthrenequinone) demonstrated superior inhibitory potency with an IC50 of 0.68 μM, compared to compound 1 (acridinone) with an IC50 of 3.87 μM, and compound 3 (benzodioxole) with an IC50 of 101.7 μM. In contrast, semicarbazones (4 and 5) showed no BChE inhibition up to the highest concentration tested (300 μM). Importantly, all five compounds were found to be non-cytotoxic. Our results suggest that these compounds have potential as drug prototypes targeting different phases of AD. Compounds 3, 4, and 5 may be more effective in the early phase, when AChE activity remains high; compound 1 could be useful in the intermediate phase; and compound 2 appears particularly promising for the severe phase, when BChE plays a more dominant role.
Collapse
Affiliation(s)
- Denise Cristian Ferreira Neto
- Medicinal Chemistry Group, Department of Chemistry, Military Institute of Engineering, Praça General Tibúrcio 80, 22290-270, Rio de Janeiro, RJ, Brazil; Department of Chemistry, Federal University of Roraima, Av. Cap. Ene Garcês, 2413, 69310-000, Boa Vista, Roraima, Brazil.
| | - Joyce Sobreiro Francisco Diz
- Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense (LMCBD), Military Institute of Engineering (IME), Praça General Tibúrcio 80, 22290-270, Rio de Janeiro, Brazil; Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Av. das Américas 28705, Área 4, 23020-470, Rio de Janeiro, RJ, Brazil
| | - Sulayne Janayna Araújo Guimarães
- Laboratory for Applied Cancer Immunology, Biological and Health Sciences Center, Federal University of Maranhão, Avenida dos Portugueses, 1966, Bacanga, 65080-805, São Luís, Maranhão, Brazil
| | - Eduardo Mendes Dos Santos
- Federal University of Maranhão, Postgraduate Program in Adult Health (PPGSAD), Avenida dos Portugueses, 1966, Bacanga, 65080-805, São Luís, Maranhão, Brazil
| | | | - Ana Paula Silva de Azevedo-Santos
- Laboratory for Applied Cancer Immunology, Biological and Health Sciences Center, Federal University of Maranhão, Avenida dos Portugueses, 1966, Bacanga, 65080-805, São Luís, Maranhão, Brazil
| | - Tanos Celmar Costa França
- Laboratory of Molecular Modeling Applied to the Chemical and Biological Defense (LMCBD), Military Institute of Engineering (IME), Praça General Tibúrcio 80, 22290-270, Rio de Janeiro, Brazil; Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec, H7V 1B7, Canada; Center for Basic and Applied Research, Faculty of Informatics and Management, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| | - Steven R LaPlante
- Université de Québec, INRS - Centre Armand-Frappier Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Claudia Jorge do Nascimento
- Institute of Biosciences, Federal University of the State of Rio de Janeiro, Av. Pasteur, 296, Urca, 22290-250, Rio de Janeiro, Brazil
| | - Josélia Alencar Lima
- Federal University of Maranhão, Postgraduate Program in Adult Health (PPGSAD), Avenida dos Portugueses, 1966, Bacanga, 65080-805, São Luís, Maranhão, Brazil.
| |
Collapse
|
3
|
Pelegrino ADF, Attarha M, Toussaint PJ, Ouellet L, Grant SJ, Van Vleet T, de Villers-Sidani E. Cholinergic neurotransmission in the anterior cingulate cortex is associated with cognitive performance in healthy older adults: Baseline characteristics of the Improving Neurological Health in Aging via Neuroplasticity-based Computerized Exercise (INHANCE) trial. NEUROIMAGE. REPORTS 2025; 5:100234. [PMID: 40191405 PMCID: PMC11970925 DOI: 10.1016/j.ynirp.2025.100234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Aging is associated with dysfunction in the cholinergic system, including degeneration of basal forebrain cholinergic terminals that innervate the cortex, which directly contributes to age- and disease-related cognitive decline. In this study, we used [18F]fluoroethoxybenzovesamicol ([18F]FEOBV) positron emission tomography (PET) imaging to assess the effect of age on cholinergic terminal integrity in predefined regions of interest and its relationship to cognitive performance in healthy older adults who underwent neuropsychological assessment and FEOBV PET brain imaging. Our results showed age-related reductions in FEOBV binding, particularly in the anterior cingulate cortex-the primary region of interest-as well as in the striatum, posterior cingulate cortex, and primary auditory cortex. Notably, FEOBV binding in the anterior cingulate cortex was positively correlated with cognitive performance on the NIH EXAMINER Executive Composite Score. These findings suggest that [18F] FEOBV PET imaging can be used as a reliable biomarker to assess cholinergic changes in the human brain and indicate that preserving the cholinergic integrity of the basal forebrain may help maintain cognitive function and protect against age-related cognitive decline.
Collapse
Affiliation(s)
- Ana de Figueiredo Pelegrino
- McGill University, Montreal Neurological Institute and Hospital, 3801 University Street, Montréal, Quebec, H3A 2B4, Canada
| | - Mouna Attarha
- Posit Science Corporation, 160 Pine St Suite 200, San Francisco, CA, 94111, United States
| | - Paule-Joanne Toussaint
- McGill University, Montreal Neurological Institute and Hospital, 3801 University Street, Montréal, Quebec, H3A 2B4, Canada
| | - Lydia Ouellet
- McGill University, Montreal Neurological Institute and Hospital, 3801 University Street, Montréal, Quebec, H3A 2B4, Canada
| | - Sarah-Jane Grant
- Posit Science Corporation, 160 Pine St Suite 200, San Francisco, CA, 94111, United States
| | - Thomas Van Vleet
- Posit Science Corporation, 160 Pine St Suite 200, San Francisco, CA, 94111, United States
| | - Etienne de Villers-Sidani
- McGill University, Montreal Neurological Institute and Hospital, 3801 University Street, Montréal, Quebec, H3A 2B4, Canada
| |
Collapse
|
4
|
Pennington KR, Debs L, Chung S, Bava J, Garin CM, Vale FL, Bick SK, Englot DJ, Terry AV, Constantinidis C, Blake DT. Basal forebrain activation improves working memory in senescent monkeys. Brain Stimul 2025; 18:185-194. [PMID: 39924100 DOI: 10.1016/j.brs.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025] Open
Abstract
Brain aging contributes to cognitive decline and risk of dementia. Degeneration of the basal forebrain cholinergic system parallels these changes in aging, Alzheimer's dementia, Parkinson's dementia, and Lewy body dementia, and thus is a common element linked to executive function across the lifespan and in disease states. Here, we tested the potential of one-hour daily intermittent basal forebrain stimulation to improve cognition in senescent Rhesus monkeys, and its mechanisms of action. Stimulation in five animals improved working memory duration in each animal over 8-12 weeks, with peak improvements observed in the first four weeks. In an ensuing three month period without stimulation, improvements were retained. With additional stimulation, performance remained above baseline throughout the 15 months of the study. Studies with a cholinesterase inhibitor in five animals produced inconsistent improvements in behavior. One of five animals improved significantly. Manipulating the stimulation pattern demonstrated selectivity for both stimulation and recovery period duration in two animals. Brain stimulation led to acute increases in cerebrospinal fluid levels of tissue plasminogen activator, which is an activating element for two brain neurotrophins, Nerve Growth Factor (NGF) and Brain-Derived Growth Factor (BDNF), in four animals. Stimulation also led to improved glucose utilization in stimulated hemispheres relative to contralateral in three animals. Glucose utilization also consistently declines with aging and some dementias. Together, these findings suggest that intermittent stimulation of the nucleus basalis of Meynert improves executive function and reverses some aspects of brain aging.
Collapse
Affiliation(s)
- Kendyl R Pennington
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Luca Debs
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sophia Chung
- Neuroscience Program, Vanderbilt University, Nashville, TN, 37235, USA
| | - Janki Bava
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Clément M Garin
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fernando L Vale
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sarah K Bick
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Neurosurgery, Vanderbilt University, Nashville, TN, USA
| | - Dario J Englot
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Neurosurgery, Vanderbilt University, Nashville, TN, USA
| | - Alvin V Terry
- Dept Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Christos Constantinidis
- Neuroscience Program, Vanderbilt University, Nashville, TN, 37235, USA; Dept Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA; Dept Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| | - David T Blake
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
5
|
Ananth MR, Gardus JD, Huang C, Palekar N, Slifstein M, Zaborszky L, Parsey RV, Talmage DA, DeLorenzo C, Role LW. A central role for acetylcholine in entorhinal cortex function and dysfunction with age in humans and mice. Cell Rep 2025; 44:115249. [PMID: 39891909 DOI: 10.1016/j.celrep.2025.115249] [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: 05/31/2024] [Revised: 11/15/2024] [Accepted: 01/09/2025] [Indexed: 02/03/2025] Open
Abstract
Structural and functional changes in the entorhinal cortex (EC) are among the earliest signs of cognitive aging. Here, we ask whether a compromised cholinergic system influences early EC impairments and plays a primary role in EC cognition. We evaluated the relationship between loss of integrity of cholinergic inputs to the EC and cognitive deficits in otherwise healthy humans and mice. Using in vivo imaging (PET/MRI) in older humans and high-resolution imaging in wild-type mice and mice with genetic susceptibility to Alzheimer's disease pathology, we establish that loss of cholinergic input to the EC is, in fact, an early feature in cognitive aging. Through mechanistic studies in mice, we find a central role for EC-projecting cholinergic neurons in the expression of EC-related behaviors. Our data demonstrate that alterations to the cholinergic EC are an early, conserved feature of cognitive aging across species and may serve as an early predictor of cognitive status.
Collapse
Affiliation(s)
- Mala R Ananth
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA.
| | - John D Gardus
- Department of Psychiatry and Behavioral Health, Stony Brook Medicine, Stony Brook, NY, USA
| | - Chuan Huang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA; Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nikhil Palekar
- Department of Psychiatry and Behavioral Health, Stony Brook Medicine, Stony Brook, NY, USA
| | - Mark Slifstein
- Department of Psychiatry and Behavioral Health, Stony Brook Medicine, Stony Brook, NY, USA
| | - Laszlo Zaborszky
- Center for Molecular and Behavioral Neuroscience, Rutgers University, New Newark, NJ, USA
| | - Ramin V Parsey
- Department of Psychiatry and Behavioral Health, Stony Brook Medicine, Stony Brook, NY, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - David A Talmage
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA
| | - Christine DeLorenzo
- Department of Psychiatry and Behavioral Health, Stony Brook Medicine, Stony Brook, NY, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Lorna W Role
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Manca R, De Marco M, Soininen H, Ruffini L, Venneri A. Changes in neurotransmitter-related functional connectivity along the Alzheimer's disease continuum. Brain Commun 2025; 7:fcaf008. [PMID: 39980737 PMCID: PMC11840171 DOI: 10.1093/braincomms/fcaf008] [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: 08/28/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
Alzheimer's disease may be associated with early dopamine dysfunction. However, its effects on neurofunctional alterations in the neurotransmission pathways remain elusive. In this study, positron emission tomography atlases and functional MRI data for 86 older adults with mild cognitive impairment Alzheimer's disease (MCI), 58 with mild Alzheimer's disease-dementia and 76 cognitively unimpaired were combined to investigate connectivity alterations associated with the dopaminergic and cholinergic systems. A cross-sectional design was used to compare neurotransmitter-related functional connectivity across groups and associations between functional connectivity and cognitive performance. The findings show that the Alzheimer's disease dementia group showed a decline in mesocorticolimbic dopamine-related connectivity in the precuneus but heightened connectivity in the thalamus, whereas the Alzheimer's disease-MCI group showed a decline in nigrostriatal connectivity in the left temporal areas. Acetylcholine-related connectivity decline was observed in both Alzheimer's disease-MCI and Alzheimer's disease-dementia primarily in the temporo-parietal areas. Episodic memory scores correlated positively with acetylcholine- and dopamine-related connectivity in the temporo-parietal cortex and negatively with dopamine-related functional connectivity in the fronto-thalamic areas. This study shows that connectivity alterations in acetylcholine and dopamine functional pathways parallel cognitive decline in Alzheimer's disease and might be a clinically relevant marker in early Alzheimer's disease.
Collapse
Affiliation(s)
- Riccardo Manca
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Matteo De Marco
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, 70210 Kuopio, Finland
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, 43126 Parma, Italy
| | - Annalena Venneri
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| |
Collapse
|
7
|
Cichon N, Grabowska W, Gorniak L, Stela M, Harmata P, Ceremuga M, Bijak M. Mechanistic and Therapeutic Insights into Flavonoid-Based Inhibition of Acetylcholinesterase: Implications for Neurodegenerative Diseases. Nutrients 2024; 17:78. [PMID: 39796512 PMCID: PMC11722824 DOI: 10.3390/nu17010078] [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: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Flavonoids are naturally occurring polyphenolic compounds known for their extensive range of biological activities. This review focuses on the inhibitory effects of flavonoids on acetylcholinesterase (AChE) and their potential as therapeutic agents for cognitive dysfunction. AChE, a serine hydrolase that plays a crucial role in cholinergic neurotransmission, is a key target in the treatment of cognitive impairments due to its function in acetylcholine hydrolysis. Natural polyphenolic compounds, particularly flavonoids, have demonstrated significant inhibition of AChE, positioning them as promising alternatives or adjuncts in neuropharmacology. This study specifically examines flavonoids such as quercetin, apigenin, kaempferol, and naringenin, investigating their inhibitory efficacy, binding mechanisms, and additional neuroprotective properties, including their antioxidant and anti-inflammatory effects. In vitro, in vivo, and in silico analyses reveal that these flavonoids effectively interact with both the active and peripheral anionic sites of AChE, resulting in increased acetylcholine levels and the stabilization of cholinergic signaling. Their mechanisms of action extend beyond mere enzymatic inhibition, as they also exhibit antioxidant and anti-amyloidogenic properties, thereby offering a multifaceted approach to neuroprotection. Given these findings, flavonoids hold considerable therapeutic potential as modulators of AChE, with implications for enhancing cognitive function and treating neurodegenerative diseases. Future studies should prioritize the enhancement of flavonoid bioavailability, evaluate their efficacy in clinical settings, and explore their potential synergistic effects when combined with established therapies to fully harness their potential as neurotherapeutic agents.
Collapse
Affiliation(s)
- Natalia Cichon
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland (L.G.); (M.S.); (M.B.)
| | - Weronika Grabowska
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland (L.G.); (M.S.); (M.B.)
| | - Leslaw Gorniak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland (L.G.); (M.S.); (M.B.)
| | - Maksymilian Stela
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland (L.G.); (M.S.); (M.B.)
| | - Piotr Harmata
- Faculty of Advanced Technologies and Chemistry, Military University of Technology, 2 gen. S. Kaliskiego St., 00-908 Warsaw, Poland;
| | - Michal Ceremuga
- Military Institute of Armoured and Automotive Technology, Okuniewska 1, 05-070 Sulejówek, Poland;
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland (L.G.); (M.S.); (M.B.)
| |
Collapse
|
8
|
Badal R, Ranjan S, Kumar L, Shekhawat L, Patel AK, Yadav P, Prajapati PK. Alzheimer's disease: A case study involving EEG-based fE/I ratio and pTau-181 protein analysis through nasal administration of Saraswata Ghrita. J Alzheimers Dis Rep 2024; 8:1763-1774. [PMID: 40034345 PMCID: PMC11863747 DOI: 10.1177/25424823241306771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/11/2024] [Indexed: 03/05/2025] Open
Abstract
Background Alzheimer's disease (AD) is a progressive neurodegenerative disorder that impairs memory, language, and cognitive functions and currently has no definitive cure. Saraswata Ghrita (SG), a traditional Ayurvedic remedy administered nasally, offers a holistic approach and is believed to directly affect brain functions through its unique delivery route. Objective This study aimed to evaluate the effectiveness of SG in improving cognitive function and neurochemical biomarkers in a patient with AD. Key outcomes included electroencephalography-based excitation/inhibition (fE/I) ratio, and levels of phosphorylated Tau-181 (pTau-181), serotonin, dopamine, acetylcholine, and dehydroepiandrosterone (DHEA). Methods A 90-day proof-of-concept clinical trial was conducted with one AD patient. Nasal administration of SG was performed twice daily. Measurements included EEG spectral power analysis across 1-48 Hz, cognitive function assessed by Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog), Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), and Quality of Life in Alzheimer's Disease (QoL-AD) scales, and biochemical analyses of pTau-181, serotonin, dopamine, acetylcholine, and DHEA. Results Notable improvements were observed: ADAS-Cog score decreased from 40 to 36, QoL-AD score increased from 23 to 31, MMSE score improved from 13 to 18, and MoCA score increased from 8 to 13. Biochemical markers showed a decrease in pTau-181 (12.50 pg/ml to 6.28 pg/ml), an increase in acetylcholine (13.73 pg/ml to 31.83 pg/ml), while serotonin and DHEA levels rose, and dopamine levels decreased (39.14 pg/ml to 36.21 pg/ml). Conclusions SG demonstrated potential in enhancing cognitive functions and neurochemical markers in AD, with the nasal route proving safe and effective. These findings suggest the value of traditional Ayurvedic treatments in contemporary AD management.
Collapse
Affiliation(s)
- Robin Badal
- Department of Rasashastra & Bhaishajya Kalpana, All India Institute of Ayurveda, New Delhi, India
| | - Shivani Ranjan
- Department of Electrical Engineering, Indian Institute of Technology Delhi, New Delhi, India
| | - Lalan Kumar
- Department of Electrical Engineering, Bharti School of Telecommunication, and Yardi School of Artificial Intelligence,
Indian Institute of Technology Delhi,
New Delhi, India
| | - Lokesh Shekhawat
- Department of Psychiatry, Atal Bihari Vajpayee Institute of Medical Sciences (ABVIMS) and Dr Ram Manohar Lohia Hospital,
New Delhi, India
| | - Ashok Kumar Patel
- School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Pramod Yadav
- Department of Rasashastra & Bhaishajya Kalpana, All India Institute of Ayurveda, New Delhi, India
| | | |
Collapse
|
9
|
Slater NM, Melzer TR, Myall DJ, Anderson TJ, Dalrymple-Alford JC. Cholinergic Basal Forebrain Integrity and Cognition in Parkinson's Disease: A Reappraisal of Magnetic Resonance Imaging Evidence. Mov Disord 2024; 39:2155-2172. [PMID: 39360864 DOI: 10.1002/mds.30023] [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: 06/14/2024] [Revised: 08/23/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Cognitive impairment is a well-recognized and debilitating symptom of Parkinson's disease (PD). Degradation in the cortical cholinergic system is thought to be a key contributor. Both postmortem and in vivo cholinergic positron emission tomography (PET) studies have provided valuable evidence of cholinergic system changes in PD, which are pronounced in PD dementia (PDD). A growing body of literature has employed magnetic resonance imaging (MRI), a noninvasive, more cost-effective alternative to PET, to examine cholinergic system structural changes in PD. This review provides a comprehensive discussion of the methodologies and findings of studies that have focused on the relationship between cholinergic basal forebrain (cBF) integrity, based on T1- and diffusion-weighted MRI, and cognitive function in PD. Nucleus basalis of Meynert (Ch4) volume has been consistently reduced in cognitively impaired PD samples and has shown potential utility as a prognostic indicator for future cognitive decline. However, the extent of structural changes in Ch4, especially in early stages of cognitive decline in PD, remains unclear. In addition, evidence for structural change in anterior cBF regions in PD has not been well established. This review underscores the importance of continued cross-sectional and longitudinal research to elucidate the role of cholinergic dysfunction in the cognitive manifestations of PD. © 2024 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Nicola M Slater
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Tracy R Melzer
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Daniel J Myall
- New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Tim J Anderson
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
- Department of Neurology, Christchurch Hospital, Te Whatu Ora Waitaha Canterbury, Christchurch, New Zealand
| | - John C Dalrymple-Alford
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
10
|
Pennington KR, Debs L, Chung S, Bava J, Garin CM, Vale FL, Bick SK, Englot DJ, Terry AV, Constantinidis C, Blake DT. Basal forebrain activation improves working memory in senescent monkeys. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582925. [PMID: 39574741 PMCID: PMC11580932 DOI: 10.1101/2024.03.01.582925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Brain aging contributes to cognitive decline and risk of dementia. Degeneration of the basal forebrain cholinergic system parallels these changes in aging, Alzheimer's dementia, Parkinson's dementia, and Lewy body dementia, and thus is a common element linked to executive function across the lifespan and in disease states. Here, we tested the potential of one-hour daily intermittent basal forebrain stimulation to improve cognition in senescent monkeys, and its mechanisms of action. Stimulation in five animals improved working memory duration in 8-12 weeks across all animals, with peak improvements observed in the first four weeks. In an ensuing three month period without stimulation, improvements were retained. With additional stimulation, performance remained above baseline throughout the 15 months of the study. Studies with a cholinesterase inhibitor produced inconsistent improvements in behavior. One of five animals improved significantly. Manipulating the stimulation pattern demonstrated selectivity for both stimulation and recovery period duration. Brain stimulation led to acute increases in cerebrospinal levels of tissue plasminogen activator, which is an activating element for two brain neurotrophins, Nerve Growth Factor (NGF) and Brain-Derived Growth Factor (BDNF). Stimulation also led to improved glucose utilization in stimulated hemispheres relative to contralateral. Glucose utilization also consistently declines with aging and some dementias. Together, these findings suggest that intermittent stimulation of the nucleus basalis of Meynert improves executive function and reverses some aspects of brain aging.
Collapse
Affiliation(s)
- Kendyl R Pennington
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| | - Luca Debs
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sophia Chung
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235
| | - Janki Bava
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Clément M Garin
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Fernando L Vale
- Dept Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA
| | - Sarah K Bick
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Neurosurgery, Vanderbilt University, Nashville TN
| | - Dario J Englot
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Neurosurgery, Vanderbilt University, Nashville TN
| | - Alvin V Terry
- Dept Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Christos Constantinidis
- Neuroscience Program, Vanderbilt University, Nashville, TN 37235
- Dept Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
- Dept Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232
| | - David T Blake
- Dept Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA
| |
Collapse
|
11
|
Yu Z, Luo F. The Role of Reactive Oxygen Species in Alzheimer's Disease: From Mechanism to Biomaterials Therapy. Adv Healthc Mater 2024; 13:e2304373. [PMID: 38508583 DOI: 10.1002/adhm.202304373] [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: 12/09/2023] [Revised: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Alzheimer's disease (AD) is a chronic, insidious, and progressive neurodegenerative disease that remains a clinical challenge for society. The fully approved drug lecanemab exhibits the prospect of therapy against the pathological processes, while debatable adverse events conflict with the drug concentration required for the anticipated therapeutic effects. Reactive oxygen species (ROS) are involved in the pathological progression of AD, as has been demonstrated in much research regarding oxidative stress (OS). The contradiction between anticipated dosage and adverse event may be resolved through targeted transport by biomaterials and get therapeutic effects through pathological progression via regulation of ROS. Besides, biomaterials fix delivery issues by promoting the penetration of drugs across the blood-brain barrier (BBB), protecting the drug from peripheral degradation, and elevating bioavailability. The goal is to comprehensively understand the mechanisms of ROS in the progression of AD disease and the potential of ROS-related biomaterials in the treatment of AD. This review focuses on OS and its connection with AD and novel biomaterials in recent years against AD via OS to inspire novel biomaterial development. Revisiting these biomaterials and mechanisms associated with OS in AD via thorough investigations presents a considerable potential and bright future for improving effective interventions for AD.
Collapse
Affiliation(s)
- Zhuohang Yu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
12
|
Sharma P, Sharma S, Yadav Y, Shukla P, Sagar R. Current pharmacophore based approaches for the development of new anti-Alzheimer's agents. Bioorg Med Chem 2024; 113:117926. [PMID: 39306973 DOI: 10.1016/j.bmc.2024.117926] [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: 07/24/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 10/13/2024]
Abstract
Amyloid beta peptide (Aβ) and hyperphosphorylated neuronal tau proteins accumulate in neurofibrillary tangles in Alzheimer's disease (AD), a chronic neurodegenerative illness. Chronic inflammation in the brain, which promotes disease progression, is another feature of the Alzheimer's disease pathogenesis. Approximately 60-70 % of dementia cases are caused by AD. The development of effective therapies for the treatment of AD is urgently needed given the severity of the condition and its rapidly rising prevalence. Cholinesterase inhibitors, beta-amyloid (A-beta), tau inhibitors, and many other medications are currently used as preventive medicine for AD. These medications can temporarily suppress dementia symptoms but cannot halt or reverse the disease's progression. Many international pharmaceutical companies have tried numerous times to develop an amyloid clearing medication based on the amyloid hypothesis, but without success. Therefore, the amyloid theory may not be entirely plausible. This review mainly covers the recent and important reported pharmacophores as the starting point to discuss already known targets like tau, butyrylcholinesterase, amyloid beta, and acetylcholinesterase and covers the literature between years 2019-2024.
Collapse
Affiliation(s)
- Prachi Sharma
- Department of Chemistry, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India
| | - Sunil Sharma
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Yogesh Yadav
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Paritosh Shukla
- Department of Chemistry, Birla Institute of Technology and Sciences, Pilani, Rajasthan 333031, India.
| | - Ram Sagar
- Glycochemistry Laboratory, School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
13
|
Galgani A, Scotto M, Giorgi FS. The Neuroanatomy of Induced Pluripotent Stem Cells: In Vitro Models of Subcortical Nuclei in Neurodegenerative Disorders. Curr Issues Mol Biol 2024; 46:10180-10199. [PMID: 39329959 PMCID: PMC11430477 DOI: 10.3390/cimb46090607] [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: 08/08/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Neuromodulatory subcortical systems (NSSs) are monoaminergic and cholinergic neuronal groups that are markedly and precociously involved in the pathogenesis of many neurodegenerative disorders (NDDs), including Parkinson's and Alzheimer's diseases. In humans, although many tools have been developed to infer information on these nuclei, encompassing neuroimaging and neurophysiological methods, a detailed and specific direct evaluation of their cellular features in vivo has been difficult to obtain until recent years. The development of induced pluripotent stem cell (iPSC) models has allowed research to deeply delve into the cellular and molecular biology of NSS neurons. In fact, iPSCs can be produced easily and non-invasively from patients' fibroblasts or circulating blood monocytes, by de-differentiating those cells using specific protocols, and then be re-differentiated towards neural phenotypes, which may reproduce the specific features of the correspondent brain neurons (including NSS ones) from the same patient. In this review, we summarized findings obtained in the field of NDDs using iPSCs, with the aim to understand how reliably these might represent in vitro models of NSS. We found that most of the current literature in the field of iPSCs and NSSs in NDDs has focused on midbrain dopaminergic neurons in Parkinson's disease, providing interesting results on cellular pathophysiology and even leading to the first human autologous transplantation. Differentiation protocols for noradrenergic, cholinergic, and serotoninergic neurons have also been recently defined and published. Thus, it might be expected that in the near future, this approach could extend to other NSSs and other NDDs.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Marco Scotto
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
- Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Filippo S. Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
- IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| |
Collapse
|
14
|
Skandalakis GP, Neudorfer C, Payne CA, Bond E, Tavakkoli AD, Barrios-Martinez J, Trutti AC, Koutsarnakis C, Coenen VA, Komaitis S, Hadjipanayis CG, Stranjalis G, Yeh FC, Banihashemi L, Hong J, Lozano AM, Kogan M, Horn A, Evans LT, Kalyvas A. Establishing connectivity through microdissections of midbrain stimulation-related neural circuits. Brain 2024; 147:3083-3098. [PMID: 38808482 PMCID: PMC11370807 DOI: 10.1093/brain/awae173] [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: 12/14/2023] [Revised: 03/15/2024] [Accepted: 04/21/2024] [Indexed: 05/30/2024] Open
Abstract
Comprehensive understanding of the neural circuits involving the ventral tegmental area is essential for elucidating the anatomofunctional mechanisms governing human behaviour, in addition to the therapeutic and adverse effects of deep brain stimulation for neuropsychiatric diseases. Although the ventral tegmental area has been targeted successfully with deep brain stimulation for different neuropsychiatric diseases, the axonal connectivity of the region is not fully understood. Here, using fibre microdissections in human cadaveric hemispheres, population-based high-definition fibre tractography and previously reported deep brain stimulation hotspots, we find that the ventral tegmental area participates in an intricate network involving the serotonergic pontine nuclei, basal ganglia, limbic system, basal forebrain and prefrontal cortex, which is implicated in the treatment of obsessive-compulsive disorder, major depressive disorder, Alzheimer's disease, cluster headaches and aggressive behaviours.
Collapse
Affiliation(s)
- Georgios P Skandalakis
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Clemens Neudorfer
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Caitlin A Payne
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Evalina Bond
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Armin D Tavakkoli
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | | | - Anne C Trutti
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam 15926, The Netherlands
| | - Christos Koutsarnakis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Volker A Coenen
- Department of Stereotactic and Functional Neurosurgery, Medical Center of the University of Freiburg, Freiburg 79106, Germany
- Medical Faculty of the University of Freiburg, Freiburg 79110, Germany
- Center for Deep Brain Stimulation, Medical Center of the University of Freiburg, Freiburg 79106, Germany
| | - Spyridon Komaitis
- Queens Medical Center, Nottingham University Hospitals NHS Foundation Trust, Nottingham NG7 2UH, UK
| | | | - George Stranjalis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Layla Banihashemi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jennifer Hong
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Andres M Lozano
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Andreas Horn
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Linton T Evans
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Aristotelis Kalyvas
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
15
|
Xia Y, Dore V, Fripp J, Bourgeat P, Laws SM, Fowler CJ, Rainey-Smith SR, Martins RN, Rowe C, Masters CL, Coulson EJ, Maruff P. Association of Basal Forebrain Atrophy With Cognitive Decline in Early Alzheimer Disease. Neurology 2024; 103:e209626. [PMID: 38885444 PMCID: PMC11254448 DOI: 10.1212/wnl.0000000000209626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In early Alzheimer disease (AD), β-amyloid (Aβ) deposition is associated with volume loss in the basal forebrain (BF) and cognitive decline. However, the extent to which Aβ-related BF atrophy manifests as cognitive decline is not understood. This study sought to characterize the relationship between BF atrophy and the decline in memory and attention in patients with early AD. METHODS Participants from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study who completed Aβ-PET imaging and repeated MRI and cognitive assessments were included. At baseline, participants were classified based on their clinical dementia stage and Aβ status, yielding groups that were cognitively unimpaired (CU) Aβ-, CU Aβ+, and mild cognitive impairment (MCI) Aβ+. Linear mixed-effects models were used to assess changes in volumetric measures of BF subregions and the hippocampus and changes in AIBL memory and attention composite scores for each group compared with CU Aβ- participants. Associations between Aβ burden, brain atrophy, and cognitive decline were evaluated and explored further using mediation analyses. RESULTS The cohort included 476 participants (72.6 ± 5.9 years, 55.0% female) with longitudinal data from a median follow-up period of 6.1 years. Compared with the CU Aβ- group (n = 308), both CU Aβ+ (n = 107) and MCI Aβ+ (n = 61) adults showed faster decline in BF and hippocampal volumes and in memory and attention (Cohen d = 0.73-1.74). Rates of atrophy in BF subregions and the hippocampus correlated with cognitive decline, and each individually mediated the impact of Aβ burden on memory and attention decline. When all mediators were considered simultaneously, hippocampal atrophy primarily influenced the effect of Aβ burden on memory decline (β [SE] = -0.139 [0.032], proportion mediated [PM] = 28.0%) while the atrophy of the posterior nucleus basalis of Meynert in the BF (β [SE] = -0.068 [0.029], PM = 13.1%) and hippocampus (β [SE] = -0.121 [0.033], PM = 23.4%) distinctively influenced Aβ-related attention decline. DISCUSSION These findings highlight the significant role of BF atrophy in the complex pathway linking Aβ to cognitive impairment in early stages of AD. Volumetric assessment of BF subregions could be essential in elucidating the relationships between the brain structure and behavior in AD.
Collapse
Affiliation(s)
- Ying Xia
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Vincent Dore
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Jurgen Fripp
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Pierrick Bourgeat
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Simon M Laws
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher J Fowler
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Stephanie R Rainey-Smith
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Ralph N Martins
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Christopher Rowe
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Colin L Masters
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Elizabeth J Coulson
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| | - Paul Maruff
- From the The Australian e-Health Research Centre (Y.X., V.D., J.F., P.B.), CSIRO Health and Biosecurity, Brisbane; Department of Nuclear Medicine and Centre for PET (V.D., C.R.), Austin Health, Melbourne; Centre for Precision Health (S.M.L.), Edith Cowan University; Collaborative Genomics and Translation Group (S.M.L.), School of Medical and Health Sciences, Edith Cowan University, Joondalup; Curtin Medical School (S.M.L.), Curtin University, Bentley; The Florey Institute of Neuroscience and Mental Health (C.J.F., C.R., C.L.M., P.M.), The University of Melbourne; Centre for Healthy Ageing (S.R.R.-S.), Health Futures Institute, Murdoch University; Australian Alzheimer's Research Foundation (S.R.R.-S., R.N.M.), Sarich Neuroscience Research Institute, Nedlands; School of Psychological Science (S.R.R.-S.), University of Western Australia, Crawley; School of Medical and Health Sciences (S.R.R.-S., R.N.M.), Edith Cowan University, Joondalup; Department of Biomedical Sciences (R.N.M.), Macquarie University, Sydney; Queensland Brain Institute (E.J.C.), and School of Biomedical Sciences (E.J.C.), The University of Queensland, Brisbane; and Cogstate Ltd. (P.M.), Melbourne, Australia
| |
Collapse
|
16
|
Okkels N, Grothe MJ, Taylor JP, Hasselbalch SG, Fedorova TD, Knudsen K, van der Zee S, van Laar T, Bohnen NI, Borghammer P, Horsager J. Cholinergic changes in Lewy body disease: implications for presentation, progression and subtypes. Brain 2024; 147:2308-2324. [PMID: 38437860 DOI: 10.1093/brain/awae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
Cholinergic degeneration is significant in Lewy body disease, including Parkinson's disease, dementia with Lewy bodies, and isolated REM sleep behaviour disorder. Extensive research has demonstrated cholinergic alterations in the CNS of these disorders. More recently, studies have revealed cholinergic denervation in organs that receive parasympathetic denervation. This enables a comprehensive review of cholinergic changes in Lewy body disease, encompassing both central and peripheral regions, various disease stages and diagnostic categories. Across studies, brain regions affected in Lewy body dementia show equal or greater levels of cholinergic impairment compared to the brain regions affected in Lewy body disease without dementia. This observation suggests a continuum of cholinergic alterations between these disorders. Patients without dementia exhibit relative sparing of limbic regions, whereas occipital and superior temporal regions appear to be affected to a similar extent in patients with and without dementia. This implies that posterior cholinergic cell groups in the basal forebrain are affected in the early stages of Lewy body disorders, while more anterior regions are typically affected later in the disease progression. The topographical changes observed in patients affected by comorbid Alzheimer pathology may reflect a combination of changes seen in pure forms of Lewy body disease and those seen in Alzheimer's disease. This suggests that Alzheimer co-pathology is important to understand cholinergic degeneration in Lewy body disease. Thalamic cholinergic innervation is more affected in Lewy body patients with dementia compared to those without dementia, and this may contribute to the distinct clinical presentations observed in these groups. In patients with Alzheimer's disease, the thalamus is variably affected, suggesting a different sequential involvement of cholinergic cell groups in Alzheimer's disease compared to Lewy body disease. Patients with isolated REM sleep behaviour disorder demonstrate cholinergic denervation in abdominal organs that receive parasympathetic innervation from the dorsal motor nucleus of the vagus, similar to patients who experienced this sleep disorder in their prodrome. This implies that REM sleep behaviour disorder is important for understanding peripheral cholinergic changes in both prodromal and manifest phases of Lewy body disease. In conclusion, cholinergic changes in Lewy body disease carry implications for understanding phenotypes and the influence of Alzheimer co-pathology, delineating subtypes and pathological spreading routes, and for developing tailored treatments targeting the cholinergic system.
Collapse
Affiliation(s)
- Niels Okkels
- Department of Neurology, Aarhus University Hospital, 8200 Aarhus N, Denmark
- Department of Nuclear Medicine and PET, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Reina Sofia Alzheimer's Centre, CIEN Foundation-ISCIII, 28031 Madrid, Spain
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Center, Department of Neurology, Copenhagen University Hospital, 2100 Copenhagen Ø, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2100 Copenhagen Ø, Denmark
| | - Tatyana D Fedorova
- Department of Nuclear Medicine and PET, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Karoline Knudsen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Sygrid van der Zee
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Teus van Laar
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Nicolaas I Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
- Neurology Service and GRECC, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI 48109, USA
- Parkinson's Foundation Research Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, 8200 Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus N, Denmark
| | - Jacob Horsager
- Department of Nuclear Medicine and PET, Aarhus University Hospital, 8200 Aarhus N, Denmark
| |
Collapse
|
17
|
Shafiee N, Fonov V, Dadar M, Spreng RN, Collins DL. Degeneration in Nucleus basalis of Meynert signals earliest stage of Alzheimer's disease progression. Neurobiol Aging 2024; 139:54-63. [PMID: 38608458 DOI: 10.1016/j.neurobiolaging.2024.03.003] [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: 10/12/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
Nucleus Basalis of Meynert (NbM), a crucial source of cholinergic projection to the entorhinal cortex (EC) and hippocampus (HC), has shown sensitivity to neurofibrillary degeneration in the early stages of Alzheimer's Disease. Using deformation-based morphometry (DBM) on up-sampled MRI scans from 1447 Alzheimer's Disease Neuroimaging Initiative participants, we aimed to quantify NbM degeneration along the disease trajectory. Results from cross-sectional analysis revealed significant differences of NbM volume between cognitively normal and early mild cognitive impairment cohorts, confirming recent studies suggesting that NbM degeneration happens before degeneration in the EC or HC. Longitudinal linear mixed-effect models were then used to compare trajectories of volume change after realigning all participants into a common timeline based on their cognitive decline. Results indicated the earliest deviations in NbM volumes from the cognitively healthy trajectory, challenging the prevailing idea that Alzheimer's originates in the EC. Converging evidence from cross-sectional and longitudinal models suggest that the NbM may be a focal target of early AD progression, which is often obscured by normal age-related decline.
Collapse
Affiliation(s)
- Neda Shafiee
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Vladimir Fonov
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Mahsa Dadar
- Department of Psychiatry, Douglas Mental Health University Institute and Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - R Nathan Spreng
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Douglas Mental Health University Institute and Douglas Research Centre, McGill University, Montreal, QC, Canada; Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Psychology, McGill University, Montreal, QC, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Schumacher J, Ray N, Teipel S, Storch A. Associations of cholinergic system integrity with cognitive decline in GBA1 and LRRK2 mutation carriers. NPJ Parkinsons Dis 2024; 10:127. [PMID: 38951174 PMCID: PMC11217433 DOI: 10.1038/s41531-024-00743-w] [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: 01/16/2024] [Accepted: 06/14/2024] [Indexed: 07/03/2024] Open
Abstract
In Parkinson's disease (PD), GBA1- and LRRK2-mutations are associated with different clinical phenotypes which might be related to differential involvement of the cholinergic system. We investigated cholinergic integrity in 149 asymptomatic GBA1 and 169 asymptomatic LRRK2 mutation carriers, 112 LRRK2 and 60 GBA1 carriers with PD, 492 idiopathic PD, and 180 controls from the PPMI cohort. Basal forebrain volumes were extracted and white matter pathways from nucleus basalis of Meynert (NBM) to cortex and from pedunculopontine nucleus (PPN) to thalamus were assessed with a free water-corrected DTI model. Bayesian ANCOVAs were conducted for group comparisons and Bayesian linear mixed models to assess associations with cognitive decline. Basal forebrain volumes were increased in asymptomatic GBA1 (Bayes Factor against the null hypothesis (BF10) = 75.2) and asymptomatic LRRK2 (BF10 = 57.0) compared to controls. Basal forebrain volumes were increased in LRRK2- compared to GBA1-PD (BF10 = 14.5) and idiopathic PD (BF10 = 3.6*107), with no difference between idiopathic PD and PD-GBA1 (BF10 = 0.25). Mean diffusivity along the medial NBM pathway was decreased in asymptomatic GBA1 compared to controls (BF10 = 30.3). Over 5 years, idiopathic PD and PD-GBA1 declined across all cognitive domains whereas PD-LRRK2 patients only declined in processing speed. We found an interaction between basal forebrain volume and time in predicting multiple cognitive domains in idiopathic PD and PD-GBA1, but not in PD-LRRK2. While LRRK2 and GBA1 mutations are both associated with increased basal forebrain volume at asymptomatic stages, this increase persists at the symptomatic PD stage only in LRRK2 and might be related to slower cognitive decline in these patients.
Collapse
Affiliation(s)
- Julia Schumacher
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany.
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany.
| | - Nicola Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, UK
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
- Department of Psychosomatic Medicine, University Medical Center Rostock, 18147, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University Medical Center Rostock, 18147, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147, Rostock, Germany
| |
Collapse
|
19
|
Lam AKF, Carrick J, Kao CH, Phillips CL, Zheng YZ, Yee BJ, Kim JW, Grunstein RR, Naismith SL, D’Rozario AL. Electroencephalographic slowing during REM sleep in older adults with subjective cognitive impairment and mild cognitive impairment. Sleep 2024; 47:zsae051. [PMID: 38394454 PMCID: PMC11168761 DOI: 10.1093/sleep/zsae051] [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: 09/19/2023] [Revised: 12/27/2023] [Indexed: 02/25/2024] Open
Abstract
STUDY OBJECTIVES In older adults with Alzheimer's disease, slowing of electroencephalographic (EEG) activity during REM sleep has been observed. Few studies have examined EEG slowing during REM in those with mild cognitive impairment (MCI) and none have examined its relationship with cognition in this at-risk population. METHODS Two hundred and ten older adults (mean age = 67.0, SD = 8.2 years) underwent comprehensive neuropsychological, medical, and psychiatric assessment and overnight polysomnography. Participants were classified as subjective cognitive impairment (SCI; n = 75), non-amnestic MCI (naMCI, n = 85), and amnestic MCI (aMCI, n = 50). REM EEG slowing was defined as (δ + θ)/(α + σ + β) power and calculated for frontal, central, parietal, and occipital regions. Analysis of variance compared REM EEG slowing between groups. Correlations between REM EEG slowing and cognition, including learning and memory, visuospatial and executive functions, were examined within each subgroup. RESULTS The aMCI group had significantly greater REM EEG slowing in the parietal and occipital regions compared to the naMCI and SCI groups (partial η2 = 0.06, p < 0.05 and 0.06, p < 0.05, respectively), and greater EEG slowing in the central region compared to SCI group (partial η2 = 0.03, p < 0.05). Greater REM EEG slowing in parietal (r = -0.49) and occipital regions (r = -0.38 [O1/M2] and -0.33 [O2/M1]) were associated with poorer visuospatial performance in naMCI. CONCLUSIONS REM EEG slowing may differentiate older adults with memory impairment from those without. Longitudinal studies are now warranted to examine the prognostic utility of REM EEG slowing for cognitive and dementia trajectories.
Collapse
Affiliation(s)
- Aaron Kin Fu Lam
- School of Psychology, University of Sydney, Camperdown, NSW, Australia
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- School of Psychological Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
| | - James Carrick
- School of Psychology, University of Sydney, Camperdown, NSW, Australia
| | - Chien-Hui Kao
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- School of Psychological Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
| | - Craig L Phillips
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- School of Psychological Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
| | - Yi Zhong Zheng
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
| | - Brendon J Yee
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- Central Clinical School, University of Sydney, Camperdown, NSW, Australia
| | - Jong Won Kim
- Department of Healthcare IT, Inje University, Gimhae, Gyeongsangnam-do, South Korea
| | - Ronald R Grunstein
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- Sydney Medical School, Faculty of Medicine and Health, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Sharon L Naismith
- School of Psychology, University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Angela L D’Rozario
- Woolcock Institute of Medical Research, Centre for Sleep and Chronobiology, Glebe, NSW, Australia
- School of Psychological Sciences, Faculty of Medicine, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
20
|
Lin M, Zhou Y, Liang P, Zheng R, Du M, Ke X, Zhang W, Shang P. Identification of Alzheimer's disease biomarkers and their immune function characterization. Arch Med Sci 2024; 21:233-257. [PMID: 40190307 PMCID: PMC11969561 DOI: 10.5114/aoms/188721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/15/2024] [Indexed: 04/09/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disease with neurogenic fiber tangles caused by amyloid-β protein plaques and tau protein hyperphosphorylation as the pathological manifestations. This study was based on multi-omics to investigate the mechanisms and immune characterization of AD. Material and methods Based on bulk RNA-seq (GSE122063 and GSE97760), we screened potential biomarkers for AD by differential expression analysis and machine learning algorithms. Then, we analyzed the expression characteristics and immune functions of the above biomarkers by scRNA-seq (single-cell RNA sequencing) data analysis (GSM4996463 and GSM4996462) and immune infiltration analysis. Results Five biomarkers (RBM3, GOLGA8A, ALS2, FSD2, and LOC100287628) were identified using machine learning algorithms. Single-cell analysis revealed distinct expression patterns of these biomarkers in astrocytes from AD samples compared to normal samples. Additionally, three key biomarkers were selected based on interaction networks, and the diagnostic models indicated high diagnostic efficacy for these biomarkers. Based on immune infiltration and correlation analyses, RBM3, GOLGA8A, and ALS2 were all highly correlated with CD8 T cell content in the immune microenvironment of AD. Conclusions The biomarkers identified in this study demonstrate significant diagnostic potential for AD. Notably, the downregulation of RBM3 in astrocytes and the decreased presence of CD8 T cells infiltrating brain tissue are potential risk factors for AD.
Collapse
Affiliation(s)
- Mingkai Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yue Zhou
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peixian Liang
- Department of Ultrasound, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruoyi Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minwei Du
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xintong Ke
- College of Teacher’s Education, Guangdong University of Education, Guangzhou, China
| | - Wenjing Zhang
- Department of Respiratory Medicine, Puyang People’s Hospital, Henan, China
| | - Pei Shang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Mayo Clinic, United States
| |
Collapse
|
21
|
Zerva MC, Triantafylloudis C, Paspaliaris V, Skoulakis EMC, Papanikolopoulou K. Choline Metabolites Reverse Differentially the Habituation Deficit and Elevated Memory of Tau Null Drosophila. Cells 2024; 13:746. [PMID: 38727282 PMCID: PMC11083674 DOI: 10.3390/cells13090746] [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: 03/12/2024] [Revised: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Impaired neuronal plasticity and cognitive decline are cardinal features of Alzheimer's disease and related Tauopathies. Aberrantly modified Tau protein and neurotransmitter imbalance, predominantly involving acetylcholine, have been linked to these symptoms. In Drosophila, we have shown that dTau loss specifically enhances associative long-term olfactory memory, impairs foot shock habituation, and deregulates proteins involved in the regulation of neurotransmitter levels, particularly acetylcholine. Interestingly, upon choline treatment, the habituation and memory performance of mutants are restored to that of control flies. Based on these surprising results, we decided to use our well-established genetic model to understand how habituation deficits and memory performance correlate with different aspects of choline physiology as an essential component of the neurotransmitter acetylcholine, the lipid phosphatidylcholine, and the osmoregulator betaine. The results revealed that the two observed phenotypes are reversed by different choline metabolites, implying that they are governed by different underlying mechanisms. This work can contribute to a broader knowledge about the physiologic function of Tau, which may be translated into understanding the mechanisms of Tauopathies.
Collapse
Affiliation(s)
- Maria-Christina Zerva
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, 16672 Vari, Greece (V.P.)
- Athens International Master’s Program in Neurosciences, Department of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Christos Triantafylloudis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, 16672 Vari, Greece (V.P.)
- Master’s Program in Molecular Biomedicine, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vassilis Paspaliaris
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, 16672 Vari, Greece (V.P.)
- Laboratory of Experimental Physiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efthimios M. C. Skoulakis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, 16672 Vari, Greece (V.P.)
| | - Katerina Papanikolopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Centre “Alexander Fleming”, 16672 Vari, Greece (V.P.)
| |
Collapse
|
22
|
Lane RM, Darreh-Shori T, Junge C, Li D, Yang Q, Edwards AL, Graham DL, Moore K, Mummery CJ. Onset of Alzheimer disease in apolipoprotein ɛ4 carriers is earlier in butyrylcholinesterase K variant carriers. BMC Neurol 2024; 24:116. [PMID: 38594621 PMCID: PMC11003149 DOI: 10.1186/s12883-024-03611-5] [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: 12/19/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The authors sought to examine the impact of the K-variant of butyrylcholinesterase (BCHE-K) carrier status on age-at-diagnosis of Alzheimer disease (AD) in APOE4 carriers. METHODS Patients aged 50-74 years with cerebrospinal fluid (CSF) biomarker-confirmed AD, were recruited to clinical trial (NCT03186989 since June 14, 2017). Baseline demographics, disease characteristics, and biomarkers were evaluated in 45 patients according to BCHE-K and APOE4 allelic status in this post-hoc study. RESULTS In APOE4 carriers (N = 33), the mean age-at-diagnosis of AD in BCHE-K carriers (n = 11) was 6.4 years earlier than in BCHE-K noncarriers (n = 22, P < .001, ANOVA). In APOE4 noncarriers (N = 12) there was no observed influence of BCHE-K. APOE4 carriers with BCHE-K also exhibited slightly higher amyloid and tau accumulations compared to BCHE-K noncarriers. A predominantly amyloid, limited tau, and limbic-amnestic phenotype was exemplified by APOE4 homozygotes with BCHE-K. In the overall population, multiple regression analyses demonstrated an association of amyloid accumulation with APOE4 carrier status (P < .029), larger total brain ventricle volume (P < .021), less synaptic injury (Ng, P < .001), and less tau pathophysiology (p-tau181, P < .005). In contrast, tau pathophysiology was associated with more neuroaxonal damage (NfL, P = .002), more synaptic injury (Ng, P < .001), and higher levels of glial activation (YKL-40, P = .01). CONCLUSION These findings have implications for the genetic architecture of prognosis in early AD, not the genetics of susceptibility to AD. In patients with early AD aged less than 75 years, the mean age-at-diagnosis of AD in APOE4 carriers was reduced by over 6 years in BCHE-K carriers versus noncarriers. The functional status of glia may explain many of the effects of APOE4 and BCHE-K on the early AD phenotype. TRIAL REGISTRATION NCT03186989 since June 14, 2017.
Collapse
Affiliation(s)
- Roger M Lane
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA.
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Stockholm, Sweden
| | - Candice Junge
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Dan Li
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Qingqing Yang
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | | | | - Katrina Moore
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | |
Collapse
|
23
|
Zhang D, Zhou L, Lu C, Feng T, Liu J, Wu T. Free-Water Imaging of the Nucleus Basalis of Meynert in Patients With Idiopathic REM Sleep Behavior Disorder and Parkinson Disease. Neurology 2024; 102:e209220. [PMID: 38489578 DOI: 10.1212/wnl.0000000000209220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/23/2023] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cognitive impairments are common in idiopathic REM sleep behavior disorder (iRBD), in which the cholinergic degeneration of nucleus basalis of Meynert (NBM) may play an important role. However, the progressive changes of NBM, the relationship between progressive NBM degeneration and progression of cognitive impairments, and whether degeneration of the NBM can predict cognitive decline in patients with iRBD remain unclear. This study aimed to investigate the cross-sectional and longitudinal microstructural alterations in the NBM of patients with iRBD using free-water imaging and whether free water in the NBM is related to cognitive, mood, and autonomic function. METHODS We compared the baseline free-water values in the NBM between 59 healthy controls (HCs), 57 patients with iRBD, 57 patients with Parkinson disease (PD) with normal cognition (PD-NC), and 64 patients with PD with cognitive impairment (PD-CI). Thirty patients with iRBD and 40 HCs had one longitudinal data. In patients with iRBD, we explored the associations between baseline and longitudinal changes of free-water values in the NBM and clinical characteristics and whether baseline free-water values in the NBM could predict cognitive decline. RESULTS IRBD, PD-NC, and PD-CI groups had significantly increased free-water values in the NBM compared with HCs, whereas PD-CI had higher free-water values compared with iRBD and PD-NC. In patients with iRBD, free-water values in the NBM were progressively elevated over follow-up and correlated with the progression of cognitive impairment and depression. Free-water values in the NBM could predict cognitive decline in the iRBD group. Furthermore, we found that patients with iRBD with cognitive impairment had higher relative change of free-water value in the NBM compared with patients with iRBD with normal cognition over follow-up. DISCUSSION This study proves that free-water values in the NBM are elevated cross-sectionally and longitudinally and are associated with the progression of cognitive impairment and depression in patients with iRBD. Moreover, the free-water value in the NBM can predict cognitive decline in patients with iRBD. Whether free-water imaging of the NBM has the potential to be a marker for monitoring progressive cognitive impairment and predicting the conversion to dementia in synucleinopathies needs further investigation.
Collapse
Affiliation(s)
- Dongling Zhang
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Liche Zhou
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Chenxi Lu
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Tao Feng
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jun Liu
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Tao Wu
- From the Center for Movement Disorders (D.Z., T.F., T.W.), Department of Neurology, Beijing Tiantan Hospital, Capital Medical University; China National Clinical Research Center for Neurological Diseases (D.Z., T.F., T.W.), Beijing; Department of Neurology and Institute of Neurology (L.Z., J.L.), Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; and Center for Brain Imaging Science and Technology (C.L.), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Zadrozny M, Drapich P, Gasiorowska-Bien A, Niewiadomski W, Harrington CR, Wischik CM, Riedel G, Niewiadomska G. Neuroprotection of Cholinergic Neurons with a Tau Aggregation Inhibitor and Rivastigmine in an Alzheimer's-like Tauopathy Mouse Model. Cells 2024; 13:642. [PMID: 38607082 PMCID: PMC11011792 DOI: 10.3390/cells13070642] [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: 02/27/2024] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Basal forebrain cholinergic dysfunction, most likely linked with tau protein aggregation, is a characteristic feature of Alzheimer's disease (AD). Recent evidence suggests that tau protein is a putative target for the treatment of dementia, and the tau aggregation inhibitor, hydromethylthionine mesylate (HMTM), has emerged as a potential disease-modifying treatment. However, its efficacy was diminished in patients already receiving approved acetylcholinesterase inhibitors. In this study, we ask whether this negative interaction can also be mimicked in experimental tau models of AD and whether the underlying mechanism can be understood. From a previous age profiling study, 6-month-old line 1 (L1) tau transgenic mice were characterized by a severe reduction in several cholinergic markers. We therefore assessed whether long-term pre-exposure with the acetylcholinesterase inhibitor rivastigmine alone and in conjunction with the tau aggregation inhibitor HMTM can reverse cholinergic deficits in L1. Rivastigmine and HMTM, and combinations of the two compounds were administered orally for 11 weeks to both L1 and wild-type mice. The brains were sectioned with a focus on the basal forebrain, motor cortex and hippocampus. Immunohistochemical staining and quantification of choline acetyltransferase (ChAT), tyrosine kinase A (TrkA)-positive neurons and relative optical intensity (ROI) for vesicular acetylcholine transporter (VAChT), and acetylcholinesterase (AChE) reactivity confirmed reversal of the diminished cholinergic phenotype of interneurons (nucleus accumbens, striatum) and projection neurons (medial septum, nucleus basalis magnocellularis) by HMTM, to a greater extent than by rivastigmine alone in L1 mice. Combined administration did not yield additivity but, in most proxies, led to antagonistic effects in which rivastigmine decreased the benefits shown with HMTM alone. Local markers (VAChT and AChE) in target structures of the basal forebrain, motor cortex and hippocampal CA3 seemed to be normalized by HMTM, but not by rivastigmine or the combination of both drugs. HMTM, which was developed as a tau aggregation inhibitor, strongly decreased the tau load in L1 mice, however, not in combination with rivastigmine. Taken together, these data confirm a cholinergic phenotype in L1 tau transgenic mice that resembles the deficits observed in AD patients. This phenotype is reversible by HMTM, but at the same time appears to be subject to a homeostatic regulation induced by chronic pre-treatment with an acetylcholinesterase inhibitor, which interferes with the efficacy of HMTM. The strongest phenotypic reversal coincided with a normalization of the tau load in the cortex and hippocampus of L1, suggesting that tau accumulation underpins the loss of cholinergic markers in the basal forebrain and its projection targets.
Collapse
Affiliation(s)
- Maciej Zadrozny
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Anna Gasiorowska-Bien
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
| | - Charles R. Harrington
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Claude M. Wischik
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
- TauRx Therapeutics Ltd., Aberdeen AB24 3FX, UK
| | - Gernot Riedel
- School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.R.H.); (C.M.W.); (G.R.)
| | - Grazyna Niewiadomska
- Mossakowski Medical Research Institute, 02-106 Warsaw, Poland; (M.Z.); (P.D.); (A.G.-B.); (W.N.)
- Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| |
Collapse
|
25
|
Lai YLL, Hsu FT, Yeh SY, Kuo YT, Lin HH, Lin YC, Kuo LW, Chen CY, Liu HS. Atrophy of the cholinergic regions advances from early to late mild cognitive impairment. Neuroradiology 2024; 66:543-556. [PMID: 38240769 DOI: 10.1007/s00234-024-03290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/10/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE We investigated the volumetric changes in the components of the cholinergic pathway for patients with early mild cognitive impairment (EMCI) and those with late mild cognitive impairment (LMCI). The effect of patients' apolipoprotein 4 (APOE-ε4) allele status on the structural changes were analyzed. METHODS Structural magnetic resonance imaging data were collected. Patients' demographic information, plasma data, and validated global cognitive composite scores were included. Relevant features were extracted for constructing machine learning models to differentiate between EMCI (n = 312) and LMCI (n = 541) and predict patients' neurocognitive function. The data were analyzed primarily through one-way analysis of variance and two-way analysis of covariance. RESULTS Considerable differences were observed in cholinergic structural changes between patients with EMCI and LMCI. Cholinergic atrophy was more prominent in the LMCI cohort than in the EMCI cohort (P < 0.05 family-wise error corrected). APOE-ε4 differentially affected cholinergic atrophy in the LMCI and EMCI cohorts. For LMCI cohort, APOE-ε4 carriers exhibited increased brain atrophy (left amygdala: P = 0.001; right amygdala: P = 0.006, and right Ch123, P = 0.032). EMCI and LCMI patients showed distinctive associations of gray matter volumes in cholinergic regions with executive (R2 = 0.063 and 0.030 for EMCI and LMCI, respectively) and language (R2 = 0.095 and 0.042 for EMCI and LMCI, respectively) function. CONCLUSIONS Our data confirmed significant cholinergic atrophy differences between early and late stages of mild cognitive impairment. The impact of the APOE-ε4 allele on cholinergic atrophy varied between the LMCI and EMCI groups.
Collapse
Affiliation(s)
- Ying-Liang Larry Lai
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Shu-Yi Yeh
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Tzu Kuo
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Hui-Hsien Lin
- CT/MR Division, Rotary Trading CO., LTD, Taipei, Taiwan
| | - Yi-Chun Lin
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Yu Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Medical Imaging, Taipei Medical University Hospital, Medical University, Taipei, Taiwan.
| | - Hua-Shan Liu
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
26
|
Li B, Deng S, Jiang H, Zhu W, Zhuo B, Du Y, Meng Z. The mechanistic effects of acupuncture in rodent neurodegenerative disease models: a literature review. Front Neurosci 2024; 18:1323555. [PMID: 38500484 PMCID: PMC10944972 DOI: 10.3389/fnins.2024.1323555] [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: 10/19/2023] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Neurodegenerative diseases refer to a battery of medical conditions that affect the survival and function of neurons in the brain, which are mainly presented with progressive loss of cognitive and/or motor function. Acupuncture showed benign effects in improving neurological deficits, especially on movement and cognitive function impairment. Here, we reviewed the therapeutic mechanisms of acupuncture at the neural circuit level in movement and cognition disorders, summarizing the influence of acupuncture in the dopaminergic system, glutamatergic system, γ-amino butyric acid-ergic (GABAergic) system, serotonergic system, cholinergic system, and glial cells at the circuit and synaptic levels. These findings can provide targets for clinical treatment and perspectives for further studies.
Collapse
Affiliation(s)
- Boxuan Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shizhe Deng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hailun Jiang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Weiming Zhu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bifang Zhuo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzheng Du
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihong Meng
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
27
|
López-Ortiz S, Caruso G, Emanuele E, Menéndez H, Peñín-Grandes S, Guerrera CS, Caraci F, Nisticò R, Lucia A, Santos-Lozano A, Lista S. Digging into the intrinsic capacity concept: Can it be applied to Alzheimer's disease? Prog Neurobiol 2024; 234:102574. [PMID: 38266702 DOI: 10.1016/j.pneurobio.2024.102574] [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: 06/06/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Historically, aging research has largely centered on disease pathology rather than promoting healthy aging. The World Health Organization's (WHO) policy framework (2015-2030) underscores the significance of fostering the contributions of older individuals to their families, communities, and economies. The WHO has introduced the concept of intrinsic capacity (IC) as a key metric for healthy aging, encompassing five primary domains: locomotion, vitality, sensory, cognitive, and psychological. Past AD research, constrained by methodological limitations, has focused on single outcome measures, sidelining the complexity of the disease. Our current scientific milieu, however, is primed to adopt the IC concept. This is due to three critical considerations: (I) the decline in IC is linked to neurocognitive disorders, including AD, (II) cognition, a key component of IC, is deeply affected in AD, and (III) the cognitive decline associated with AD involves multiple factors and pathophysiological pathways. Our study explores the application of the IC concept to AD patients, offering a comprehensive model that could revolutionize the disease's diagnosis and prognosis. There is a dearth of information on the biological characteristics of IC, which are a result of complex interactions within biological systems. Employing a systems biology approach, integrating omics technologies, could aid in unraveling these interactions and understanding IC from a holistic viewpoint. This comprehensive analysis of IC could be leveraged in clinical settings, equipping healthcare providers to assess AD patients' health status more effectively and devise personalized therapeutic interventions in accordance with the precision medicine paradigm. We aimed to determine whether the IC concept could be extended from older individuals to patients with AD, thereby presenting a model that could significantly enhance the diagnosis and prognosis of this disease.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | | | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Saúl Peñín-Grandes
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Claudia Savia Guerrera
- Department of Educational Sciences, University of Catania, 95125 Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", 00133 Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00143 Rome, Italy
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, 28670 Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), 28029 Madrid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain
| | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain.
| |
Collapse
|
28
|
Ananth MR, Gardus JD, Huang C, Palekar N, Slifstein M, Zaborszky L, Parsey RV, Talmage DA, DeLorenzo C, Role LW. Loss of cholinergic input to the entorhinal cortex is an early indicator of cognitive impairment in natural aging of humans and mice. RESEARCH SQUARE 2024:rs.3.rs-3851086. [PMID: 38260541 PMCID: PMC10802688 DOI: 10.21203/rs.3.rs-3851086/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
In a series of translational experiments using fully quantitative positron emission tomography (PET) imaging with a new tracer specific for the vesicular acetylcholine transporter ([18F]VAT) in vivo in humans, and genetically targeted cholinergic markers in mice, we evaluated whether changes to the cholinergic system were an early feature of age-related cognitive decline. We found that deficits in cholinergic innervation of the entorhinal cortex (EC) and decline in performance on behavioral tasks engaging the EC are, strikingly, early features of the aging process. In human studies, we recruited older adult volunteers that were physically healthy and without prior clinical diagnosis of cognitive impairment. Using [18F]VAT PET imaging, we demonstrate that there is measurable loss of cholinergic inputs to the EC that can serve as an early signature of decline in EC cognitive performance. These deficits are specific to the cholinergic circuit between the medial septum and vertical limb of the diagonal band (MS/vDB; CH1/2) to the EC. Using diffusion imaging, we further demonstrate impaired structural connectivity in the tracts between the MS/vDB and EC in older adults with mild cognitive impairment. Experiments in mouse, designed to parallel and extend upon the human studies, used high resolution imaging to evaluate cholinergic terminal density and immediate early gene (IEG) activity of EC neurons in healthy aging mice and in mice with genetic susceptibility to accelerated accumulation amyloid beta plaques and hyperphosphorylated mouse tau. Across species and aging conditions, we find that the integrity of cholinergic projections to the EC directly correlates with the extent of EC activation and with performance on EC-related object recognition memory tasks. Silencing EC-projecting cholinergic neurons in young, healthy mice during the object-location memory task impairs object recognition performance, mimicking aging. Taken together we identify a role for acetylcholine in normal EC function and establish loss of cholinergic input to the EC as an early, conserved feature of age-related cognitive decline in both humans and rodents.
Collapse
|
29
|
Rodriguez-Hernandez MA, Alemany I, Olofsson JK, Diaz-Galvan P, Nemy M, Westman E, Barroso J, Ferreira D, Cedres N. Degeneration of the cholinergic system in individuals with subjective cognitive decline: A systematic review. Neurosci Biobehav Rev 2024; 157:105534. [PMID: 38220033 DOI: 10.1016/j.neubiorev.2024.105534] [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: 10/10/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Subjective cognitive decline (SCD) is a risk factor for future cognitive impairment and dementia. It is uncertain whether the neurodegeneration of the cholinergic system is already present in SCD individuals. We aimed to review the current evidence about the association between SCD and biomarkers of degeneration in the cholinergic system. METHOD Original articles were extracted from three databases: Pubmed, Web of Sciences, and Scopus, in January 2023. Two researchers screened the studies independently. RESULTS A total of 11 research articles were selected. SCD was mostly based on amnestic cognitive complaints. Cholinergic system biomarkers included neuroimaging markers of basal forebrain volume, functional connectivity, transcranial magnetic stimulation, or biofluid. The evidence showed associations between basal forebrain atrophy, poorer connectivity of the cholinergic system, and SCD CONCLUSIONS: Degenerative changes in the cholinergic system can be present in SCD. Subjective complaints may help when identifying individuals with brain changes that are associated with cognitive impairment. These findings may have important implications in targeting individuals that may benefit from cholinergic-target treatments at very early stages of neurodegenerative diseases.
Collapse
Affiliation(s)
- Marta A Rodriguez-Hernandez
- Department of Psychology, Faculty of Health Sciences, University Fernando Pessoa-Canarias, Santa María de Guia, Spain
| | - Iris Alemany
- Department of Psychology, Faculty of Health Sciences, University Fernando Pessoa-Canarias, Santa María de Guia, Spain
| | - Jonas K Olofsson
- Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-lab), Stockholm University, Stockholm, Sweden
| | | | - Milan Nemy
- Department of Cybernetics, Faculty of Electrical Engineering, Czech Technical University in Prague, Prague, Czech Republic; Department of Biomedical Engineering and Assistive Technology, Czech Institute of Informatics, Robotics and Cybernetics, Czech Technical University in Prague, Prague, Czech Republic; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Center for Alzheimer Research, Stockholm, Sweden; Division of Clinical Geriatrics, Care Sciences and Society. Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Center for Alzheimer Research, Stockholm, Sweden; Division of Clinical Geriatrics, Care Sciences and Society. Karolinska Institutet, Stockholm, Sweden; Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Jose Barroso
- Department of Psychology, Faculty of Health Sciences, University Fernando Pessoa-Canarias, Santa María de Guia, Spain
| | - Daniel Ferreira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Center for Alzheimer Research, Stockholm, Sweden; Division of Clinical Geriatrics, Care Sciences and Society. Karolinska Institutet, Stockholm, Sweden
| | - Nira Cedres
- Department of Psychology, Faculty of Health Sciences, University Fernando Pessoa-Canarias, Santa María de Guia, Spain; Department of Psychology, Sensory Cognitive Interaction Laboratory (SCI-lab), Stockholm University, Stockholm, Sweden; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Center for Alzheimer Research, Stockholm, Sweden; Division of Clinical Geriatrics, Care Sciences and Society. Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
30
|
Pérez-González D, Lao-Rodríguez AB, Aedo-Sánchez C, Malmierca MS. Acetylcholine modulates the precision of prediction error in the auditory cortex. eLife 2024; 12:RP91475. [PMID: 38241174 PMCID: PMC10942646 DOI: 10.7554/elife.91475] [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] [Indexed: 01/21/2024] Open
Abstract
A fundamental property of sensory systems is their ability to detect novel stimuli in the ambient environment. The auditory brain contains neurons that decrease their response to repetitive sounds but increase their firing rate to novel or deviant stimuli; the difference between both responses is known as stimulus-specific adaptation or neuronal mismatch (nMM). Here, we tested the effect of microiontophoretic applications of ACh on the neuronal responses in the auditory cortex (AC) of anesthetized rats during an auditory oddball paradigm, including cascade controls. Results indicate that ACh modulates the nMM, affecting prediction error responses but not repetition suppression, and this effect is manifested predominantly in infragranular cortical layers. The differential effect of ACh on responses to standards, relative to deviants (in terms of averages and variances), was consistent with the representational sharpening that accompanies an increase in the precision of prediction errors. These findings suggest that ACh plays an important role in modulating prediction error signaling in the AC and gating the access of these signals to higher cognitive levels.
Collapse
Affiliation(s)
- David Pérez-González
- Cognitive and Auditory Neuroscience Laboratory, Institute of Neuroscience of Castilla y León, Calle Pintor Fernando GallegoSalamancaSpain
- Institute for Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Department of Basic Psychology, Psychobiology and Behavioural Science Methodology, Faculty of Psychology, Campus Ciudad Jardín, University of SalamancaSalamancaSpain
| | - Ana Belén Lao-Rodríguez
- Cognitive and Auditory Neuroscience Laboratory, Institute of Neuroscience of Castilla y León, Calle Pintor Fernando GallegoSalamancaSpain
- Institute for Biomedical Research of Salamanca (IBSAL)SalamancaSpain
| | - Cristian Aedo-Sánchez
- Cognitive and Auditory Neuroscience Laboratory, Institute of Neuroscience of Castilla y León, Calle Pintor Fernando GallegoSalamancaSpain
- Institute for Biomedical Research of Salamanca (IBSAL)SalamancaSpain
| | - Manuel S Malmierca
- Cognitive and Auditory Neuroscience Laboratory, Institute of Neuroscience of Castilla y León, Calle Pintor Fernando GallegoSalamancaSpain
- Institute for Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Department of Biology and Pathology, Faculty of Medicine, Campus Miguel de Unamuno, University of SalamancaSalamancaSpain
| |
Collapse
|
31
|
Montoya-Pedrón A, Ocaña Montoya CM, Santos Toural JE, Acosta Lee T, Sánchez-Hechavarría ME, López-Galán E, Muñoz-Bustos GA. Contingent Negative Variation in the Evaluation of Neurocognitive Disorders Due to Possible Alzheimer's Disease. Neurol Int 2024; 16:126-138. [PMID: 38251056 PMCID: PMC10801563 DOI: 10.3390/neurolint16010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
The usefulness of Contingent Negative Variation (CNV) potential as a biomarker of neurocognitive disorders due to possible Alzheimer's disease, is based on its possible physiological correlates. However, its application in the diagnostic evaluation of these disorders is still incipient. The aim of this study is to characterize the patterns of cognitive processing of information in the domain of nonspecific global attention, by recording potential CNV in a group of patients with neurocognitive disorders due to possible Alzheimer's disease. An experimental study of cases and controls was carried out. The sample included 39 patients classified according to DSM-5 with a neurocognitive disorder subtype possibly due Alzheimer's disease, and a Control Group of 53 subjects with normal cognitive functions. CNV potential was registered using standard protocol. The analysis of variance obtained significant differences in mean values and confidence intervals of total CNV amplitude between the three study groups. The late CNV segment amplitudes makes it possible to discriminate between the level of mild and major dysfunction in the group of patients. The CNV total amplitudes of potential allows for effective discrimination between normal cognitive functioning and neurocognitive disorders due to possible Alzheimer's disease.
Collapse
Affiliation(s)
- Arquímedes Montoya-Pedrón
- Department of Clinical Neurophysiology, General Hospital “Dr. Juan Bruno Zayas Alfonso”, Santiago de Cuba 90100, Cuba
| | | | | | - Tania Acosta Lee
- Department of Clinical Neurophysiology, General Hospital “Dr. Juan Bruno Zayas Alfonso”, Santiago de Cuba 90100, Cuba
| | - Miguel Enrique Sánchez-Hechavarría
- Departamento de Ciencias Clínicas y Preclínicas, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción 4070129, Chile
- Núcleo Científico de Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad Adventista de Chile, Chillán 8320000, Chile
- Laboratorio de Psicología, Departamento de Psicología, Universidad de Concepción, Concepción 4070386, Chile
| | - Erislandis López-Galán
- Facultad de Medicina 2, Universidad de Ciencias Médicas de Santiago de Cuba, Santiago de Cuba 90100, Cuba;
| | - Gustavo Alejandro Muñoz-Bustos
- Escuela de Kinesiología, Facultad de Salud y Ciencias Sociales, Campus El Boldal, Sede Concepción, Universidad de Las Américas, Concepción 4030000, Chile
| |
Collapse
|
32
|
Yoo HS, Kim HK, Lee JH, Chun JH, Lee HS, Grothe MJ, Teipel S, Cavedo E, Vergallo A, Hampel H, Ryu YH, Cho H, Lyoo CH. Association of Basal Forebrain Volume with Amyloid, Tau, and Cognition in Alzheimer's Disease. J Alzheimers Dis 2024; 99:145-159. [PMID: 38640150 DOI: 10.3233/jad-230975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Background Degeneration of cholinergic basal forebrain (BF) neurons characterizes Alzheimer's disease (AD). However, what role the BF plays in the dynamics of AD pathophysiology has not been investigated precisely. Objective To investigate the baseline and longitudinal roles of BF along with core neuropathologies in AD. Methods In this retrospective cohort study, we enrolled 113 subjects (38 amyloid [Aβ]-negative cognitively unimpaired, 6 Aβ-positive cognitively unimpaired, 39 with prodromal AD, and 30 with AD dementia) who performed brain MRI for BF volume and cortical thickness, 18F-florbetaben PET for Aβ, 18F-flortaucipir PET for tau, and detailed cognitive testing longitudinally. We investigated the baseline and longitudinal association of BF volume with Aβ and tau standardized uptake value ratio and cognition. Results Cross-sectionally, lower BF volume was not independently associated with higher cortical Aβ, but it was associated with tau burden. Tau burden in the orbitofrontal, insular, lateral temporal, inferior temporo-occipital, and anterior cingulate cortices were associated with progressive BF atrophy. Lower BF volume was associated with faster Aβ accumulation, mainly in the prefrontal, anterior temporal, cingulate, and medial occipital cortices. BF volume was associated with progressive decline in language and memory functions regardless of baseline Aβ and tau burden. Conclusions Tau deposition affected progressive BF atrophy, which in turn accelerated amyloid deposition, leading to a vicious cycle. Also, lower baseline BF volume independently predicted deterioration in cognitive function.
Collapse
Affiliation(s)
- Han Soo Yoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Han-Kyeol Kim
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae-Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Hyun Chun
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Department of Biostatistics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Michel J Grothe
- Reina Sofia Alzheimer Center, CIEN Foundation-ISCIII, Madrid, Spain
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)-Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University Medicine Rostock, Germany
| | - Enrica Cavedo
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Harald Hampel
- Sorbonne University Alzheimer Precision Medicine, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Song R, Chen H, Zhan R, Han M, Zhao L, Shen X. Vitamin E protects dopaminergic neurons against manganese-induced neurotoxicity through stimulation of CHRM1 and KCNJ4. J Trace Elem Med Biol 2024; 81:127326. [PMID: 37939525 DOI: 10.1016/j.jtemb.2023.127326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Manganese (Mn) overexposure can induce neurotoxicity and lead to manganism. Vitamin E (Vit E) has neuroprotective effects by acting as an ROS scavenger, preventing mitochondrial dysfunction and neuronal apoptosis. However, the effects of Vit E on Mn-induced nigrostriatal system lesions remains unknown. OBJECTIVES We aim to investigate whether Vit E has protective effects on Mn-induced nigrostriatal system lesions and mRNA expression profiles in the SN of mice. METHODS Sixty 8-week-old C57BL/6 male mice were randomly divided into the Control, MnCl2, MnCl2 +Vit E, and Vit E group. Twenty-four hours after the last injection, the behaviour test was performed. The numbers of dopaminergic neurons in Substantia nigra (SN), the contents of dopamine and its metabolite levels in striatium, and the morphology of mitochondria and nuclei in the dopaminergic neurons in SN were detected by immunofluorescence staining, high-performance liquid chromatography, and transmission electron microscopy. Transcriptome analysis was used to analyze the signaling pathways and RT-PCR was used to verify the mRNA levels. RESULTS Vit E ameliorates behavioral disorders and attenuates the loss of nigral dopaminergic neurons in the Mn-induced mouse model. In addition, Vit E antagonized Mn-induced toxicity by restoring mitochondrial function. The results of transcriptome sequencing and RTPCR show that the protective effect of Vit E was related to the upregulation of CHRM1 and KCNJ4 mRNA in the SN. CONCLUSIONS Vit E has neuroprotective effects on Mn-induced neurodegeneration in the nigrostriatal system. This effect may be related to the upregulation of CHRM1 and KCNJ4 mRNA stimulated by Vit E in the SN.
Collapse
Affiliation(s)
- Ruihan Song
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Huanhuan Chen
- Qingdao Municipal Center for Disease Control and Prevention/Qingdao Institute of Preventive Medicine, Qingdao, Shandong, China
| | - Runqing Zhan
- Qingdao University Affiliated Hiser Hospital, Qingdao, Shandong, China
| | - Miaomiao Han
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Longzhu Zhao
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China
| | - Xiaoli Shen
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
34
|
Moreno-Rodriguez M, Perez SE, Martinez-Gardeazabal J, Manuel I, Malek-Ahmadi M, Rodriguez-Puertas R, Mufson EJ. Frontal Cortex Lipid Alterations During the Onset of Alzheimer's Disease. J Alzheimers Dis 2024; 98:1515-1532. [PMID: 38578893 DOI: 10.3233/jad-231485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Although sporadic Alzheimer's disease (AD) is a neurodegenerative disorder of unknown etiology, familial AD is associated with specific gene mutations. A commonality between these forms of AD is that both display multiple pathogenic events including cholinergic and lipid dysregulation. Objective We aimed to identify the relevant lipids and the activity of their related receptors in the frontal cortex and correlating them with cognition during the progression of AD. Methods MALDI-mass spectrometry imaging (MSI) and functional autoradiography was used to evaluate the distribution of phospholipids/sphingolipids and the activity of cannabinoid 1 (CB1), sphingosine 1-phosphate 1 (S1P1), and muscarinic M2/M4 receptors in the frontal cortex (FC) of people that come to autopsy with premortem clinical diagnosis of AD, mild cognitive impairment (MCI), and no cognitive impairment (NCI). Results MALDI-MSI revealed an increase in myelin-related lipids, such as diacylglycerol (DG) 36:1, DG 38:5, and phosphatidic acid (PA) 40:6 in the white matter (WM) in MCI compared to NCI, and a downregulation of WM phosphatidylinositol (PI) 38:4 and PI 38:5 levels in AD compared to NCI. Elevated levels of phosphatidylcholine (PC) 32:1, PC 34:0, and sphingomyelin 38:1 were observed in discrete lipid accumulations in the FC supragranular layers during disease progression. Muscarinic M2/M4 receptor activation in layers V-VI decreased in AD compared to MCI. CB1 receptor activity was upregulated in layers V-VI, while S1P1 was downregulated within WM in AD relative to NCI. Conclusions FC WM lipidomic alterations are associated with myelin dyshomeostasis in prodromal AD, suggesting WM lipid maintenance as a potential therapeutic target for dementia.
Collapse
Affiliation(s)
- Marta Moreno-Rodriguez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Sylvia E Perez
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | | | - Ivan Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | | | - Rafael Rodriguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
35
|
Schumacher J, Kanel P, Dyrba M, Storch A, Bohnen NI, Teipel S, Grothe MJ. Structural and molecular cholinergic imaging markers of cognitive decline in Parkinson's disease. Brain 2023; 146:4964-4973. [PMID: 37403733 PMCID: PMC10689921 DOI: 10.1093/brain/awad226] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/14/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023] Open
Abstract
Cognitive decline in Parkinson's disease is related to cholinergic system degeneration, which can be assessed in vivo using structural MRI markers of basal forebrain volume and PET measures of cortical cholinergic activity. In the present study we aimed to examine the interrelation between basal forebrain degeneration and PET-measured depletion of cortical acetylcholinesterase activity as well as their relative contribution to cognitive impairment in Parkinson's disease. This cross-sectional study included 143 Parkinson's disease participants without dementia and 52 healthy control participants who underwent structural MRI, PET scanning with 11C-methyl-4-piperidinyl propionate (PMP) as a measure of cortical acetylcholinesterase activity, and a detailed cognitive assessment. Based on the fifth percentile of the overall cortical PMP PET signal from the control group, people with Parkinson's disease were subdivided into a normo-cholinergic (n = 94) and a hypo-cholinergic group (n = 49). Volumes of functionally defined posterior and anterior basal forebrain subregions were extracted using an established automated MRI volumetry approach based on a stereotactic atlas of cholinergic basal forebrain nuclei. We used Bayesian t-tests to compare basal forebrain volumes between controls, and normo- and hypo-cholinergic Parkinson's participants after covarying out age, sex and years of education. Associations between the two cholinergic imaging measures were assessed across all people with Parkinson's disease using Bayesian correlations and their respective relations with performance in different cognitive domains were assessed with Bayesian ANCOVAs. As a specificity analysis, hippocampal volume was added to the analysis. We found evidence for a reduction of posterior basal forebrain volume in the hypo-cholinergic compared to both normo-cholinergic Parkinson's disease [Bayes factor against the null model (BF10) = 8.2] and control participants (BF10 = 6.0), while for the anterior basal forebrain the evidence was inconclusive (BF10 < 3). In continuous association analyses, posterior basal forebrain volume was significantly associated with cortical PMP PET signal in a temporo-posterior distribution. The combined models for the prediction of cognitive scores showed that both cholinergic markers (posterior basal forebrain volume and cortical PMP PET signal) were independently related to multi-domain cognitive deficits, and were more important predictors for all cognitive scores, including memory scores, than hippocampal volume. We conclude that degeneration of the posterior basal forebrain in Parkinson's disease is accompanied by functional cortical changes in acetylcholinesterase activity and that both PET and MRI cholinergic imaging markers are independently associated with multi-domain cognitive deficits in Parkinson's disease without dementia. Comparatively, hippocampal atrophy only seems to have minimal involvement in the development of early cognitive impairment in Parkinson's disease.
Collapse
Affiliation(s)
- Julia Schumacher
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
- Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
| | - Prabesh Kanel
- University of Michigan Morris K. Udall Center for Excellence in Parkinson’s Disease Research, Ann Arbor, MI 48109, USA
- University of Michigan Parkinson’s Foundation Research Center of Excellence, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
| | - Alexander Storch
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
- Department of Neurology, University Medical Center Rostock, 18147 Rostock, Germany
| | - Nicolaas I Bohnen
- University of Michigan Morris K. Udall Center for Excellence in Parkinson’s Disease Research, Ann Arbor, MI 48109, USA
- University of Michigan Parkinson’s Foundation Research Center of Excellence, Ann Arbor, MI 48109, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA
- Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI 48105, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stefan Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, 18147 Rostock, Germany
- Department of Psychosomatic Medicine, University Medical Center Rostock, 18147 Rostock, Germany
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
36
|
Mahady L, Perez SE, Malek-Ahmadi M, Mufson EJ. Oligomeric, phosphorylated, and truncated tau and spliceosome pathology within the entorhinal-hippocampal connectome across stages of Alzheimer's disease. J Comp Neurol 2023; 531:2080-2108. [PMID: 36989381 PMCID: PMC10539478 DOI: 10.1002/cne.25466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 03/31/2023]
Abstract
Neurofibrillary tangles (NFTs) contain abnormally phosphorylated tau proteins, which spread within components of the medial temporal lobe (MTL) memory circuit in Alzheimer's disease (AD). Here, we used quantitative immunohistochemistry to determine the density of posttranslational oligomeric (TOC1 and TNT1), phosphorylated (AT8), and late truncated (TauC3) tau epitopes within the MTL subfields including entorhinal cortex (EC) layer II, subiculum, Cornu Ammonis (CA) subfields, and dentate gyrus (DG) in subjects who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), and AD. We also examined whether alterations of the nuclear alternative splicing protein, SRSF2, are associated with tau pathology. Although a significant increase in TOC1, TNT1, and AT8 neuron density occurred in the EC in MCI and AD, subicular, DG granule cell, and CA1 and CA3 densities were only significantly higher in AD. TauC3 counts were not different between connectome regions and clinical groups. SRSF2 intensity in AT8-positive cells decreased significantly in all regions independent of the clinical groups examined. CA1 and subicular AT8, TauC3, and oligomeric densities correlated across clinical groups. EC AT8 counts correlated with CA subfields and subicular and DG values across clinical groups. Oligomeric and AT8 CA1, EC, and subicular density correlated with Braak stage. Decreased nuclear SRSF2 in the presence of cytoplasmic phosphorylated tau suggests a dual-hit process in NFT formation within the entorhinal hippocampal connectome during the onset of AD. Although oligomeric and phosphorylated tau follow a stereotypical pattern, clinical disease stage determined density of tau deposition and not anatomic location within the entorhinal-hippocampal connectome.
Collapse
Affiliation(s)
- Laura Mahady
- Dept. of Translational Neuroscience, Phoenix, AZ
| | | | | | - Elliott J. Mufson
- Dept. of Translational Neuroscience, Phoenix, AZ
- Dept. of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013
| |
Collapse
|
37
|
Rozalem Aranha M, Iulita MF, Montal V, Pegueroles J, Bejanin A, Vaqué-Alcázar L, Grothe MJ, Carmona-Iragui M, Videla L, Benejam B, Arranz J, Padilla C, Valldeneu S, Barroeta I, Altuna M, Fernández S, Ribas L, Valle-Tamayo N, Alcolea D, González-Ortiz S, Bargalló N, Zetterberg H, Blennow K, Blesa R, Wisniewski T, Busciglio J, Cuello AC, Lleó A, Fortea J. Basal forebrain atrophy along the Alzheimer's disease continuum in adults with Down syndrome. Alzheimers Dement 2023; 19:4817-4827. [PMID: 37021589 DOI: 10.1002/alz.12999] [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: 10/03/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Basal forebrain (BF) degeneration occurs in Down syndrome (DS)-associated Alzheimer's disease (AD). However, the dynamics of BF atrophy with age and disease progression, its impact on cognition, and its relationship with AD biomarkers have not been studied in DS. METHODS We included 234 adults with DS (150 asymptomatic, 38 prodromal AD, and 46 AD dementia) and 147 euploid controls. BF volumes were extracted from T-weighted magnetic resonance images using a stereotactic atlas in SPM12. We assessed BF volume changes with age and along the clinical AD continuum and their relationship to cognitive performance, cerebrospinal fluid (CSF) and plasma amyloid/tau/neurodegeneration biomarkers, and hippocampal volume. RESULTS In DS, BF volumes decreased with age and along the clinical AD continuum and significantly correlated with amyloid, tau, and neurofilament light chain changes in CSF and plasma, hippocampal volume, and cognitive performance. DISCUSSION BF atrophy is a potentially valuable neuroimaging biomarker of AD-related cholinergic neurodegeneration in DS.
Collapse
Affiliation(s)
- Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lídia Vaqué-Alcázar
- Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michel J Grothe
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Javier Arranz
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Concepción Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sílvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Susana Fernández
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Laia Ribas
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Natalia Valle-Tamayo
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sofía González-Ortiz
- Hospital del Mar - Parc de Salut Mar, Barcelona, Spain
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Núria Bargalló
- Neuroradiology Section, Radiology Department, Diagnostic Image Center, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
- Magnetic Resonance Image Core Facility (IDIBAPS), Barcelona, Spain
| | - Henrik Zetterberg
- Queen Square Institute of Neurology, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- UK Dementia Research Institute, University College London, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, China, Hong Kong
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry and Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, New York, USA
| | - Jorge Busciglio
- Department of Neurobiology & Behavior, Institute for Memory Impairments and Neurological Disorders (iMIND), Center for the Neurobiology of Learning and Memory, University of California at Irvine, Irvine, California, USA
| | - A Claudio Cuello
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Department of Pharmacology, Oxford University, Oxford, UK
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Facultad de Medicina - Universitat Autònoma de Barcelona, Barcelona, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| |
Collapse
|
38
|
Schumacher J, Ray NJ, Hamilton CA, Bergamino M, Donaghy PC, Firbank M, Watson R, Roberts G, Allan L, Barnett N, O'Brien JT, Thomas AJ, Taylor JP. Free water imaging of the cholinergic system in dementia with Lewy bodies and Alzheimer's disease. Alzheimers Dement 2023; 19:4549-4563. [PMID: 36919460 DOI: 10.1002/alz.13034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Degeneration of cortical cholinergic projections from the nucleus basalis of Meynert (NBM) is characteristic of dementia with Lewy bodies (DLB) and Alzheimer's disease (AD), whereas involvement of cholinergic projections from the pedunculopontine nucleus (PPN) to the thalamus is less clear. METHODS We studied both cholinergic projection systems using a free water-corrected diffusion tensor imaging (DTI) model in the following cases: 46 AD, 48 DLB, 35 mild cognitive impairment (MCI) with AD, 38 MCI with Lewy bodies, and 71 controls. RESULTS Free water in the NBM-cortical pathway was increased in both dementia and MCI groups compared to controls and associated with cognition. Free water along the PPN-thalamus tract was increased only in DLB and related to visual hallucinations. Results were largely replicated in an independent cohort. DISCUSSION While NBM-cortical projections degenerate early in AD and DLB, the thalamic cholinergic input from the PPN appears to be more selectively affected in DLB and might associate with visual hallucinations. HIGHLIGHTS Free water in the NBM-cortical cholinergic pathways is increased in AD and DLB. NBM-cortical pathway integrity is related to overall cognitive performance. Free water in the PPN-thalamus cholinergic pathway is only increased in DLB, not AD. PPN-thalamus pathway integrity might be related to visual hallucinations in DLB.
Collapse
Affiliation(s)
- Julia Schumacher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
- Department of Neurology, University Medical Center Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock-Greifswald, Rostock, Germany
| | - Nicola J Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, UK
| | - Calum A Hamilton
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - Maurizio Bergamino
- Barrow Neurological Institute, Neuroimaging Research, Phoenix, Arizona, USA
| | - Paul C Donaghy
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - Michael Firbank
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - Rosie Watson
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Gemma Roberts
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - Louise Allan
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
- University of Exeter Medical School, Exeter, UK
| | - Nicola Barnett
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge School of Medicine, Cambridge, UK
| | - Alan J Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, Nebraska4 5PL , UK
| |
Collapse
|
39
|
Padulo C, Sestieri C, Punzi M, Picerni E, Chiacchiaretta P, Tullo MG, Granzotto A, Baldassarre A, Onofrj M, Ferretti A, Delli Pizzi S, Sensi SL. Atrophy of specific amygdala subfields in subjects converting to mild cognitive impairment. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2023; 9:e12436. [PMID: 38053753 PMCID: PMC10694338 DOI: 10.1002/trc2.12436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 12/07/2023]
Abstract
Introduction Accumulating evidence indicates that the amygdala exhibits early signs of Alzheimer's disease (AD) pathology. However, it is still unknown whether the atrophy of distinct subfields of the amygdala also participates in the transition from healthy cognition to mild cognitive impairment (MCI). Methods Our sample was derived from the AD Neuroimaging Initiative 3 and consisted of 97 cognitively healthy (HC) individuals, sorted into two groups based on their clinical follow-up: 75 who remained stable (s-HC) and 22 who converted to MCI within 48 months (c-HC). Anatomical magnetic resonance (MR) images were analyzed using a semi-automatic approach that combines probabilistic methods and a priori information from ex vivo MR images and histology to segment and obtain quantitative structural metrics for different amygdala subfields in each participant. Spearman's correlations were performed between MR measures and baseline and longitudinal neuropsychological measures. We also included anatomical measurements of the whole amygdala, the hippocampus, a key target of AD-related pathology, and the whole cortical thickness as a test of spatial specificity. Results Compared with s-HC individuals, c-HC subjects showed a reduced right amygdala volume, whereas no significant difference was observed for hippocampal volumes or changes in cortical thickness. In the amygdala subfields, we observed selected atrophy patterns in the basolateral nuclear complex, anterior amygdala area, and transitional area. Macro-structural alterations in these subfields correlated with variations of global indices of cognitive performance (measured at baseline and the 48-month follow-up), suggesting that amygdala changes shape the cognitive progression to MCI. Discussion Our results provide anatomical evidence for the early involvement of the amygdala in the preclinical stages of AD. Highlights Amygdala's atrophy marks elderly progression to mild cognitive impairment (MCI).Amygdala's was observed within the basolateral and amygdaloid complexes.Macro-structural alterations were associated with cognitive decline.No atrophy was found in the hippocampus and cortex.
Collapse
Affiliation(s)
- Caterina Padulo
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Department of HumanitiesUniversity of Naples Federico IINaplesItaly
| | - Carlo Sestieri
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical Technologies (ITAB)“G. d'Annunzio” University, Chieti‐PescaraChietiItaly
| | - Miriam Punzi
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Eleonora Picerni
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Piero Chiacchiaretta
- Department of Innovative Technologies in Medicine and Dentistry“G. d'Annunzio” University of Chieti‐Pescara, ChietiChietiItaly
- Advanced Computing CoreCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Maria Giulia Tullo
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Alberto Granzotto
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Antonello Baldassarre
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Marco Onofrj
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Antonio Ferretti
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Stefano Delli Pizzi
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | - Stefano L. Sensi
- Department of Neuroscience, Imaging, and Clinical SciencesUniversity “G. d'Annunzio” of Chieti‐PescaraChietiItaly
- Institute for Advanced Biomedical Technologies (ITAB)“G. d'Annunzio” University, Chieti‐PescaraChietiItaly
- Molecular Neurology UnitCenter for Advanced Studies and Technology (CAST)University “G. d'Annunzio” of Chieti‐PescaraChietiItaly
| | | |
Collapse
|
40
|
Dávila G, Torres-Prioris MJ, López-Barroso D, Berthier ML. Turning the Spotlight to Cholinergic Pharmacotherapy of the Human Language System. CNS Drugs 2023; 37:599-637. [PMID: 37341896 PMCID: PMC10374790 DOI: 10.1007/s40263-023-01017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/22/2023]
Abstract
Even though language is essential in human communication, research on pharmacological therapies for language deficits in highly prevalent neurodegenerative and vascular brain diseases has received little attention. Emerging scientific evidence suggests that disruption of the cholinergic system may play an essential role in language deficits associated with Alzheimer's disease and vascular cognitive impairment, including post-stroke aphasia. Therefore, current models of cognitive processing are beginning to appraise the implications of the brain modulator acetylcholine in human language functions. Future work should be directed further to analyze the interplay between the cholinergic system and language, focusing on identifying brain regions receiving cholinergic innervation susceptible to modulation with pharmacotherapy to improve affected language domains. The evaluation of language deficits in pharmacological cholinergic trials for Alzheimer's disease and vascular cognitive impairment has thus far been limited to coarse-grained methods. More precise, fine-grained language testing is needed to refine patient selection for pharmacotherapy to detect subtle deficits in the initial phases of cognitive decline. Additionally, noninvasive biomarkers can help identify cholinergic depletion. However, despite the investigation of cholinergic treatment for language deficits in Alzheimer's disease and vascular cognitive impairment, data on its effectiveness are insufficient and controversial. In the case of post-stroke aphasia, cholinergic agents are showing promise, particularly when combined with speech-language therapy to promote trained-dependent neural plasticity. Future research should explore the potential benefits of cholinergic pharmacotherapy in language deficits and investigate optimal strategies for combining these agents with other therapeutic approaches.
Collapse
Affiliation(s)
- Guadalupe Dávila
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - María José Torres-Prioris
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Diana López-Barroso
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain
- Department of Psychobiology and Methodology of Behavioral Sciences, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain
| | - Marcelo L Berthier
- Cognitive Neurology and Aphasia Unit, Centro de Investigaciones Médico-Sanitarias, University of Malaga, Marqués de Beccaria 3, 29010, Malaga, Spain.
- Instituto de Investigación Biomédica de Malaga-IBIMA, Malaga, Spain.
- Language Neuroscience Research Laboratory, Faculty of Psychology and Speech Therapy, University of Malaga, Malaga, Spain.
| |
Collapse
|
41
|
Mendivil-Perez M, Velez-Pardo C, Lopera F, Kosik KS, Jimenez-Del-Rio M. PSEN1 E280A Cholinergic-like Neurons and Cerebral Spheroids Derived from Mesenchymal Stromal Cells and from Induced Pluripotent Stem Cells Are Neuropathologically Equivalent. Int J Mol Sci 2023; 24:8957. [PMID: 37240306 PMCID: PMC10218810 DOI: 10.3390/ijms24108957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological condition characterized by the severe loss of cholinergic neurons. Currently, the incomplete understanding of the loss of neurons has prevented curative treatments for familial AD (FAD). Therefore, modeling FAD in vitro is essential for studying cholinergic vulnerability. Moreover, to expedite the discovery of disease-modifying therapies that delay the onset and slow the progression of AD, we depend on trustworthy disease models. Although highly informative, induced pluripotent stem cell (iPSCs)-derived cholinergic neurons (ChNs) are time-consuming, not cost-effective, and labor-intensive. Other sources for AD modeling are urgently needed. Wild-type and presenilin (PSEN)1 p.E280A fibroblast-derived iPSCs, menstrual blood-derived menstrual stromal cells (MenSCs), and umbilical cord-derived Wharton Jelly's mesenchymal stromal cells (WJ-MSCs) were cultured in Cholinergic-N-Run and Fast-N-Spheres V2 medium to obtain WT and PSEN 1 E280A cholinergic-like neurons (ChLNs, 2D) and cerebroid spheroids (CSs, 3D), respectively, and to evaluate whether ChLNs/CSs can reproduce FAD pathology. We found that irrespective of tissue source, ChLNs/CSs successfully recapitulated the AD phenotype. PSEN 1 E280A ChLNs/CSs show accumulation of iAPPβ fragments, produce eAβ42, present TAU phosphorylation, display OS markers (e.g., oxDJ-1, p-JUN), show loss of ΔΨm, exhibit cell death markers (e.g., TP53, PUMA, CASP3), and demonstrate dysfunctional Ca2+ influx response to ACh stimuli. However, PSEN 1 E280A 2D and 3D cells derived from MenSCs and WJ-MSCs can reproduce FAD neuropathology more efficiently and faster (11 days) than ChLNs derived from mutant iPSCs (35 days). Mechanistically, MenSCs and WJ-MSCs are equivalent cell types to iPSCs for reproducing FAD in vitro.
Collapse
Affiliation(s)
- Miguel Mendivil-Perez
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| | - Kenneth S. Kosik
- Neuroscience Research Institute, Department of Molecular Cellular Developmental Biology, University of California, Santa Barbara, CA 93106, USA;
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, Calle 62#52-59, Building 1, Room 412, SIU, Medellin 050010, Colombia; (M.M.-P.); (C.V.-P.); (F.L.)
| |
Collapse
|
42
|
Hirota Y, Sakakibara Y, Takei K, Nishijima R, Sekiya M, Iijima KM. Alzheimer's Disease-Related Phospho-Tau181 Signals Are Localized to Demyelinated Axons of Parvalbumin-Positive GABAergic Interneurons in an App Knock-In Mouse Model of Amyloid-β Pathology. J Alzheimers Dis 2023:JAD230121. [PMID: 37212118 DOI: 10.3233/jad-230121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
BACKGROUND The tau protein phosphorylated at Thr181 (p-tau181) in cerebrospinal fluid and blood is a sensitive biomarker for Alzheimer's disease (AD). Increased p-tau181 levels correlate well with amyloid-β (Aβ) pathology and precede neurofibrillary tangle formation in the early stage of AD; however, the relationship between p-tau181 and Aβ-mediated pathology is less well understood. We recently reported that p-tau181 represents axonal abnormalities in mice with Aβ pathology (AppNLGF). However, from which neuronal subtype(s) these p-tau181-positive axons originate remains elusive. OBJECTIVE The main purpose of this study is to differentiate neuronal subtype(s) and elucidate damage associated with p-tau181-positive axons by immunohistochemical analysis of AppNLGF mice brains. METHODS Colocalization between p-tau181 and (1) unmyelinated axons positive for vesicular acetylcholine transporter or norepinephrine transporter and (2) myelinated axons positive for vesicular glutamate transporter, vesicular GABA transporter, or parvalbumin in the brains of 24-month-old AppNLGF and control mice without Aβ pathology were analyzed. The density of these axons was also compared. RESULTS Unmyelinated axons of cholinergic or noradrenergic neurons did not overlap with p-tau181. By contrast, p-tau181 signals colocalized with myelinated axons of parvalbumin-positive GABAergic interneurons but not of glutamatergic neurons. Interestingly, the density of unmyelinated axons was significantly decreased in AppNLGF mice, whereas that of glutamatergic, GABAergic, or p-tau181-positive axons was less affected. Instead, myelin sheaths surrounding p-tau181-positive axons were significantly reduced in AppNLGF mice. CONCLUSION This study demonstrates that p-tau181 signals colocalize with axons of parvalbumin-positive GABAergic interneurons with disrupted myelin sheaths in the brains of a mouse model of Aβ pathology.
Collapse
Affiliation(s)
- Yu Hirota
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Japan Society for the Promotion of Science, Chiyoda-ku, Tokyo, Japan
| | - Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
43
|
Ray NJ, Kanel P, Bohnen NI. Atrophy of the Cholinergic Basal Forebrain can Detect Presynaptic Cholinergic Loss in Parkinson's Disease. Ann Neurol 2023; 93:991-998. [PMID: 36597786 PMCID: PMC10192078 DOI: 10.1002/ana.26596] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/19/2022] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Structural imaging of the cholinergic basal forebrain may provide a biomarker for cholinergic system integrity that can be used in motor and non-motor outcome studies in Parkinson's disease. However, no prior studies have validated these structural metrics with cholinergic nerve terminal in vivo imaging in Parkinson's disease. Here, we correlate cholinergic basal forebrain morphometry with the topography of vesicular acetylcholine transporter in a large Parkinson's sample. METHODS [18 F]-Fluoroethoxybenzovesamicol vesicular acetylcholine transporter positron emission tomography was carried out in 101 non-demented people with Parkinson's (76.24% male, mean age 67.6 ± 7.72 years, disease duration 5.7 ± 4.4 years). Subregional cholinergic basal forebrain volumes were measured using magnetic resonance imaging morphometry. Relationships were assessed via volume-of-interest based correlation analysis. RESULTS Subregional volumes of the cholinergic basal forebrain predicted cholinergic nerve terminal loss, with most robust correlations occurring between the posterior cholinergic basal forebrain and temporofrontal, insula, cingulum, and hippocampal regions, and with modest correlations in parieto-occipital regions. Hippocampal correlations were not limited to the cholinergic basal forebrain subregion Ch1-2. Correlations were also observed in the striatum, thalamus, and brainstem. INTERPRETATION Cholinergic basal forebrain morphometry is a robust predictor of regional cerebral vesicular acetylcholine transporter bindings, especially in the anterior brain. The relative lack of correlation between parieto-occipital binding and basal forebrain volumes may reflect the presence of more diffuse synaptopathy in the posterior cortex due to etiologies that extend well beyond the cholinergic system. ANN NEUROL 2023;93:991-998.
Collapse
Affiliation(s)
- Nicola J Ray
- Health, Psychology and Communities Research Centre, Department of Psychology, Manchester Metropolitan University, Manchester, UK
| | - Prabesh Kanel
- Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan, USA
- Parkinson's Foundation Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicolaas I Bohnen
- Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan, USA
- Parkinson's Foundation Center of Excellence, University of Michigan, Ann Arbor, Michigan, USA
- Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
44
|
Whiteaker P, George AA. Discoveries and future significance of research into amyloid-beta/α7-containing nicotinic acetylcholine receptor (nAChR) interactions. Pharmacol Res 2023; 191:106743. [PMID: 37084859 PMCID: PMC10228377 DOI: 10.1016/j.phrs.2023.106743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 04/23/2023]
Abstract
Initiated by findings that Alzheimer's disease is associated with a profound loss of cholinergic markers in human brain, decades of studies have examined the interactions between specific subtypes of nicotinic acetylcholine receptors and amyloid-β [derived from the amyloid precursor protein (APP), which is cleaved to yield variable isoforms of amyloid-β]. We review the evolving understanding of amyloid-β's roles in Alzheimer's disease and pioneering studies that highlighted a role of nicotinic acetylcholine receptors in mediating important aspects of amyloid-β's effects. This review also surveys the current state of research into amyloid-β / nicotinic acetylcholine receptor interactions. The field has reached an exciting point in which common themes are emerging from the wide range of prior research and a range of accessible, relevant model systems are available to drive further progress. We highlight exciting new areas of inquiry and persistent challenges that need to be considered while conducting this research. Studies of amyloid-β and the nicotinic acetylcholine receptor populations that it interacts with provide opportunities for innovative basic and translational scientific breakthroughs related to nicotinic receptor biology, Alzheimer's disease, and cholinergic contributions to cognition more broadly.
Collapse
Affiliation(s)
- Paul Whiteaker
- Virginia Commonwealth University School of Medicine, Department of Pharmacology and Toxicology, VCU Health Sciences Research Building, Box 980613, Richmond, VA 23298-0613, USA
| | - Andrew A George
- Virginia Commonwealth University School of Medicine, Department of Pharmacology and Toxicology, VCU Health Sciences Research Building, Box 980613, Richmond, VA 23298-0613, USA.
| |
Collapse
|
45
|
Chen Q, Chen F, Long C, Zhu Y, Jiang Y, Zhu Z, Lu J, Zhang X, Nedelska Z, Hort J, Zhang B. Spatial navigation is associated with subcortical alterations and progression risk in subjective cognitive decline. Alzheimers Res Ther 2023; 15:86. [PMID: 37098612 PMCID: PMC10127414 DOI: 10.1186/s13195-023-01233-6] [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: 12/04/2022] [Accepted: 04/18/2023] [Indexed: 04/27/2023]
Abstract
BACKGROUND Subjective cognitive decline (SCD) may serve as a symptomatic indicator for preclinical Alzheimer's disease; however, SCD is a heterogeneous entity regarding clinical progression. We aimed to investigate whether spatial navigation could reveal subcortical structural alterations and the risk of progression to objective cognitive impairment in SCD individuals. METHODS One hundred and eighty participants were enrolled: those with SCD (n = 80), normal controls (NCs, n = 77), and mild cognitive impairment (MCI, n = 23). SCD participants were further divided into the SCD-good (G-SCD, n = 40) group and the SCD-bad (B-SCD, n = 40) group according to their spatial navigation performance. Volumes of subcortical structures were calculated and compared among the four groups, including basal forebrain, thalamus, caudate, putamen, pallidum, hippocampus, amygdala, and accumbens. Topological properties of the subcortical structural covariance network were also calculated. With an interval of 1.5 years ± 12 months of follow-up, the progression rate to MCI was compared between the G-SCD and B-SCD groups. RESULTS Volumes of the basal forebrain, the right hippocampus, and their respective subfields differed significantly among the four groups (p < 0.05, false discovery rate corrected). The B-SCD group showed lower volumes in the basal forebrain than the G-SCD group, especially in the Ch4p and Ch4a-i subfields. Furthermore, the structural covariance network of the basal forebrain and right hippocampal subfields showed that the B-SCD group had a larger Lambda than the G-SCD group, which suggested weakened network integration in the B-SCD group. At follow-up, the B-SCD group had a significantly higher conversion rate to MCI than the G-SCD group. CONCLUSION Compared to SCD participants with good spatial navigation performance, SCD participants with bad performance showed lower volumes in the basal forebrain, a reorganized structural covariance network of subcortical nuclei, and an increased risk of progression to MCI. Our findings indicated that spatial navigation may have great potential to identify SCD subjects at higher risk of clinical progression, which may contribute to making more precise clinical decisions for SCD individuals who seek medical help.
Collapse
Affiliation(s)
- Qian Chen
- Department of Radiology, Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Futao Chen
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Cong Long
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yajing Zhu
- Department of Radiology, Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yaoxian Jiang
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhengyang Zhu
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiaming Lu
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xin Zhang
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zuzana Nedelska
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czechia
| | - Jakub Hort
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University, University Hospital Motol, Prague, Czechia
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210008, China.
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China.
- Medical Imaging Center, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China.
- Institute of Brain Science, Nanjing University, Nanjing, China.
| |
Collapse
|
46
|
Sola E, Moyano P, Flores A, García JM, García J, Anadon MJ, Frejo MT, Pelayo A, de la Cabeza Fernandez M, Del Pino J. Cadmium-promoted thyroid hormones disruption mediates ROS, inflammation, Aβ and Tau proteins production, gliosis, spongiosis and neurodegeneration in rat basal forebrain. Chem Biol Interact 2023; 375:110428. [PMID: 36868496 DOI: 10.1016/j.cbi.2023.110428] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Cadmium (Cd) produces cognition decline following single and repeated treatment, although the complete mechanisms are still unrevealed. Basal forebrain (BF) cholinergic neurons innervate the cortex and hippocampus, regulating cognition. Cd single and repeated exposure induced BF cholinergic neuronal loss, partly through thyroid hormones (THs) disruption, which may cause the cognition decline observed following Cd exposure. However, the mechanisms through which THs disruption mediate this effect remain unknown. To research the possible mechanisms through which Cd-induced THs deficiency may mediate BF neurodegeneration, Wistar male rats were treated with Cd for 1- (1 mg/kg) or 28-days (0.1 mg/kg) with or without triiodothyronine (T3, 40 μg/kg/day). Cd exposure promoted neurodegeneration, spongiosis, gliosis and several mechanisms related to these alterations (increased H202, malondialdehyde, TNF-α, IL-1β, IL-6, BACE1, Aβ and phosphorylated-Tau levels, and decreased phosphorylated-AKT and phosphorylated-GSK-3β levels). T3 supplementation partially reversed the effects observed. Our results show that Cd induces several mechanisms that may be responsible for the neurodegeneration, spongiosis and gliosis observed in the rats' BF, which are partially mediated by a reduction in THs levels. These data may help to explain the mechanisms through which Cd induces BF neurodegeneration, possibly leading to the cognitive decline observed, providing new therapeutic tools to prevent and treat these damages.
Collapse
Affiliation(s)
- Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Andrea Flores
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - José Manuel García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Jimena García
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - María José Anadon
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - María Teresa Frejo
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain
| | - Adela Pelayo
- Department of Legal Medicine, Psychiatry and Pathology, Medicine School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Maria de la Cabeza Fernandez
- Department of Chemistry in Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041, Madrid, Spain
| | - Javier Del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
47
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
48
|
Ananth MR, Rajebhosale P, Kim R, Talmage DA, Role LW. Basal forebrain cholinergic signalling: development, connectivity and roles in cognition. Nat Rev Neurosci 2023; 24:233-251. [PMID: 36823458 PMCID: PMC10439770 DOI: 10.1038/s41583-023-00677-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 01/18/2023] [Indexed: 02/25/2023]
Abstract
Acetylcholine plays an essential role in fundamental aspects of cognition. Studies that have mapped the activity and functional connectivity of cholinergic neurons have shown that the axons of basal forebrain cholinergic neurons innervate the pallium with far more topographical and functional organization than was historically appreciated. Together with the results of studies using new probes that allow release of acetylcholine to be detected with high spatial and temporal resolution, these findings have implicated cholinergic networks in 'binding' diverse behaviours that contribute to cognition. Here, we review recent findings on the developmental origins, connectivity and function of cholinergic neurons, and explore the participation of cholinergic signalling in the encoding of cognition-related behaviours.
Collapse
Affiliation(s)
- Mala R Ananth
- Section on Circuits, Synapses, and Molecular Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Prithviraj Rajebhosale
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ronald Kim
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David A Talmage
- Section on Genetics of Neuronal Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Lorna W Role
- Section on Circuits, Synapses, and Molecular Signalling, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
49
|
Gan C, Cao X, Wang L, Sun H, Ji M, Zhang H, Yuan Y, Zhang K. Cholinergic basal forebrain atrophy in Parkinson's disease with freezing of gait. Ann Clin Transl Neurol 2023; 10:814-824. [PMID: 37000969 DOI: 10.1002/acn3.51769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/21/2023] [Accepted: 03/19/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Mounting research support that cholinergic dysfunction plays a prominent role in freezing of gait (FOG), which commonly occurs in Parkinson's disease (PD). Basal forebrain (BF), especially the cholinergic nuclei 4 (Ch4), provides the primary source of the brain cholinergic input. However, whether the degeneration of BF and its innervated cortex contribute to the pathogenesis of FOG is unknown. OBJECTIVE To explore the role of structural alterations of BF and its innervated cortical brain regions in the pathogenesis of PD patients with freezing. METHODS Magnetic resonance imaging assessments and neurological assessments were performed on 20 PD patients with FOG (PD-FOG), 20 without FOG (PD-NFOG), and 21 healthy participants. Subregion volumes of the BF were compared among groups. Local gyrification index (LGI) was computed to reveal the cortical alternations. Relationships among subregional BF volumes, LGI, and the severity of FOG were evaluated by multiple linear regression. RESULTS Our study discovered that, compared to PD-NFOG, PD-FOG exhibited significant Ch4 atrophy (p = 4.6 × 10-5 ), accompanied by decreased LGI values in the left entorhinal cortex (p = 3.00 × 10-5 ) and parahippocampal gyrus (p = 2.90 × 10-5 ). Based on the regression analysis, Ch4 volume was negatively associated with FOG severity in PD-FOG group (β = -12.224, T = -2.556, p = 0.031). INTERPRETATION Our results imply that Ch4 degeneration and microstructural disorganization of its innervated cortical brain regions may play important roles in PD-FOG.
Collapse
|
50
|
Twarowski B, Herbet M. Inflammatory Processes in Alzheimer's Disease-Pathomechanism, Diagnosis and Treatment: A Review. Int J Mol Sci 2023; 24:6518. [PMID: 37047492 PMCID: PMC10095343 DOI: 10.3390/ijms24076518] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease is one of the most commonly diagnosed cases of senile dementia in the world. It is an incurable process, most often leading to death. This disease is multifactorial, and one factor of this is inflammation. Numerous mediators secreted by inflammatory cells can cause neuronal degeneration. Neuritis may coexist with other mechanisms of Alzheimer's disease, contributing to disease progression, and may also directly underlie AD. Although much has been established about the inflammatory processes in the pathogenesis of AD, many aspects remain unexplained. The work is devoted in particular to the pathomechanism of inflammation and its role in diagnosis and treatment. An in-depth and detailed understanding of the pathomechanism of neuroinflammation in Alzheimer's disease may help in the development of diagnostic methods for early diagnosis and may contribute to the development of new therapeutic strategies for the disease.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland
| |
Collapse
|