1
|
Achouri-Rassas A, Fray S, Said Z, Ben Sassi S, Ben Ali N, Baraket G. Genetic association study between rs2234253 (p.T96K) variant of TREM2 and Alzheimer's disease in a Tunisian population. Neurol Res 2025; 47:290-295. [PMID: 40043316 DOI: 10.1080/01616412.2025.2472841] [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: 05/08/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is the leading cause of major neurodegenerative cognitive impairment. The risk of developing AD is influenced by a complex interaction of genetic predisposition and environmental factors. Among the genetic risk factors, the APOE ɛ4 allele is the most significant, while variants in the TREM2 (Triggering Receptor Expressed on Myeloid Cells 2) and ABCA7 (ATP-binding cassette transporter A7) genes have also been associated with an increased risk of AD. OBJECTIVE This study aimed to investigate the association of APOE ɛ4, TREM2 gene variants (rs75932628 [p.R47H] and rs2234253 [p.T96K]), and ABCA7 gene variants (rs142076058 and rs115550680) with sporadic AD in the Tunisian population. Methods: A case-control study was conducted including 222 Tunisian patients diagnosed with sporadic AD and 99 cognitively healthy controls. Genotyping was performed to assess the presence and association of the selected genetic variants with AD. Statistical analyses were conducted to determine the significance of genetic associations. RESULTS A significant association was found between the TREM2 rs2234253 (p.T96K) variant and AD, with the T allele identified as a risk factor in the Tunisian population. The APOE ɛ4 allele was also associated with an increased risk of developing AD. However, no significant association was observed for the ABCA7 gene variants or the TREM2 rs75932628 (p.R47H) variant in either the AD or control groups. CONCLUSION Our findings suggest that the TREM2 rs2234253 (p.T96K) variant is a significant genetic risk factor for late-onset AD (LOAD) in the Tunisian population. Further studies with larger cohorts are needed to validate these findings and explore potential gene-gene interactions contributing to AD risk.
Collapse
Affiliation(s)
- Afef Achouri-Rassas
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
| | - Saloua Fray
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
- Neurological Department, Charles Nicolle Hospital, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Zakaria Said
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
- Department of Neurology, National Institute Mongi Ben Hamida of Neurology, Tunis, Tunisia
| | - Samia Ben Sassi
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
- Department of Neurology, National Institute Mongi Ben Hamida of Neurology, Tunis, Tunisia
| | - Nadia Ben Ali
- Research Laboratory LR12SP01 Temporal Lobe Pathology, Charles Nicolle Hospital Tunis, Tunisia
- Neurological Department, Charles Nicolle Hospital, Tunis, Tunisia
- Faculty of Medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Ghada Baraket
- Faculty of Sciences of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
2
|
Iturria-Medina Y, Poole VN, Zammit AR, Yu L, Tasaki S, Hong JH, Lopes KDP, Batalha C, Ridwan AR, Vialle RA, Sanchez-Rodriguez L, Geddes MR, Abadir P, Ortlund E, De Jager P, Menon V, Beeri MS, Buchman AS, Levin Y, Morgenstern D, Schneider JA, Daouk RK, Wyss-Coray T, Seyfried NT, Arfanakis K, Rosa-Neto P, Wang Y, Bennett DA. Translating the Post-Mortem Brain Multi-Omics Molecular Taxonomy of Alzheimer's Dementia to Living Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644323. [PMID: 40196602 PMCID: PMC11974700 DOI: 10.1101/2025.03.20.644323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Alzheimer's disease (AD) dementia is characterized by significant molecular and phenotypic heterogeneity, which confounds its mechanistic understanding, diagnosis, and effective treatment. In this study, we harness the most comprehensive dataset of paired ante-mortem blood omics, clinical, psychological, and post-mortem brain multi-omics data and neuroimaging to extensively characterize and translate the molecular taxonomy of AD dementia to living individuals. First, utilizing a comprehensive integration of eight complementary molecular layers from brain multi-omics data (N = 1,189), we identified three distinct molecular AD dementia subtypes exhibiting strong associations with cognitive decline, sex, psychological traits, brain morphology, and characterized by specific cellular and molecular drivers involving immune, vascular, and oligodendrocyte precursor cells. Next, in a significant translational effort, we developed predictive models to convert these advanced brain-derived molecular profiles (AD dementia pseudotimes and subtypes) into blood-, MRI- and psychological traits-based markers. The translation results underscore both the promise of these models and the opportunities for further enhancement. Our findings enhance the understanding of AD heterogeneity, underscore the value of multi-scale molecular approaches for elucidating causal mechanisms, and lay the groundwork for the development of novel therapies in living persons that target multi-level brain molecular subtypes of AD dementia.
Collapse
Affiliation(s)
- Yasser Iturria-Medina
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Victoria N. Poole
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Andrea R. Zammit
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Lei Yu
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Shinya Tasaki
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Joon Hwan Hong
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Katia de Paiva Lopes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Caio Batalha
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Abdur Raquib Ridwan
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ricardo A. Vialle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| | - Lazaro Sanchez-Rodriguez
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada
| | - Maiya Rachel Geddes
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Peter Abadir
- Johns Hopkins University School of Medicine, Baltimore, USA
| | - Eric Ortlund
- Department of Biochemistry at Emory University School of Medicine, Atlanta, USA
| | - Philip De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michal Schnaider Beeri
- Kreiger Klein Alzheimer’s Research Center, Brain Health Institute, Rutgers Health, NJ, USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Yishai Levin
- Israel National Center for Personalized Medicine at Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- Israel National Center for Personalized Medicine at Weizmann Institute of Science, Rehovot, Israel
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | | | | | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, McConnell Brain Imaging Centre (BIC), Montreal Neurological Institute, Montreal Neurological Institute-Hospital, Montreal, QC, Canada
- Douglas Hospital Research Centre - Centre intégré universitaire de santé et services sociaux de l’Ouest-de-l’Île-de-Montréal, Verdun, Quebec, Canada
- The Peter O’Donnell Jr. Brain Institute (OBI), University of Texas Southwestern Medical Centre (UTSW). Dallas, TX, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Instituto de Assistência Médica ao Servidor Público Estadual, Sao Paulo, SP, Brazil
| |
Collapse
|
3
|
Bhardwaj S, Jindal A, Singh S, Kaur R, Kaur Grewal A. Pharmacological Evaluation of Aescin for Neuroprotection in Intracerebroventricular Streptozotocin Model of Alzheimer's Disease in Experimental Rats. Assay Drug Dev Technol 2025. [PMID: 40095493 DOI: 10.1089/adt.2024.130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Alzheimer's disease (AD) is a neurological disorder that results in the loss of memory and cognitive functions linked to redox disbalance, neuroinflammation, neurotransmitters changes, and the accumulation of amyloid-beta (1-42) plaques in AD. In this study, rats were administered with intracerebroventricular (ICV) streptozotocin (STZ) to produce AD-like symptoms in rats. ICV-STZ bilaterally, 3 mg/kg, was infused on days 1 and 3 with the help of Hamilton syringe by fixing cannula at the target position of rat brain using coordinates -2 mm (anteriposterior), 1.6 mm Mediolateral (ML), and 1.5 mm (dorsoventral). Learning and spatial memory were checked using Morris water maze and elevated plus maze apparatus. In ICV-STZ, rats lost their spatial and learning memory, increased level of prooxidant like Lipid peroxidation (LPO), nitrite and reduced glutathione (GSH), catalase, and superoxide dismutase (SOD) level. The increased level acetylcholinesterase (AChE) catalyzed acetylcholine (ACh) concentration indicates cholinergic neuron degeneration. Furthermore, we found raised inflammatory markers and altered neurotransmitters level after ICV-STZ. Administration of aescin (10, 20, and 30 mg/kg, p.o.) dose-dependently ameliorated the behavioral alteration and inhibited inflammatory markers like tumor necrosis factor-alpha, interleukin-6 (IL-6), and IL-1β. Furthermore, aescin restored antioxidants like GSH, SOD, and catalase and reduced the nitrite and lipid peroxidation level. AChE enzyme causes degradation of ACh, and its level was declined after treatment with aescin. Aescin also restored GABA, norepinephrine, and serotonin level in the brain with prevention of raised glutamate level. Moreover, the histopathological study confirmed neuronal pathogenesis, and aescin significantly achieved neuroprotective effect via preventing neuroinflammation, balancing redox potential, and inhibiting AChE enzyme.
Collapse
Affiliation(s)
| | - Anu Jindal
- G.H.G. Khalsa College of Pharmacy, Ludhiana, Punjab, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Romanpreet Kaur
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | |
Collapse
|
4
|
Wang H, Lv P. Safety and efficacy of rivastigmine and memantine combined for treatment of patients with Alzheimer's disease: a retrospective study. Am J Transl Res 2025; 17:2240-2249. [PMID: 40226022 PMCID: PMC11982832 DOI: 10.62347/yopp4162] [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: 03/01/2024] [Accepted: 12/31/2024] [Indexed: 04/15/2025]
Abstract
OBJECTIVE To assess the effects and safety of combining rivastigmine hydrogen tartrate capsules with memantine tablets for Alzheimer's disease (AD). METHODS A retrospective study was conducted on AD patients admitted to The Third People's Hospital of Yongkang from November 2021 to June 2023. There were two groups: a single drug group (n=21) given only memantine tablets, and a combination group (n=39) treated with both rivastigmine hydrogen tartrate capsules and memantine tablets. Data were collected, including age, gender, education, overall response rate, adverse reaction rate, mini-mental state examination (MMSE), activity of daily living (ADL), behavioral pathology in Alzheimer's disease scale (BEHAVE-AD), serum tumor necrosis factor-α (TNF-α), serum interleukin-6 (IL-6) and serum Tau at baseline and at week 12. RESULTS In both groups, compared to baseline, at week 12, MMSE increased, while ADL, BEHAVE-AD, serum TNF-α, IL-6, and Tau decreased (all P<0.05). After treatment, compared with the single drug group at week 12, the combination group had a higher MMSE (t=2.519, P=0.015), better effectiveness (χ2=4.331, P=0.037), and lower ADL (t=2.418, P=0.019), BEHAVE-AD (t=3.231, P=0.002), TNF-α (t=3.496, P=0.001), IL-6 (t=2.513, P=0.015) and Tau (t=2.290, P=0.026) levels. CONCLUSION The combination of the two drugs was more effective in alleviating AD symptoms with comparable safety. It also showed an edge in suppressing pro-inflammatory cytokines and Tau in AD.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Geriatric Psychiatry, The Third People's Hospital of Yongkang No. 18 Feifeng Road, Xicheng Street, Yongkang City, Jinhua 321300, Zhejiang, China
| | - Pingping Lv
- Department of Geriatric Psychiatry, The Third People's Hospital of Yongkang No. 18 Feifeng Road, Xicheng Street, Yongkang City, Jinhua 321300, Zhejiang, China
| |
Collapse
|
5
|
Pérez R, Burgos V, Cabrera-Pardo JR, Ortiz L, Camins A, Ettcheto M, Schmidt B, Nchiozem-Ngnitedem VA, Paz C. 9-Hydroxyaristoquinolone: A New Indole Alkaloid Isolated from Aristotelia chilensis with Inhibitory Activity of NF-κB in HMC-3 Microglia Cells. Int J Mol Sci 2025; 26:2419. [PMID: 40141065 PMCID: PMC11942453 DOI: 10.3390/ijms26062419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Neurodegenerative diseases are characterized by a progressive process of degeneration and neuronal death in the nervous system, with neuroinflammation being one of the main factors contributing to the progression of these diseases. Aristotelia chilensis (Maqui) is a native tree of Chile used in the Mapuche folk medicine for wounds and digestive treatment. It produces edible black berries with the highest antioxidant capacity among berries, and the Mapuche people used it for producing an alcoholic beverage. The leaves of Maqui contain indole alkaloids with different pharmacological properties that suggest neuroprotective effects. Here, the isolation and chemical characterization of a new alkaloid, named 9-hydroxyaristoquinolone, and the evaluation of its anti-inflammatory activity in the microglial cell line HMC-3, treated with LPS, are reported. 9-Hydroxyaristoquinolone protects microglia from LPS-induced morphological changes at concentrations as low as 1 µM, with a reduction in IKBα-P levels and inhibition of the NF-κB pathway, which was assessed by THP-1 NF-κB dual cell reporter and Western blot in HMC-3 cells. In silico studies suggest that 9-hydroxyaristoquinolone does not induce hepatotoxicity or genotoxicity and exhibits BBB permeability.
Collapse
Affiliation(s)
- Rebeca Pérez
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Viviana Burgos
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Temuco 4780000, Chile;
| | - Jaime R. Cabrera-Pardo
- Laboratorio de Química Aplicada y Sustentable (LabQAS), Departamento de Química, Universidad del Bío-Bío, Avenida Collao 1202, Concepcion 4051381, Chile;
- College of Dental Medicine, Roseman University of Health Sciences, 10894 S. River Front Parkway, South Jordan, UT 84095, USA
| | - Leandro Ortiz
- Instituto de Ciencias Químicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile;
| | - Antoni Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.); (M.E.)
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d’Hebron, 171, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, 43204 Reus, Spain
| | - Miren Ettcheto
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.); (M.E.)
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d’Hebron, 171, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, 43204 Reus, Spain
| | - Bernd Schmidt
- Institut für Chemie, Universität Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany; (B.S.); (V.-A.N.-N.)
| | | | - Cristian Paz
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| |
Collapse
|
6
|
Nam YR, Kang M, Kim M, Seok MJ, Yang Y, Han YE, Oh SJ, Kim DG, Son H, Chang MY, Lee SH. Preparation of human astrocytes with potent therapeutic functions from human pluripotent stem cells using ventral midbrain patterning. J Adv Res 2025; 69:181-196. [PMID: 38521186 PMCID: PMC11954835 DOI: 10.1016/j.jare.2024.03.012] [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: 11/17/2023] [Revised: 02/19/2024] [Accepted: 03/16/2024] [Indexed: 03/25/2024] Open
Abstract
INTRODUCTION Astrocytes are glial-type cells that protect neurons from toxic insults and support neuronal functions and metabolism in a healthy brain. Leveraging these physiological functions, transplantation of astrocytes or their derivatives has emerged as a potential therapeutic approach for neurodegenerative disorders. METHODS To substantiate the clinical application of astrocyte-based therapy, we aimed to prepare human astrocytes with potent therapeutic capacities from human pluripotent stem cells (hPSCs). To that end, we used ventral midbrain patterning during the differentiation of hPSCs into astrocytes, based on the roles of midbrain-specific factors in potentiating glial neurotrophic/anti-inflammatory activity. To assess the therapeutic effects of human midbrain-type astrocytes, we transplanted them into mouse models of Parkinson's disease (PD) and Alzheimer's disease (AD). RESULTS Through a comprehensive series of in-vitro and in-vivo experiments, we were able to establish that the midbrain-type astrocytes exhibited the abilities to effectively combat oxidative stress, counter excitotoxic glutamate, and manage pathological protein aggregates. Our strategy for preparing midbrain-type astrocytes yielded promising results, demonstrating the strong therapeutic potential of these cells in various neurotoxic contexts. Particularly noteworthy is their efficacy in PD and AD-specific proteopathic conditions, in which the midbrain-type astrocytes outperformed forebrain-type astrocytes derived by the same organoid-based method. CONCLUSION The enhanced functions of the midbrain-type astrocytes extended to their ability to release signaling molecules that inhibited neuronal deterioration and senescence while steering microglial cells away from a pro-inflammatory state. This success was evident in both in-vitro studies using human cells and in-vivo experiments conducted in mouse models of PD and AD. In the end, our human midbrain-type astrocytes demonstrated remarkable effectiveness in alleviating neurodegeneration, neuroinflammation, and the pathologies associated with the accumulation of α-synuclein and Amyloid β proteins.
Collapse
Affiliation(s)
- Ye Rim Nam
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Minji Kang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Minji Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Min Jong Seok
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Yunseon Yang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Young Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Korea
| | - Mi-Yoon Chang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Premedicine, College of Medicine, Hanyang University, Korea; Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea.
| | - Sang-Hun Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea; Biomedical Research Institute, Hanyang University, Seoul, Korea; Department of Biochemistry & Molecular Biology, College of Medicine, Hanyang University, Korea.
| |
Collapse
|
7
|
Li L, Liu Z. SRF Facilitates Transcriptional Inhibition of Gem Expression by m6A Methyltransferase METTL3 to Suppress Neuronal Damage in Epilepsy. Mol Neurobiol 2025; 62:2903-2925. [PMID: 39190265 DOI: 10.1007/s12035-024-04396-x] [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: 11/01/2023] [Accepted: 07/21/2024] [Indexed: 08/28/2024]
Abstract
A bioinformatics analysis was conducted to screen for relevant expression datasets of the transcription factor SRF knockout mice. The aim was to investigate the relationship between SRF and m6A-related genes, predict how SRF regulates the m6A modification of GEM genes mediated by METTL3, and explore potential molecular mechanisms associated with neurotrauma. Disease gene databases such as GeneCards, DisGeNET, and Phenolyzer, and transcription factor databases TFDB and TRRUST, were used to obtain epilepsy-related genes and transcription factors. The intersection was then selected. Expression data of SRF knockout epilepsy mice were obtained from the GEO database and used to filter differentially expressed genes. Important module genes related to the disease were selected through WGCNA co-expression analysis. The intersection between these genes and the differentially expressed genes was performed, followed by PPI network analysis and GO/KEGG enrichment analysis. Furthermore, the core genes were selected using the cytoHubba plugin of the Cytoscape software. Differential expression analysis was performed on m6A-related factors in the GEO dataset, and the relationship between SRF and m6A-related factors and core genes was analyzed. The m6A binding sites of SRF with the METTL3 promoter and target gene Gem were predicted using the AnimalTFDB and SRAMP websites, respectively. We found that the transcription factor SRF may be a key gene in epilepsy during neuronal development. Further WGCNA analysis showed that 129 module genes were associated with SRF knockout epilepsy, and these differentially expressed genes were mainly enriched in the neuroactive ligand-receptor interaction pathway. The final results indicate that knocking out SRF may inhibit the transcription of METTL3, thereby inhibiting the m6A modification of Gem and leading to upregulation of Gem expression, thereby playing an important role in neuronal damage. Knocking out the SRF gene may inhibit the transcription of m6A methyltransferase METTL3, thereby inhibiting the m6A modification of GEM genes mediated by METTL3, promoting GEM gene expression, and leading to the occurrence of epilepsy-related neuron injury. Further investigation revealed that SRF overexpression can potentially enhance the transcription of METTL3, thus promoting m6A modification of GEM, resulting in downregulation of GEM expression. This process regulates oxidative stress in epileptic mouse neurons, suppresses inflammatory responses, and mitigates associated damage. Additionally, an in vitro neuronal epileptic model was established, and experimental techniques such as qRT-PCR and WB were employed to assess the expression of SRF, METTL3, and GEM in hippocampal tissues and neurons. The experimental results were consistent with our predictions, demonstrating that overexpression of SRF can inhibit the development of epilepsy-related neuronal damage. This study reveals that knockout of the SRF gene may suppress the transcription of m6A methyltransferase METTL3, thereby inhibiting m6A modification of the GEM gene mediated by METTL3 and subsequently promoting the expression of the GEM gene, leading to the occurrence of epilepsy-related neuronal damage.
Collapse
Affiliation(s)
- Lianling Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, P. R. China.
| | - Zhiguo Liu
- Department of Neurosurgery, Centtal Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, P. R. China
| |
Collapse
|
8
|
Zhang LH, Liu ST, Zhao Q, Liu XY, Liu T, Zhang Q, Liu MH, Zhao WX. Role of triggering receptor expressed on myeloid cells 2 in the pathogenesis of non-alcoholic fatty liver disease. World J Hepatol 2025; 17:102328. [PMID: 40027566 PMCID: PMC11866134 DOI: 10.4254/wjh.v17.i2.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/04/2025] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive disease. Without effective interventions, NAFLD can gradually develop to non-alcoholic steatohepatitis, fatty liver fibrosis, liver cirrhosis and even hepatocellular carcinoma. It is still to investigate the precise molecular mechanism behind the pathophysiology of NAFLD. Triggering receptor expressed on myeloid cells 2 (TREM2) can sense tissue injury and mediate immune remodeling, thereby inducing phagocytosis, lipid metabolism, and metabolic transfer, promoting cell survival and combating inflammatory activation. NAFLD might develop as a result of TREM2's regulatory role. We here briefly summarize the biological characteristics of TREM2 and its functions in the disease progression of NAFLD. Moreover, we propose to broaden the therapeutic strategy for NAFLD by targeting TREM2.
Collapse
Affiliation(s)
- Li-Hui Zhang
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Su-Tong Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Qing Zhao
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Xiao-Yan Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Tong Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Qiang Zhang
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Ming-Hao Liu
- Department of Spleen, Stomach and Hepatobiliary Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
| | - Wen-Xia Zhao
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China
- Department of Spleen, Stomach, Liver and Gallbladder Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China.
| |
Collapse
|
9
|
Mongelli M, De Angelis B, delle Cave V, Greco G, De Arcangelis A, Bernagozzi A, Salvemini C, Calabrese M, Christille JM, Cavalli A, Gustincich S, Monaci MG. The Public Knowledge of Precision Medicine and Genomic Research: A Survey in the Aosta Valley. J Pers Med 2025; 15:80. [PMID: 40137396 PMCID: PMC11943031 DOI: 10.3390/jpm15030080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Precision medicine (PM) considers the genetic variability of individuals to identify tailored diagnosis and treatments. It relies on the possibility of gathering the widest possible health data and genetic information from individuals to obtain a broad pool of comparative data. To achieve this goal, the Region of Valle d'Aosta, since 2019, has co-financed the research center CMP3VdA, aiming to sequence 5000 genomes of patients with neurodevelopmental, neurodegenerative, oncological, and organ transplantation diseases, and to investigate the genetic variability of the resident population. Methods: This paper presents the results of an online survey of 472 (328F) respondents regarding willingness to participate in the genomic project and awareness, attitudes, and concerns about PM. Results: The main results show that the vast majority (92.6%) would be willing to participate-a higher percentage than in previous studies. Age, education, and prior experience in the healthcare sector are significant factors influencing the awareness of PM. Additionally, subgroups organized by age, gender, and religiosity show significant differences with respect to participants' reasons for participating in research and which types of biological samples they would be willing to donate. Conclusions: Our findings can serve as a guide for stakeholders-particularly policymakers-to target institutional communication and achieve maximum participation in genomic research projects.
Collapse
Affiliation(s)
- Matteo Mongelli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Biagio De Angelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Valeria delle Cave
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Giuliano Greco
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Arianna De Arcangelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Andrea Bernagozzi
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Chiara Salvemini
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Matteo Calabrese
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Jean Marc Christille
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Andrea Cavalli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Computational and Chemical Biology, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Stefano Gustincich
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Center for Human Technologies, Non-Coding RNAs and RNA-Based Therapeutics, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152 Genova, Italy
| | - Maria Grazia Monaci
- Department of Human and Social Science, University of Valle d’Aosta, Strada Cappuccini 2A, 11100 Aosta, Italy
| |
Collapse
|
10
|
Cao ZL, Zhu LX, Wang HM, Zhu LJ. Microglial Regulation of Neural Networks in Neuropsychiatric Disorders. Neuroscientist 2025:10738584251316558. [PMID: 39932233 DOI: 10.1177/10738584251316558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Microglia serve as vital innate immune cells in the central nervous system, playing crucial roles in the generation and development of brain neurons, as well as mediating a series of immune and inflammatory responses. The morphologic transitions of microglia are closely linked to their function. With the advent of single-cell sequencing technology, the diversity of microglial subtypes is increasingly recognized. The intricate interactions between microglia and neuronal networks have significant implications for psychiatric disorders and neurodegenerative diseases. A deeper investigation of microglia in neurologic diseases such as Alzheimer disease, depression, and epilepsy can provide valuable insights in understanding the pathogenesis of diseases and exploring novel therapeutic strategies, thereby addressing issues related to central nervous system disorders.
Collapse
Affiliation(s)
- Zi-Lin Cao
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Xia Zhu
- Patent Examination Cooperation (JIANGSU) Center of the Patent Office, China National Intellectual Property Administration (CNIPA), Suzhou, China
| | - Hong-Mei Wang
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| | - Li-Juan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Bhardwaj V, Kumari S, Dhapola R, Sharma P, Beura SK, Singh SK, Vellingiri B, HariKrishnaReddy D. Shedding light on microglial dysregulation in Alzheimer's disease: exploring molecular mechanisms and therapeutic avenues. Inflammopharmacology 2025; 33:679-702. [PMID: 39609333 DOI: 10.1007/s10787-024-01598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 10/26/2024] [Indexed: 11/30/2024]
Abstract
Alzheimer's disease (AD) stands out as the foremost prevalent neurodegenerative disorder, characterized by a complex etiology. Various mechanisms have been proposed to elucidate its onset, encompassing amyloid-beta (Aβ) toxicity, tau hyperphosphorylation, oxidative stress and reactive gliosis. The hallmark of AD comprises Aβ and tau aggregation. These misfolded protein aggregates trigger the activation of glial cells, primarily microglia. Microglial cells serve as a major source of inflammatory mediators and their cytotoxic activation has been implicated in various aspects of AD pathology. Activated microglia can adopt M1 or M2 phenotypes, where M1 promotes inflammation by increasing pro-inflammatory cytokines and M2 suppresses inflammation by boosting anti-inflammatory factors. Overexpressed pro-inflammatory cytokines include interleukin (IL)-1β, IL-6 and tumor necrosis factor-α (TNF-α) in adjacent brain regions. Furthermore, microglial signaling pathways dysregulated in AD are myeloid differentiation primary-response protein 88 (Myd 88), colony-stimulating factor-1 receptor (CSF1R) and dedicator of cytokinesis 2 (DOCK2), which alter the physiology. Despite numerous findings, the causative role of microglia-mediated neuroinflammation in AD remains elusive. This review concisely explores cellular and molecular mechanisms of activated microglia and their correlation with AD pathogenesis. Additionally, it highlights promising therapeutics targeting microglia modulation, currently undergoing preclinical and clinical studies, for developing effective treatment for AD.
Collapse
Affiliation(s)
- Vanshu Bhardwaj
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Sunil Kumar Singh
- Department of Bio-Chemistry, School of Basic Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
12
|
Mallick R, Basak S, Chowdhury P, Bhowmik P, Das RK, Banerjee A, Paul S, Pathak S, Duttaroy AK. Targeting Cytokine-Mediated Inflammation in Brain Disorders: Developing New Treatment Strategies. Pharmaceuticals (Basel) 2025; 18:104. [PMID: 39861166 PMCID: PMC11769149 DOI: 10.3390/ph18010104] [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: 11/22/2024] [Revised: 12/26/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Cytokine-mediated inflammation is increasingly recognized for playing a vital role in the pathophysiology of a wide range of brain disorders, including neurodegenerative, psychiatric, and neurodevelopmental problems. Pro-inflammatory cytokines such as interleukin-1 (IL-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) cause neuroinflammation, alter brain function, and accelerate disease development. Despite progress in understanding these pathways, effective medicines targeting brain inflammation are still limited. Traditional anti-inflammatory and immunomodulatory drugs are effective in peripheral inflammatory illnesses. Still, they face substantial hurdles when applied to the central nervous system (CNS), such as the blood-brain barrier (BBB) and unwanted systemic effects. This review highlights the developing treatment techniques for modifying cytokine-driven neuroinflammation, focusing on advances that selectively target critical cytokines involved in brain pathology. Novel approaches, including cytokine-specific inhibitors, antibody-based therapeutics, gene- and RNA-based interventions, and sophisticated drug delivery systems like nanoparticles, show promise with respect to lowering neuroinflammation with greater specificity and safety. Furthermore, developments in biomarker discoveries and neuroimaging techniques are improving our ability to monitor inflammatory responses, allowing for more accurate and personalized treatment regimens. Preclinical and clinical trial data demonstrate the therapeutic potential of these tailored techniques. However, significant challenges remain, such as improving delivery across the BBB and reducing off-target effects. As research advances, the creation of personalized, cytokine-centered therapeutics has the potential to alter the therapy landscape for brain illnesses, giving patients hope for better results and a higher quality of life.
Collapse
Affiliation(s)
- Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India;
| | - Premanjali Chowdhury
- Institute of Public Health and Clinical Nutrition, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Prasenjit Bhowmik
- Department of Chemistry, Uppsala Biomedical Centre, Uppsala University, SE-751 23 Uppsala, Sweden;
- Department of Textile Engineering, Green University of Bangladesh, Narayanganj 1461, Bangladesh
| | - Ranjit K. Das
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai 603103, India; (A.B.); (S.P.)
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Queretaro 76130, Mexico;
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Chennai 603103, India; (A.B.); (S.P.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
13
|
Dolatshahi M, Commean PK, Rahmani F, Xu Y, Liu J, Hosseinzadeh Kassani S, Naghashzadeh M, Lloyd L, Nguyen C, McBee Kemper A, Hantler N, Ly M, Yu G, Flores S, Ippolito JE, Song SK, Sirlin CB, Dai W, Mittendorfer B, Morris JC, Benzinger TLS, Raji CA. Relationships between abdominal adipose tissue and neuroinflammation with diffusion basis spectrum imaging in midlife obesity. Obesity (Silver Spring) 2025; 33:41-53. [PMID: 39517107 DOI: 10.1002/oby.24188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE This study investigated how obesity, BMI ≥ 30 kg/m2, abdominal adiposity, and systemic inflammation relate to neuroinflammation using diffusion basis spectrum imaging. METHODS We analyzed data from 98 cognitively normal midlife participants (mean age: 49.4 [SD 6.2] years; 34 males [34.7%]; 56 with obesity [57.1%]). Participants underwent brain and abdominal magnetic resonance imaging (MRI), blood tests, and amyloid positron emission tomography (PET) imaging. Abdominal visceral and subcutaneous adipose tissue (VAT and SAT, respectively) was segmented, and Centiloids were calculated. Diffusion basis spectrum imaging parameter maps were created using an in-house script, and tract-based spatial statistics assessed white matter differences in high versus low BMI values, VAT, SAT, insulin resistance, systemic inflammation, and Centiloids, with age and sex as covariates. RESULTS Obesity, high VAT, and high SAT were linked to lower axial diffusivity, reduced fiber fraction, and increased restricted fraction in white matter. Obesity was additionally associated with higher hindered fraction and lower fractional anisotropy. Also, individuals with high C-reactive protein showed lower axial diffusivity. Higher restricted fraction correlated with continuous BMI and SAT particularly in male individuals, whereas VAT effects were similar in male and female individuals. CONCLUSIONS The findings suggest that, at midlife, obesity and abdominal fat are associated with reduced brain axonal density and increased inflammation, with visceral fat playing a significant role in both sexes.
Collapse
Affiliation(s)
- Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Paul K Commean
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yifei Xu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingxia Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | - Mahshid Naghashzadeh
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - LaKisha Lloyd
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Caitlyn Nguyen
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Abby McBee Kemper
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Nancy Hantler
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Maria Ly
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Gary Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Claude B Sirlin
- Liver Imaging Group, Department of Radiology, University of California, Los Angeles, California, USA
| | - Weiying Dai
- Department of Computer Science, State University of New York at Binghamton, Binghamton, New York, USA
| | - Bettina Mittendorfer
- Departments of Medicine and Nutrition & Exercise Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
14
|
Wen CH, Kang HY, Chan JY. Brain Amyloid-β Peptide Is Associated with Pain Intensity and Cognitive Dysfunction in Osteoarthritic Patients. Int J Mol Sci 2024; 25:12575. [PMID: 39684287 PMCID: PMC11641244 DOI: 10.3390/ijms252312575] [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: 11/07/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Considerable studies have demonstrated that osteoarthritis (OA) is a risk factor for dementia. The precise mechanisms underlying the association between OA and increased risk for cognitive dysfunction, however, remain unclear. This study aimed at exploring the associations between pro-inflammatory cytokines/chemokines, biomarkers of Alzheimer's disease (AD), pain intensity, and cognitive decline in knee joint OA patients. A total of 50 patients (26 in OA group and 24 in non-OA control group) were enrolled in this prospective, observational study. The visual analogue scale (VAS) score for pain intensity and Cognitive Abilities Screening Instrument (CASI) score for cognitive functions were examined in both groups. The plasma and cerebrospinal fluid (CSF) levels of pro-inflammatory molecules (IL-1β, IL-6, TNF-α, fractalkine, BDNF, MCP-1, and TGF-β), as well as biomarkers of AD (Aβ40, Aβ42, total-tau, and phospho-tau), were measured by multiplex immunoassay. Correlations among plasma or CSF biomarkers and questionnaire scores were assessed using Pearson's correlation coefficient and simple linear regressions. There were more patients in the OA group whose CASI cutoff percentiles were
Collapse
Affiliation(s)
- Chun-Hsien Wen
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Anesthesiology, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung 82144, Taiwan
- Department of Nursing, Meiho University, Pingtung 912009, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hong-Yo Kang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Julie Y.H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833401, Taiwan
| |
Collapse
|
15
|
Nie RZ, Luo HM, Liu YP, Wang SS, Hou YJ, Chen C, Wang H, Lv HL, Tao XY, Jing ZH, Zhang HK, Li PF. Food Functional Factors in Alzheimer's Disease Intervention: Current Research Progress. Nutrients 2024; 16:3998. [PMID: 39683391 DOI: 10.3390/nu16233998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is a complex multifactorial neurodegenerative disease. With the escalating aging of the global population, the societal burden of this disease is increasing. Although drugs are available for the treatment of AD, their efficacy is limited and there remains no effective cure. Therefore, the identification of safe and effective prevention and treatment strategies is urgently needed. Functional factors in foods encompass a variety of natural and safe bioactive substances that show potential in the prevention and treatment of AD. However, current research focused on the use of these functional factors for the prevention and treatment of AD is in its initial stages, and a complete theoretical and application system remains to be determined. An increasing number of recent studies have found that functional factors such as polyphenols, polysaccharides, unsaturated fatty acids, melatonin, and caffeine have positive effects in delaying the progression of AD and improving cognitive function. For example, polyphenols exhibit antioxidant, anti-inflammatory, and neuroprotective effects, and polysaccharides promote neuronal growth and inhibit inflammation and oxidative stress. Additionally, unsaturated fatty acids inhibit Aβ production and Tau protein phosphorylation and reduce neuroinflammation, and melatonin has been shown to protect nerve cells and improve cognitive function by regulating mitochondrial homeostasis and autophagy. Caffeine has also been shown to inhibit inflammation and reduce neuronal damage. Future research should further explore the mechanisms of action of these functional factors and develop relevant functional foods or nutritional supplements to provide new strategies and support for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rong-Zu Nie
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Huo-Min Luo
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Ya-Ping Liu
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Shuang-Shuang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Yan-Jie Hou
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Chen Chen
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hang Wang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hui-Lin Lv
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Xing-Yue Tao
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Zhao-Hui Jing
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Hao-Kun Zhang
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Pei-Feng Li
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Henan Key Laboratory of Cold Chain Food Quality and Safety Control, Zhengzhou University of Light Industry, Zhengzhou 450001, China
- Key Laboratory of Cold Chain Food Processing and Safety Control, Ministry of Education, Zhengzhou University of Light Industry, Zhengzhou 450001, China
| |
Collapse
|
16
|
Shah S, Jain H. Microglia-Associated Neuroinflammation in Alzheimer’s Disease and Its Therapeutic Potential. NEUROGLIA 2024; 5:452-466. [DOI: 10.3390/neuroglia5040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Background: Neuroinflammation has long been implicated in the progression of amyloid beta (Aβ) accumulation and the decline of cognitive function in Alzheimer’s disease (AD). The phenotype balance between A1 (toxic) and A2 (safe) microglial phenotypes to toxic illness in AD has become a hot research topic at present. Currently, many transcription factors, downstream signaling pathways, and molecular mechanisms that regulate the polarization of microglia are being explored. Furthermore, microglia may also exert a complex role in AD through the transformation of Aβ plaques or debris clearance, reflected in Aβ phagocytosis. One of the mediators of neuroinflammation in AD is the activated microglia. Therefore, the regulation of microglial function may be the key to successfully treating AD. Methods: This paper is a review article. PubMed, Embase, Scopus, and research meeting abstracts were searched up to 2024 for studies of microglia and neuroinflammation in Alzheimer’s Disease. Systematic information retrieval was performed, and appropriate studies were isolated based on important information available in the studies. The information from each of the articles was understood and extracted to form a database. Results: The similar neuropathological results between several animals and AD cases show the possibility of implementing microglia-related changes as an earlier diagnostic marker for AD in humans. The gene sets identified in various transcriptomic studies further foster this avenue of research by offering potential targets for therapeutic development. Substantial evidence, both in vitro and in vivo, has suggested that the loss of the normal A2 phenotype and the activation of toxic A1 microglia contribute to neurodegeneration in AD. Conclusions: Promoting or restoring the polarization of microglia towards the A2 phenotype may thus represent an effective therapeutic strategy for ameliorating neuroinflammation and progressive neurocognitive impairments. Multiple studies suggest that microglia-associated neuroinflammation at a special stage could also be protective, and, therefore, intervention should be delicate so that a beneficial response is retained.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA
| | - Hritvik Jain
- Department of Internal Medicine, All India Institute of Medical Sciencies, Jodhpur 342005, India
| |
Collapse
|
17
|
Lin H, Su L, Mao D, Yang G, Huang Q, Lan Y, Zeng J, Song W, Liang G, Wei Q, Zou D, Li R, Zou C. Identification of altered immune cell types and molecular mechanisms in Alzheimer's disease progression by single-cell RNA sequencing. Front Aging Neurosci 2024; 16:1477327. [PMID: 39610716 PMCID: PMC11602448 DOI: 10.3389/fnagi.2024.1477327] [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: 08/07/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by gradual loss of cognitive function. Understanding the molecular mechanisms is crucial for developing effective therapies. Methods Data from single-cell RNA sequencing (scRNA-seq) in the GSE181279 dataset and gene chips in the GSE63060 and GSE63061 datasets were collected and analyzed to identify immune cell types and differentially expressed genes. Cell communication, pseudotime trajectory, enrichment analysis, co- expression network, and short time-series expression miner were analyzed to identify disease-specific molecular and cellular mechanisms. Results We identified eight cell types (B cells, monocytes, natural killer cells, gamma-delta T cells, CD8+ T cells, Tem/Temra cytotoxic T cells, Tem/Trm cytotoxic T cells, and mucosal-associated invariant T cells) using scRNA-seq. AD samples were enriched in monocytes, CD8+ T cells, Tem/Temra cytotoxic T cells, and Tem/Trm cytotoxic T cells, whereas samples from healthy controls were enriched in natural killer and mucosal-associated invariant T cells. Five co-expression modules that were identified through weighted gene correlation network analysis were enriched in immune- inflammatory pathways. Candidate genes with higher area under the receiver operating characteristic curve values were screened, and the expression trend of Ubiquitin-Fold Modifier Conjugating Enzyme 1 (UFC1) gradually decreased from healthy controls to mild cognitive impairment and then to AD. Conclusion Our study suggests that peripheral immune cells may be a potential therapeutic target for AD. Candidate genes, particularly UFC1, may serve as potential biomarkers for progression of AD.
Collapse
Affiliation(s)
- Hua Lin
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Su
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Daniel Mao
- Department of Biology, Pennsylvania State University, University Park, PA, United States
| | - Grace Yang
- State College Area High School, State College, PA, United States
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenyi Song
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guining Liang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qingyan Wei
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongjie Li
- Department of Geriatrics, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chanhua Zou
- Department of Comprehensive Internal Medicine, Guangxi Medical University Caner Hospital, Nanning, China
| |
Collapse
|
18
|
Smeralda CL, Pandit S, Turrini S, Reilly J, Palmisano A, Sprugnoli G, Hampel H, Benussi A, Borroni B, Press D, Rotenberg A, El Fakhri G, Koch G, Rossi S, Santarnecchi E. The role of parvalbumin interneuron dysfunction across neurodegenerative dementias. Ageing Res Rev 2024; 101:102509. [PMID: 39306248 DOI: 10.1016/j.arr.2024.102509] [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: 07/05/2024] [Revised: 09/15/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024]
Abstract
Parvalbumin-positive (PV+) basket neurons are fast-spiking, non-adapting inhibitory interneurons whose oscillatory activity is essential for regulating cortical excitation/inhibition balance. Their dysfunction results in cortical hyperexcitability and gamma rhythm disruption, which have recently gained substantial traction as contributing factors as well as potential therapeutic targets for the treatment of Alzheimer's Disease (AD). Recent evidence indicates that PV+ cells are also impaired in Frontotemporal Dementia (FTD) and Dementia with Lewy bodies (DLB). However, no attempt has been made to integrate these findings into a coherent pathophysiological framework addressing the contribution of PV+ interneuron dysfunction to the generation of cortical hyperexcitability and gamma rhythm disruption in FTD and DLB. To fill this gap, we epitomized the most recent evidence on PV+ interneuron impairment in AD, FTD, and DLB, focusing on its contribution to the generation of cortical hyperexcitability and gamma oscillatory disruption and their interplay with misfolded protein accumulation, neuronal death, and clinical symptoms' onset. Our work deepens the current understanding concerning the role of PV+ interneuron dysfunction across neurodegenerative dementias, highlighting commonalities and differences among AD, FTD, and DLB, thus paving the way for identifying novel biomarkers and potential therapeutic targets for the treatment of these diseases.
Collapse
Affiliation(s)
- Carmelo Luca Smeralda
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Siddhartha Pandit
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sonia Turrini
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Centro Studi e Ricerche in Neuroscienze Cognitive, Dipartimento di Psicologia, University of Bologna, Italy
| | - Julianne Reilly
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Annalisa Palmisano
- Chair of Lifespan Developmental Neuroscience, TUD Dresden University of Technology, Dresden, Germany
| | - Giulia Sprugnoli
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Neurology Unit, Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniel Press
- Cognitive Neurology Unit, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Brookline, MA, USA
| | - Alexander Rotenberg
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giacomo Koch
- Human Physiology Unit, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Experimental Neuropsychophysiology Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Simone Rossi
- Siena Brain Investigation & Neuromodulation Lab (Si-BIN Lab), Unit of Neurology and Clinical Neurophysiology, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Precision Neuroscience & Neuromodulation Program, Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
de Oliveira BH, Lins EF, Kunde NF, Salgado ASI, Martins LM, Bobinski F, Vieira WF, Cassano P, Quialheiro A, Martins DF. Transcranial photobiomodulation increases cognition and serum BDNF levels in adults over 50 years: A randomized, double-blind, placebo-controlled trial. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 260:113041. [PMID: 39423445 DOI: 10.1016/j.jphotobiol.2024.113041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND There is a significant lack of therapeutic options for mild cognitive impairment (MCI), which is rapidly becoming a global epidemic due to aging. Transcranial photobiomodulation (t-PBM) involves delivering near-infrared light (NIR) to the scalp, targeting cortical areas of the brain. NIR t-PBM has recently emerged as a potential therapy for various neurodegenerative conditions, including memory issues. AIMS This study aimed to evaluate cognition scores (primary outcome), depression, anxiety, resilience scores, neuroplasticity, and neurodegeneration biomarkers (secondary outcomes) in individuals with MCI undergoing t-PBM therapy or receiving a placebo. MATERIALS AND METHODS A total of 93 older adult individuals with MCI were randomly assigned to either a t-PBM (n = 47) or Placebo (n = 46) group. Clinical assessments were conducted at baseline, 60 days post-treatment, and a 150-day follow-up. We also measured serum levels of brain-derived neurotrophic factor (BDNF), a neuroplasticity biomarker, as well as neuron-specific enolase (NSE) and calcium-binding protein B (S100B), which are neurodegeneration biomarkers. Intervention effects were analyzed using repeated measures (RM) two-way ANOVA followed by Tukey post hoc test. Fischer's exact test and Generalized Estimating Equations (GEE) were also applied. RESULTS Of the 93 older adults individuals invited to participate, 76 (t-PBM: 40, placebo: 36) completed the study. The t-PBM significantly improved cognition as measured by the Montreal Cognitive Assessment (MoCA) compared to placebo (p = 0.0301). The delta values for MoCA scores were 3.20 in the t-PBM group and 1.97 in the placebo group. This effect persisted until the three-month follow-up, accompanied by increased BDNF levels in the t-PBM group but not in the placebo group (p = 0.0046). The delta values for BDNF were 821.94 in the t-PBM group and 359.41 in the placebo group. t-PBM did not alter depression, anxiety, resilience scores, nor the levels of NSE and S100B in individuals with MCI. CONCLUSION The t-PBM increases cognitive function and BDNF levels in adults with MCI. Its application as an adjunctive treatment may play a crucial role in preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Bruna H de Oliveira
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil; Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Elisa F Lins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil; Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Naiara F Kunde
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | | | - Leidiane M Martins
- Department of morphological sciences, Federal University Santa Catarina, Florianópolis, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil; Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Willians F Vieira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil; Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Paolo Cassano
- Photobiomodulation, Division of Neuropsychiatry, Depression Clinical and Research Program Center for Anxiety and Traumatic Stress Disorders Associate Professor Harvard Medical School, Massachusetts General, Boston, USA
| | - Anna Quialheiro
- The Artificial Intelligence and Health Research Unit, Polytechnic University of Health, CESPU, Portugal
| | - Daniel F Martins
- Experimental Neuroscience Laboratory (LaNEx), University of South Santa Catarina, Palhoça, Santa Catarina, Brazil; Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, Santa Catarina, Brazil.
| |
Collapse
|
20
|
Milos T, Vuic B, Balic N, Farkas V, Nedic Erjavec G, Svob Strac D, Nikolac Perkovic M, Pivac N. Cerebrospinal fluid in the differential diagnosis of Alzheimer's disease: an update of the literature. Expert Rev Neurother 2024; 24:1063-1079. [PMID: 39233323 DOI: 10.1080/14737175.2024.2400683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION The importance of cerebrospinal fluid (CSF) biomarkers in Alzheimer's disease (AD) diagnosis is rapidly increasing, and there is a growing interest in the use of CSF biomarkers in monitoring the response to therapy, especially in the light of newly available approaches to the therapy of neurodegenerative diseases. AREAS COVERED In this review we discuss the most relevant measures of neurodegeneration that are being used to distinguish patients with AD from healthy controls and individuals with mild cognitive impairment, in order to provide an overview of the latest information available in the scientific literature. We focus on markers related to amyloid processing, markers associated with neurofibrillary tangles, neuroinflammation, neuroaxonal injury and degeneration, synaptic loss and dysfunction, and markers of α-synuclein pathology. EXPERT OPINION In addition to neuropsychological evaluation, core CSF biomarkers (Aβ42, t-tau, and p-tau181) have been recommended for improvement of timely, accurate and differential diagnosis of AD, as well as to assess the risk and rate of disease progression. In addition to the core CSF biomarkers, various other markers related to synaptic dysfunction, neuroinflammation, and glial activation (neurogranin, SNAP-25, Nfl, YKL-40, TREM2) are now investigated and have yet to be validated for future potential clinical use in AD diagnosis.
Collapse
Affiliation(s)
- Tina Milos
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Barbara Vuic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Nikola Balic
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | - Vladimir Farkas
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
| | | | | | | | - Nela Pivac
- Division of Molecular Medicine, Ruder Boskovic Institute, Zagreb, Croatia
- University of Applied Sciences Hrvatsko Zagorje Krapina, Krapina, Croatia
| |
Collapse
|
21
|
Choi JH, Lee J, Kang U, Chang H, Cho KH. Network dynamics-based subtyping of Alzheimer's disease with microglial genetic risk factors. Alzheimers Res Ther 2024; 16:229. [PMID: 39415193 PMCID: PMC11481771 DOI: 10.1186/s13195-024-01583-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The potential of microglia as a target for Alzheimer's disease (AD) treatment is promising, yet the clinical and pathological diversity within microglia, driven by genetic factors, poses a significant challenge. Subtyping AD is imperative to enable precise and effective treatment strategies. However, existing subtyping methods fail to comprehensively address the intricate complexities of AD pathogenesis, particularly concerning genetic risk factors. To address this gap, we have employed systems biology approaches for AD subtyping and identified potential therapeutic targets. METHODS We constructed patient-specific microglial molecular regulatory network models by utilizing existing literature and single-cell RNA sequencing data. The combination of large-scale computer simulations and dynamic network analysis enabled us to subtype AD patients according to their distinct molecular regulatory mechanisms. For each identified subtype, we suggested optimal targets for effective AD treatment. RESULTS To investigate heterogeneity in AD and identify potential therapeutic targets, we constructed a microglia molecular regulatory network model. The network model incorporated 20 known risk factors and crucial signaling pathways associated with microglial functionality, such as inflammation, anti-inflammation, phagocytosis, and autophagy. Probabilistic simulations with patient-specific genomic data and subsequent dynamics analysis revealed nine distinct AD subtypes characterized by core feedback mechanisms involving SPI1, CASS4, and MEF2C. Moreover, we identified PICALM, MEF2C, and LAT2 as common therapeutic targets among several subtypes. Furthermore, we clarified the reasons for the previous contradictory experimental results that suggested both the activation and inhibition of AKT or INPP5D could activate AD through dynamic analysis. This highlights the multifaceted nature of microglial network regulation. CONCLUSIONS These results offer a means to classify AD patients by their genetic risk factors, clarify inconsistent experimental findings, and advance the development of treatments tailored to individual genotypes for AD.
Collapse
Affiliation(s)
- Jae Hyuk Choi
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jonghoon Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Uiryong Kang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hongjun Chang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
22
|
Cao C, Fu G, Xu R, Li N. Coupling of Alzheimer's Disease Genetic Risk Factors with Viral Susceptibility and Inflammation. Aging Dis 2024; 15:2028-2050. [PMID: 37962454 PMCID: PMC11346407 DOI: 10.14336/ad.2023.1017] [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: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by persistent cognitive decline. Amyloid plaque deposition and neurofibrillary tangles are the main pathological features of AD brain, though mechanisms leading to the formation of lesions remain to be understood. Genetic efforts through genome-wide association studies (GWAS) have identified dozens of risk genes influencing the pathogenesis and progression of AD, some of which have been revealed in close association with increased viral susceptibilities and abnormal inflammatory responses in AD patients. In the present study, we try to present a list of AD candidate genes that have been shown to affect viral infection and inflammatory responses. Understanding of how AD susceptibility genes interact with the viral life cycle and potential inflammatory pathways would provide possible therapeutic targets for both AD and infectious diseases.
Collapse
Affiliation(s)
| | | | - Ruodan Xu
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Ning Li
- Department of Biomedical Engineering and Technology, Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
23
|
Wang Q, Chen S, Wang J, Shang H, Chen X. Advancements in Pharmacological Treatment of Alzheimer's Disease: The Advent of Disease-Modifying Therapies (DMTs). Brain Sci 2024; 14:990. [PMID: 39452004 PMCID: PMC11506318 DOI: 10.3390/brainsci14100990] [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: 09/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
The landscape of pharmacological treatment for Alzheimer's disease (AD) has undergone significant transformations with the advent of disease-modifying therapies (DMTs) targeting β-Amyloid (Aβ) accumulation, one of the hallmark pathologies of AD. The approval and market introduction of monoclonal antibodies mark the dawn of a new era in AD therapeutics as well. Furthermore, considerable progress has also been made in the development of new drugs targeting non-Aβ and non-Tau protein pathways. These advancements are key in tackling the root causes of AD, offering hope for treatments that both relieve symptoms and slow disease progression, improving patient outcomes and quality of life. This review aims to provide a comprehensive update on the advances in drug development and application for AD, including those currently in clinical trials and those already approved for the market to treat patients.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Sihui Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China; (S.C.); (H.S.)
| | - Junhui Wang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada;
- Thyropathy Hospital, Sunsimiao Hospital, Beijing University of Chinese Medicine, Tongchuan 727000, China
| | - Huifang Shang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China; (S.C.); (H.S.)
| | - Xueping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China; (S.C.); (H.S.)
| |
Collapse
|
24
|
Liguori M, Bianco A, Introna A, Consiglio A, Milella G, Abbatangelo E, D'Errico E, Licciulli F, Grillo G, Simone IL. An early Transcriptomic Investigation in Adult Patients with Spinal Muscular Atrophy Under Treatment with Nusinersen. J Mol Neurosci 2024; 74:89. [PMID: 39325116 PMCID: PMC11427494 DOI: 10.1007/s12031-024-02251-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/17/2024] [Indexed: 09/27/2024]
Abstract
Spinal muscular atrophy (SMA) is a rare degenerative disorder with loss of motor neurons caused by mutations in the SMN1 gene. Nusinersen, an antisense oligonucleotide, was approved for SMA treatment to compensate the deficit of the encoded protein SMN by modulating the pre-mRNA splicing of SMN2, the centromeric homologous of SMN1, thus inducing the production of a greater amount of biologically active protein. Here, we reported a 10-month transcriptomics investigation in 10 adult SMA who received nusinersen to search for early genetic markers for clinical monitoring. By comparing their profiles with age-matched healthy controls (HC), we also analyzed the changes in miRNA/mRNAs expression and miRNA-target gene interactions possibly associated with SMA. A multidisciplinary approach of HT-NGS followed by bioinformatics/biostatistics analysis was applied. Within the study interval, those SMA patients who showed some clinical improvements were characterized by having the SMN2/SMN1 ratio slightly increased over the time, while in the stable ones the ratio decreased, suggesting that the estimation of SMN2/SMN1 expression may be an early indicator of nusinersen efficacy. On the other hand, the expression of 38/147 genes/genetic regions DE at T0 between SMA and HC like TRADD and JUND resulted "restored" at T10. We also confirmed the dysregulation of miR-146a(-5p), miR-324-5p and miR-423-5p in SMA subjects. Of interest, miR-146a-5p targeted SMN1, in line with experimental evidence showing the key role of astrocyte-produced miR-146a in SMA motor neuron loss. Molecular pathways such as NOTCH, NF-kappa B, and Toll-like receptor signalings seem to be involved in the SMA pathogenesis.
Collapse
Grants
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- D.U.P. n.246/2019, D.D. n. 3 of 13 January 2021 Apulian Regional Council
- Consiglio Nazionale Delle Ricerche (CNR)
Collapse
Affiliation(s)
- Maria Liguori
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy.
| | - Annalisa Bianco
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy
| | - Alessandro Introna
- Neurology Unit, Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Arianna Consiglio
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy
| | - Giammarco Milella
- Neurology Unit, Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Elena Abbatangelo
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy
| | - Eustachio D'Errico
- Neurology Unit, Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Flavio Licciulli
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy
| | - Giorgio Grillo
- National Research Council, Department of Biomedicine, Institute of Biomedical Technologies - Bari Unit, 70125, Bari, Italy
| | | |
Collapse
|
25
|
Zhou X, Wang S, Yu D, Niu T. Investigating CR1 as an indicated Gene for mild cognitive impairment in type 2 diabetes mellitus. Diabetol Metab Syndr 2024; 16:206. [PMID: 39182129 PMCID: PMC11344402 DOI: 10.1186/s13098-024-01449-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) has beenis known as an important risk factor for cognitive impairment. Meanwhile, the liver plays a central role in the development of T2DM and insulin resistance. The present study attempted to identify and validate marker genes for mild cognitive impairment (MCI) in patients with T2DM. METHODS In this study, insulin resistance-related differentially expressed genes were identified from the liver tissues of individuals with T2DM and those with normal glucose tolerance using the Gene Expression Omnibus database and MCI-associated genes were identified using the GeneCards database. Next, enrichment analysis was performed with overlapping T2DM and MCI genes, followed by the identification of specific genes using the LASSO logistic regression and SVM-RFE algorithms. An important experiment involved the implementation of clinical and in vitro validation using real-time quantitative polymerase chain reaction (RT-qPCR). Finally, multiple linear regression, binary logistic regression, and receiver operating characteristic curve analyses were performed to investigate the relationship between the key gene and cognitive function in these patients. RESULT The present study identified 40 overlapping genes between MCI and T2DM, with subsequent enrichment analysis revealing their significant association with the roles of neuronal and glial projections. The marker gene complement receptor 1(CR1) was identified for both diseases using two regression algorithms. Based on RT-qPCR validation in 65 T2DM patients with MCI (MCI group) and 65 T2DM patients without MCI (NC group), a significant upregulation of CR1 mRNA in peripheral blood mononuclear cells was observed in the MCI group (P < 0.001). Furthermore, the CR1 gene level was significantly negatively associated with MoCA and MMSE scores, which reflect the overall cognitive function, and positively correlated with TMTB scores, which indicate the executive function. Finally, elevated CR1 mRNA levels were identified as an independent risk factor for MCI (OR = 1.481, P < 0.001). CONCLUSION These findings suggest that CR1 is an important predictor of MCI in patients with T2DM. Thus, CR1 has potential clinical significance, which may offer new ideas and directions for the management and treatment of T2DM. The identification and clinical validation of dysregulated marker genes in both T2DM and MCI can offer valuable insights into the intrinsic association between these two conditions. The current study insights may inspire the development of novel strategies for addressing the complicated issues related to cognitive impairment associated with diabetes.
Collapse
Affiliation(s)
- Xueling Zhou
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Shaohua Wang
- School of Medicine, Southeast University, Nanjing, China.
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| | - Dandan Yu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Tong Niu
- School of Medicine, Southeast University, Nanjing, China
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
26
|
Basile S, Parisi C, Bellia F, Zimbone S, Arrabito G, Gulli D, Pignataro B, Giuffrida ML, Sortino S, Copani A. Red-Light-Photosensitized Tyrosine 10 Nitration of β-Amyloid 1-42 Diverts the Protein from Forming Toxic Aggregates. ACS Chem Neurosci 2024; 15:2916-2924. [PMID: 39036818 DOI: 10.1021/acschemneuro.4c00284] [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: 07/23/2024] Open
Abstract
Several studies have highlighted the presence of nitration damage following neuroinflammation in Alzheimer's disease (AD). Accordingly, post-transcriptional modifications of β-amyloid (Aβ), including peptide nitration, have been explored as a marker of the disease. However, the implications of Aβ nitration in terms of aggregation propensity and neurotoxicity are still debated. Here, we show new data obtained using a photoactivatable peroxynitrite generator (BPT-NO) to overcome the limitations associated with chemical nitration methods. We found that the photoactivation of BPT-NO with the highly biocompatible red light selectively induces the nitration of tyrosine 10 of freshly solubilized full-length Aβ1-42. Photonitrated Aβ1-42 was, therefore, investigated for aggregation states and functions. It resulted that photonitrated Aβ1-42 did not aggregate into small oligomers but rather self-assembled into large amorphous aggregates. When tested on neuronal-like SH-SY5Y cells and microglial C57BL/6 BV2 cells, photonitrated Aβ1-42 showed to be free of neurotoxicity and able to induce phagocytic microglia cells. We propose that light-controlled nitration of the multiple forms in which Aβ occurs (i.e., monomers, oligomers, fibrils) could be a tool to assess in real-time the impact of tyrosine nitration on the amyloidogenic and toxic properties of Aβ1-42.
Collapse
Affiliation(s)
- Sarah Basile
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Cristina Parisi
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Francesco Bellia
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| | - Stefania Zimbone
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| | - Giuseppe Arrabito
- Department of Physics and Chemistry (DiFC) Emilio Segrè, University of Palermo, 90128 Palermo, Italy
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | - Daniele Gulli
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | - Bruno Pignataro
- Department of Physics and Chemistry (DiFC) Emilio Segrè, University of Palermo, 90128 Palermo, Italy
- ATeN Center, University of Palermo, 90128 Palermo, Italy
| | | | - Salvatore Sortino
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
| | - Agata Copani
- Department of Drug and Health Sciences (DSFS), University of Catania, 95125 Catania, Italy
- Institute of Crystallography (IC), National Research Council, 95126 Catania, Italy
| |
Collapse
|
27
|
Lista S, Imbimbo BP, Grasso M, Fidilio A, Emanuele E, Minoretti P, López-Ortiz S, Martín-Hernández J, Gabelle A, Caruso G, Malaguti M, Melchiorri D, Santos-Lozano A, Imbimbo C, Heneka MT, Caraci F. Tracking neuroinflammatory biomarkers in Alzheimer's disease: a strategy for individualized therapeutic approaches? J Neuroinflammation 2024; 21:187. [PMID: 39080712 PMCID: PMC11289964 DOI: 10.1186/s12974-024-03163-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Recent trials of anti-amyloid-β (Aβ) monoclonal antibodies, including lecanemab and donanemab, in early Alzheimer disease (AD) showed that these drugs have limited clinical benefits and their use comes with a significant risk of serious adverse events. Thus, it seems crucial to explore complementary therapeutic approaches. Genome-wide association studies identified robust associations between AD and several AD risk genes related to immune response, including but not restricted to CD33 and TREM2. Here, we critically reviewed the current knowledge on candidate neuroinflammatory biomarkers and their role in characterizing the pathophysiology of AD. MAIN BODY Neuroinflammation is recognized to be a crucial and contributing component of AD pathogenesis. The fact that neuroinflammation is most likely present from earliest pre-stages of AD and co-occurs with the deposition of Aβ reinforces the need to precisely define the sequence and nature of neuroinflammatory events. Numerous clinical trials involving anti-inflammatory drugs previously yielded unfavorable outcomes in early and mild-to-moderate AD. Although the reasons behind these failures remain unclear, these may include the time and the target selected for intervention. Indeed, in our review, we observed a stage-dependent neuroinflammatory process in the AD brain. While the initial activation of glial cells counteracts early brain Aβ deposition, the downregulation in the functional state of microglia occurs at more advanced disease stages. To address this issue, personalized neuroinflammatory modulation therapy is required. The emergence of reliable blood-based neuroinflammatory biomarkers, particularly glial fibrillary acidic protein, a marker of reactive astrocytes, may facilitate the classification of AD patients based on the ATI(N) biomarker framework. This expands upon the traditional classification of Aβ ("A"), tau ("T"), and neurodegeneration ("N"), by incorporating a novel inflammatory component ("I"). CONCLUSIONS The present review outlines the current knowledge on potential neuroinflammatory biomarkers and, importantly, emphasizes the role of longitudinal analyses, which are needed to accurately monitor the dynamics of cerebral inflammation. Such a precise information on time and place will be required before anti-inflammatory therapeutic interventions can be considered for clinical evaluation. We propose that an effective anti-neuroinflammatory therapy should specifically target microglia and astrocytes, while considering the individual ATI(N) status of patients.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, 43122, Parma, Italy
| | | | | | | | | | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
| | - Audrey Gabelle
- CMRR, Memory Resources and Research Center, Montpellier University of Excellence i-site, 34295, Montpellier, France
| | - Giuseppe Caruso
- Oasi Research Institute-IRCCS, 94018, Troina, Italy
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 40126, Bologna, Italy
| | - Daniela Melchiorri
- Department of Physiology and Pharmacology, Sapienza University, 00185, Rome, Italy
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012, Valladolid, Spain
- Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital, 12 de Octubre ('imas12'), 28041, Madrid, Spain
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100, Pavia, Italy
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367, Esch-Belval, Luxembourg.
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, 94018, Troina, Italy.
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
28
|
Zabot GC, Medeiros EB, Macarini BMN, Peruchi BB, Keller GS, Lídio AV, Boaventura A, de Jesus LC, de Bem Silveira G, Silveira PCL, Chede BC, Réus GZ, Budni J. The involvement of neuroinflammation in an animal model of dementia and depression. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:110999. [PMID: 38552774 DOI: 10.1016/j.pnpbp.2024.110999] [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: 07/14/2023] [Revised: 03/26/2024] [Accepted: 03/26/2024] [Indexed: 04/21/2024]
Abstract
Alzheimer's disease (AD) and depression are inflammatory pathologies, leading to increased inflammatory response and neurotoxicity. Therefore, this study aimed to evaluate the effect of the treatment with fluoxetine and/or galantamine and/or donepezil on the levels of proinflammatory and anti-inflammatory cytokines in a mixed animal model of depression and dementia. Adult male Wistar rats underwent chronic mild stress (CMS) protocol for 40 days and were subjected to stereotaxic surgery for intra-hippocampal administration of amyloid-beta (Aꞵ) peptide or artificial cerebrospinal fluid (ACSF) to mimic the dementia animal model. On the 42nd day, animals were treated with water, galantamine, donepezil, and/or fluoxetine, orally for 17 days. On the 57th and 58th days, the Splash and Y-maze tests for behavior analysis were performed. The frontal cortex and hippocampus were used to analyze the tumor necrosis factor alfa (TNF-α), interleukin 1 beta (IL-1ꞵ), IL-6, and IL-10 levels. The results of this study show that animals subjected to CMS and administration of Aꞵ had anhedonia, cognitive impairment, increased TNF-α and IL-1ꞵ levels in the frontal cortex, and reduced IL-10 levels in the hippocampus. All treatment groups were able to reverse the cognitive impairment. Only donepezil did not decrease the TNF-α levels in the hippocampus. Fluoxetine + galantamine and fluoxetine + donepezil reversed the anhedonia. Fluoxetine reversed the anhedonia and IL-1ꞵ levels in the frontal cortex. In addition, fluoxetine + donepezil reversed the reduction of IL-10 levels in the hippocampus. The results indicate a pathophysiological interaction between AD and depression, and the association of medications in the future may be a possible therapeutic strategy to reduce inflammation, especially the fluoxetine-associated treatments.
Collapse
Affiliation(s)
- Gabriel Casagrande Zabot
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Eduarda Behenck Medeiros
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Bárbara Machado Naspolini Macarini
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Bruno Búrigo Peruchi
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gabriela Serafim Keller
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Adrielly Vargas Lídio
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Amanda Boaventura
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Laura Ceolin de Jesus
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Physiopathology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Beatriz Costa Chede
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Gislaine Zilli Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil
| | - Josiane Budni
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences (PPGCS), University of Southern Santa Catarina (UNESC), Criciúma, Brazil.
| |
Collapse
|
29
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Singh A, Ansari VA, Mahmood T, Hasan SM, Wasim R, Maheshwari S, Akhtar J, Sheikh S, Vishwakarma VK. Targeting Abnormal Tau Phosphorylation for Alzheimer's Therapeutics. Horm Metab Res 2024; 56:482-488. [PMID: 38350636 DOI: 10.1055/a-2238-1384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative disorder characterized by progressive memory and cognitive decline, posing a formidable public health challenge. This review explores the intricate interplay between two pivotal players in AD pathogenesis: β-amyloid (Aβ) and tau protein. While the amyloid cascade theory has long dominated AD research, recent developments have ignited debates about its centrality. Aβ plaques and tau NFTs are hallmark pathologies in AD. Aducanumab and lecanemab, monoclonal antibodies targeting Aβ, have been approved, albeit amidst controversy, raising questions about the therapeutic efficacy of Aβ-focused interventions. On the other hand, tau, specifically its hyperphosphorylation, disrupts microtubule stability and contributes to neuronal dysfunction. Various post-translational modifications of tau drive its aggregation into NFTs. Emerging treatments targeting tau, such as GSK-3β and CDK5 inhibitors, have shown promise in preclinical and clinical studies. Restoring the equilibrium between protein kinases and phosphatases, notably protein phosphatase-2A (PP2A), is a promising avenue for AD therapy, as tau is primarily regulated by its phosphorylation state. Activation of tau-specific phosphatases offers potential for mitigating tau pathology. The evolving landscape of AD drug development emphasizes tau-centric therapies and reevaluation of the amyloid cascade hypothesis. Additionally, exploring the role of neuroinflammation and its interaction with tau pathology present promising research directions.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | | | | | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Suvaiv Sheikh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | |
Collapse
|
31
|
Chi J, Hu J, Wu N, Cai H, Lin C, Lai Y, Huang J, Li W, Su P, Li M, Xu L. Causal effects for neurodegenerative diseases on the risk of myocardial infarction: a two-sample Mendelian randomization study. Aging (Albany NY) 2024; 16:9944-9958. [PMID: 38850523 PMCID: PMC11210233 DOI: 10.18632/aging.205909] [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: 11/22/2023] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
Several studies have demonstrated a correlation between neurodegenerative diseases (NDDs) and myocardial infarction (MI), yet the precise causal relationship between these remains elusive. This study aimed to investigate the potential causal associations of genetically predicted Alzheimer's disease (AD), dementia with Lewy bodies (DLB), Parkinson's disease (PD), and multiple sclerosis (MS) with MI using two-sample Mendelian randomization (TSMR). Various methods, including inverse variance weighted (IVW), weighted median (WM), MR-Egger regression, weighted mode, and simple mode, were employed to estimate the effects of genetically predicted NDDs on MI. To validate the analysis, we assessed pleiotropic effects, heterogeneity, and conducted leave-one-out sensitivity analysis. We identified that genetic predisposition to NDDs was suggestively associated with higher odds of MI (OR_IVW=1.07, OR_MR-Egger=1.08, OR_WM=1.07, OR_weighted mode=1.07, OR_simple mode=1.10, all P<0.05). Furthermore, we observed significant associations of genetically predicted DLB with MI (OR_IVW=1.07, OR_MR-Egger=1.11, OR_WM=1.09, OR_weighted mode=1.09, all P<0.05). However, there was no significant causal evidence of genetically predicted PD and MS in MI. Across all MR analyses, no horizontal pleiotropy or statistical heterogeneity was observed (all P>0.05). Additionally, results from MRPRESSO and leave-one-out sensitivity analysis confirmed the robustness of the causal effect estimations for genetically predicted AD, DLB, PD, and MS on MI. This study provides further support for the causal effects of AD on MI and, for the first time, establishes robust causal evidence for the detrimental effect of DLB on the risk of MI. Our findings emphasize the importance of monitoring the cardiovascular function of the elderly experiencing neurodegenerative changes.
Collapse
Affiliation(s)
- Jianing Chi
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaman Hu
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ningxia Wu
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hua Cai
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cailong Lin
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yingying Lai
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianyu Huang
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
| | - Weihua Li
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Peng Su
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Min Li
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
| | - Lin Xu
- Department of Geriatric Cardiology, General Hospital of Southern Theater Command, Guangzhou, China
- Branch of National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Guangzhou, China
- Guangzhou Key Laboratory of Cardiac Rehabilitation, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Pereira JD, Teixeira LCR, Mamede I, Alves MT, Caramelli P, Luizon MR, Veloso AA, Gomes KB. miRNAs in cerebrospinal fluid associated with Alzheimer's disease: A systematic review and pathway analysis using a data mining and machine learning approach. J Neurochem 2024; 168:977-994. [PMID: 38390627 DOI: 10.1111/jnc.16060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/24/2024]
Abstract
Alzheimer's disease (AD) is the most common type and accounts for 60%-70% of the reported cases of dementia. MicroRNAs (miRNAs) are small non-coding RNAs that play a crucial role in gene expression regulation. Although the diagnosis of AD is primarily clinical, several miRNAs have been associated with AD and considered as potential markers for diagnosis and progression of AD. We sought to match AD-related miRNAs in cerebrospinal fluid (CSF) found in the GeoDataSets, evaluated by machine learning, with miRNAs listed in a systematic review, and a pathway analysis. Using machine learning approaches, we identified most differentially expressed miRNAs in Gene Expression Omnibus (GEO), which were validated by the systematic review, using the acronym PECO-Population (P): Patients with AD, Exposure (E): expression of miRNAs, Comparison (C): Healthy individuals, and Objective (O): miRNAs differentially expressed in CSF. Additionally, pathway enrichment analysis was performed to identify the main pathways involving at least four miRNAs selected. Four miRNAs were identified for differentiating between patients with and without AD in machine learning combined to systematic review, and followed the pathways analysis: miRNA-30a-3p, miRNA-193a-5p, miRNA-143-3p, miRNA-145-5p. The pathways epidermal growth factor, MAPK, TGF-beta and ATM-dependent DNA damage response, were regulated by these miRNAs, but only the MAPK pathway presented higher relevance after a randomic pathway analysis. These findings have the potential to assist in the development of diagnostic tests for AD using miRNAs as biomarkers, as well as provide understanding of the relationship between different pathophysiological mechanisms of AD.
Collapse
Affiliation(s)
- Jessica Diniz Pereira
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Izabela Mamede
- Intituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Paulo Caramelli
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Rizzatti Luizon
- Intituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adriano Alonso Veloso
- Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
33
|
Shamim T, Asif HM, Abida Ejaz S, Hussain Z, Wani TA, Sumreen L, Abdullah M, Ahmed Z, Iqbal J, Kim SJ, Shah MK. Investigations of Limeum Indicum Plant for Diabetes Mellitus and Alzheimer's Disease Dual Therapy: Phytochemical, GC-MS Chemical Profiling, Enzyme Inhibition, Molecular Docking and In-Vivo Studies. Chem Biodivers 2024; 21:e202301858. [PMID: 38608202 DOI: 10.1002/cbdv.202301858] [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: 11/21/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/14/2024]
Abstract
Limeum indicum has been widely utilized in traditional medicine but no experimental work has been done on this herb. The primary objective of this study was to conduct a phytochemical analysis and assess the multifunctional capabilities of aforementioned plant in dual therapy for Alzheimer's disease (AD) and Type 2 diabetes (T2D). The phytochemical screening of ethanol, methanol extract, and their derived fractions of Limeum indicum was conducted using GC-MS, HPLC, UV-analysis and FTIR. The antioxidant capacity was evaluated by DPPH method. The inhibitory potential of the extracts/fractions against α-, β-glucosidase acetylcholinesterase (AChE), butyrylcholinesterase (BChE) and monoaminine oxidases (MAO-A & B) was evaluated. Results revealed that acetonitrile fraction has highest inhibitory potential against α-glucosidase (IC50=68.47±0.05 μg/mL), methanol extract against β-glucosidase (IC50=91.12±0.07 μg/mL), ethyl acetate fraction against AChE (IC50=59.0±0.02 μg/mL), ethanol extract against BChE (28.41±0.01 μg/mL), n-hexane fraction against MAO-A (IC50=150.5±0.31 μg/mL) and methanol extract for MAO-B (IC50=75.95±0.13 μg/mL). The docking analysis of extracts\fractions suggested the best binding scores within the active pocket of the respective enzymes. During the in-vivo investigation, ethanol extract produced hypoglycemic effect (134.52±2.79 and 119.38±1.40 mg/dl) after 21 days treatment at dose level of 250 and 500 mg/Kg. Histopathological findings further supported the in-vivo studies.
Collapse
Affiliation(s)
- Tahira Shamim
- University College of Conventional Medicine, Faculty of Medicine & Allied Health Sciences, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Hafiz Muhammad Asif
- University College of Conventional Medicine, Faculty of Medicine & Allied Health Sciences, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Zahid Hussain
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
- Center for Advance Drug Research, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Tanveer A Wani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O.Box 2452, 11451, Riyadh, Saudi Arabia
| | - Laila Sumreen
- University College of Conventional Medicine, Faculty of Medicine & Allied Health Sciences, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Muhammad Abdullah
- Cholistan Institute of Desert Studies, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Zubair Ahmed
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Jamshed Iqbal
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
- Center for Advance Drug Research, COMSATS University Islamabad, Abbottabad Campus, 22060, Abbottabad, Pakistan
| | - Song Ja Kim
- College of Natural Sciences, Department of Biological Sciences, Kongju National University, 32588, Gongju, South Korea
| | - Muhammad Kamal Shah
- Faculty of Veterinary and Animal Sciences, Gomal University, 29220, Dera Ismail Khan, Pakistan
| |
Collapse
|
34
|
Yu WY, Sun TH, Hsu KC, Wang CC, Chien SY, Tsai CH, Yang YW. Comparative analysis of machine learning algorithms for Alzheimer's disease classification using EEG signals and genetic information. Comput Biol Med 2024; 176:108621. [PMID: 38763067 DOI: 10.1016/j.compbiomed.2024.108621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairments, and behavioral changes. The presence of abnormal beta-amyloid plaques and tau protein tangles in the brain is known to be associated with AD. However, current limitations of imaging technology hinder the direct detection of these substances. Consequently, researchers are exploring alternative approaches, such as indirect assessments involving monitoring brain signals, cognitive decline levels, and blood biomarkers. Recent studies have highlighted the potential of integrating genetic information into these approaches to enhance early detection and diagnosis, offering a more comprehensive understanding of AD pathology beyond the constraints of existing imaging methods. Our study utilized electroencephalography (EEG) signals, genotypes, and polygenic risk scores (PRSs) as features for machine learning models. We compared the performance of gradient boosting (XGB), random forest (RF), and support vector machine (SVM) to determine the optimal model. Statistical analysis revealed significant correlations between EEG signals and clinical manifestations, demonstrating the ability to distinguish the complexity of AD from other diseases by using genetic information. By integrating EEG with genetic data in an SVM model, we achieved exceptional classification performance, with an accuracy of 0.920 and an area under the curve of 0.916. This study presents a novel approach of utilizing real-time EEG data and genetic background information for multimodal machine learning. The experimental results validate the effectiveness of this concept, providing deeper insights into the actual condition of patients with AD and overcoming the limitations associated with single-oriented data.
Collapse
Affiliation(s)
- Wei-Yang Yu
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Ting-Hsuan Sun
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Kai-Cheng Hsu
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan; Department of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Chun Wang
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Shang-Yu Chien
- Artificial Intelligence Center, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Chon-Haw Tsai
- Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan; Neuroscience Laboratory, Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan; Neuroscience and Brain Disease Center, College of Medicine, China Medical University, 40402, Taichung, Taiwan
| | - Yu-Wan Yang
- Department of Neurology, China Medical University Hospital, Taichung, 40447, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
35
|
Wang W, Wang Y, Wang F, Xie G, Liu S, Li Z, Wang P, Liu J, Lin L. Gastrodin regulates the TLR4/TRAF6/NF-κB pathway to reduce neuroinflammation and microglial activation in an AD model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155518. [PMID: 38552431 DOI: 10.1016/j.phymed.2024.155518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/25/2024] [Accepted: 03/07/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Gastrodia elata (Orchidaceae) is a medicinal plant used in traditional Chinese medicine. The rhizomes contain numerous active components, of which Gastrodin (p-hydroxymethylphenyl-B-D-glucopyranoside) forms the basis of the traditional medicine Gastrodiae Rhizoma. Gastrodin is also found in other medicinal plants and has neuroprotective, antioxidant, and anti-inflammatory effects. Neuroinflammation plays a crucial role in neurodegeneration. Research indicates that consuming meals and drinks containing Gastrodiaelata can enhance cognitive functioning and memory in elderly patients. The mechanisms relevant to the problem have not been completely understood. PURPOSE The aim was to examine the in vivo and in vitro anti-neuroinflammatory effects of Gastrodin. STUDY DESIGN The neuroprotective effects of Gastrodin on the TLR4/TRAF6/NF-κB pathway and Stat3 phosphorylation in LPS-treated C57BL/6 mice and BV-2 cells were investigated. METHODS 1. C57BL/6 mice were assigned to model, gastrodin, donepezil, and control groups (n = 10 per group). The Gastrodin group received 100 mg/kg/d for five days, and the Dopenezil group 1.3 mg/kg/d. A neuroinflammation model was established by administering intraperitoneal injections of 2 mg/kg LPS to all groups, excluding the control. To induce microglial activation in Gastrodin-treated mouse microglial BV-2 cells, 1 µg/ml LPS was introduced for 24 h Morris water mazes were utilized to evaluate learning and spatial memory. Expression and subcellular localization of TLR4/TRAF6/NF-κB axis-related proteins and p-Stat3, Iba-1, GFAP, iNOS, and CD206 were assessed by immunofluorescence, western blots, and ELISA. qRT-PCR was performed to determine and measure IL-1β, TNF-α, cell migration, and phagocytosis. Overexpression of TRAF6 was induced by transfection, and the effect of Gastrodin on IL-1β and p-NF-κB p65 levels was assessed. RESULTS 1. In mice, gastrodin treatment mitigated LPS-induced deficits in learning and spatial memory, as well as reducing neuroinflammation in the hippocampus, expression of TLR4/TRAF6/NF-κB pathway proteins, activation of microglia and astrocytes, and phosphorylation of Stat3. 2. Gastrodin pretreatment improved LPS-induced inflammation in vitro, reducing expression of TLR4/TRAF6/NF-κB-associated proteins and p-Stat3, inducing microglial transformation from M1 to M2, and inhibiting migration and phagocytosis. Overexpression of TRAF6 inhibited the Gastrodin-induced effects. CONCLUSION Gastrodin suppresses neuroinflammation and microglial activation by modifying the TLR4/TRAF6/NF-κB pathway and Stat3 phosphorylation.
Collapse
Affiliation(s)
- Wensheng Wang
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China; Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Yu Wang
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China
| | - Fengjie Wang
- Department of Medicine, Hubei Minzu University, Enshi 445000, China
| | - Guangjing Xie
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Shangzhi Liu
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Zefei Li
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China
| | - Ping Wang
- Hubei Research Institute of Geriatrics, Collaborative Innovation Center of Hubei Province, Hubei University of Chinese Medicine, No. 16, Huangjiahu West Road, Hongshan District, Wuhan 430065, China.
| | - Junfeng Liu
- Key Laboratory of TCM Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China.
| | - Li Lin
- Laboratory of Medical Molecular and Cellular Biology, College of Basic Medical Sciences, Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, No.16 of Huangjia Lake Western Road, Hong Shan District, Wuhan 430065, China.
| |
Collapse
|
36
|
Siafarikas N. Personalized medicine in old age psychiatry and Alzheimer's disease. Front Psychiatry 2024; 15:1297798. [PMID: 38751423 PMCID: PMC11094449 DOI: 10.3389/fpsyt.2024.1297798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Elderly patients show us unfolded lives with unique individual characteristics. An increasing life span is associated with increasing physical and mental disease burden. Alzheimer's disease (AD) is an increasing challenge in old age. AD cannot be cured but it can be treated. The complexity of old age and AD offer targets for personalized medicine (PM). Targets for stratification of patients, detection of patients at risk for AD or for future targeted therapy are plentiful and can be found in several omic-levels.
Collapse
Affiliation(s)
- Nikias Siafarikas
- Department of Geriatric Psychiatry, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
37
|
Yang C, Liu G, Zeng X, Xiang Y, Chen X, Le W. Therapeutic effects of long-term HBOT on Alzheimer's disease neuropathologies and cognitive impairment in APP swe/PS1 dE9 mice. Redox Biol 2024; 70:103006. [PMID: 38241837 PMCID: PMC10831255 DOI: 10.1016/j.redox.2023.103006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 01/21/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder with the pathological hallmarks of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Although there is a hope that anti-amyloid monoclonal antibodies may emerge as a new therapy for AD, the high cost and side effect is a big concern. Non-drug therapy is attracting more attention and may provide a better resolution for the treatment of AD. Given the fact that hypoxia contributes to the pathogenesis of AD, hyperbaric oxygen therapy (HBOT) may be an effective intervention that can alleviate hypoxia and improve AD. However, it remains unclear whether long-term HBOT intervention in the early stage of AD can slow AD progression and ultimately prevent cognitive impairment in this disease. In this study we applied consecutive 3-month HBOT interventions on 3-month-old APPswe/PS1dE9 AD mice which represent the early stage of AD. When the APPswe/PS1dE9 mice at 9-month-old which represent the disease stage we measured cognitive function, 24-h blood oxygen saturation, Aβ and tau pathologies, vascular structure and function, and neuroinflammation in APPswe/PS1dE9 mice. Our results showed that long-term HBOT can attenuate the impairments in cognitive function observed in 9-month-old APPswe/PS1dE9 mice. Most importantly, HBOT effectively reduced the progression of Aβ plaques deposition, hyperphosphorylated tau protein aggregation, and neuronal and synaptic degeneration in the AD mice. Further, long-term HBOT was able to enhance blood oxygen saturation level. Besides, long-term HBOT can improve vascular structure and function, and reduce neuroinflammation in AD mice. This study is the first to demonstrate that long-term HBOT intervention in the early stage of AD can attenuate cognitive impairment and AD-like pathologies. Overall, these findings highlight the potential of long-term HBOT as a disease-modifying approach for AD treatment.
Collapse
Affiliation(s)
- Cui Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Guangdong Liu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xianrong Zeng
- Department of Hyperbaric Oxygen, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Xiang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| |
Collapse
|
38
|
Speers AB, Wright KM, Brandes MS, Kedjejian N, Matthews DG, Caruso M, Harris CJ, Koike S, Nguyen T, Quinn JF, Soumyanath A, Gray NE. Mode of administration influences plasma levels of active Centella asiatica compounds in 5xFAD mice while markers of neuroinflammation remain unaltered. Front Neurosci 2024; 18:1277626. [PMID: 38591068 PMCID: PMC10999680 DOI: 10.3389/fnins.2024.1277626] [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: 08/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction A water extract of Centella asiatica (L.) Urban [Apiaceae] (CAW) has demonstrated cognitive-enhancing effects in mouse models of Alzheimer's disease and aging, the magnitude of which is influenced by whether CAW is delivered in the drinking water or the diet. These cognitive benefits are accompanied by improvements in oxidative stress and mitochondrial function in the brain, two pathways related to the neuroinflammatory response. The effect of CAW on neuroinflammation, however, has not been directly studied. Here, we investigated the effect of CAW on neuroinflammation in 5xFAD mice and compared plasma levels of CAW's active compounds following two modes of CAW administration. Methods Eight-to-nine-month-old male and female 5xFAD mice and their wild-type littermates were administered CAW in their diet or drinking water (0 or 1,000 mg/kg/day) for five weeks. Immunohistochemistry was performed for β-amyloid (Aβ), glial fibrillary acidic protein (GFAP), and Griffonia simplicifolia lectin I (GSL I) in the cortex and hippocampus. Gene expression of inflammatory mediators (IL-6, TNFα, IL-1β, TREM2, AIF1, CX3CR1, CX3CL1, CD36, C3AR1, RAGE, CCR6, CD3E) was measured in the deep grey matter. Results CAW decreased cortical Aβ plaque burden in female 5xFAD mice administered CAW in the drinking water but had no effect on Aβ plaques in other treatment groups. CAW did not impact elevated levels of GFAP or GSL I in 5xFAD mice, regardless of sex, brain region, or mode of CAW administration. In the deep grey matter, CAW increased C3AR1 expression in 5xFAD females administered CAW in the drinking water and decreased IL-1β expression in 5xFAD males administered CAW in the diet. CAW had no effect, however, on gene expression levels of any other inflammatory mediator in the deep grey, for either sex or mode of CAW administration. Mice administered CAW in the drinking water versus the diet had significantly higher plasma levels of CAW compounds. Discussion CAW had little impact on the neuroinflammatory markers selected for evaluation in the present study, suggesting that the cognitive benefits of CAW may not be mediated by an anti-inflammatory effect or that additional molecular markers are needed to fully characterize the effect of CAW on neuroinflammation.
Collapse
Affiliation(s)
- Alex B. Speers
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nareg Kedjejian
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Christopher J. Harris
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
39
|
Royall DR, Palmer RF. INFLAMMATION's cognitive impact revealed by a novel "Line of Identity" approach. PLoS One 2024; 19:e0295386. [PMID: 38517924 PMCID: PMC10959355 DOI: 10.1371/journal.pone.0295386] [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: 03/01/2023] [Accepted: 11/22/2023] [Indexed: 03/24/2024] Open
Abstract
IMPORTANCE Dementia is an "overdetermined" syndrome. Few individuals are demented by any single biomarker, while several may independently explain small fractions of dementia severity. It may be advantageous to identify individuals afflicted by a specific biomarker to guide individualized treatment. OBJECTIVE We aim to validate a psychometric classifier to identify persons adversely impacted by inflammation and replicate it in a second cohort. DESIGN Secondary analyses of data collected by the Texas Alzheimer's Research and Care Consortium (TARCC) (N = 3497) and the Alzheimer's Disease Neuroimaging Initiative (ADNI) (N = 1737). SETTING Two large, well-characterized multi-center convenience samples. PARTICIPANTS Volunteers with normal cognition (NC), Mild Cognitive Impairment (MCI) or clinical "Alzheimer's Disease (AD)". EXPOSURE Participants were assigned to "Afflicted" or "Resilient" classes on the basis of a psychometric classifier derived by confirmatory factor analysis. MAIN OUTCOME(S) AND MEASURE(S) The groups were contrasted on multiple assessments and biomarkers. The groups were also contrasted regarding 4-year prospective conversions to "AD" from non-demented baseline diagnoses (controls and MCI). The Afflicted groups were predicted to have adverse levels of inflammation-related blood-based biomarkers, greater dementia severity and greater risk of prospective conversion. RESULTS In ADNI /plasma, 47.1% of subjects were assigned to the Afflicted class. 44.6% of TARCC's subjects were afflicted, 49.5% of non-Hispanic Whites (NHW) and 37.2% of Mexican Americans (MA). There was greater dementia severity in the Afflicted class [by ANOVA: ADNI /F(1) = 686.99, p <0.001; TARCC /F(1) = 1544.01, p <0.001]. "INFLAMMATION" factor composite scores were significantly higher (adverse) in Afflicted subjects [by ANOVA in ADNI /plasma F(1) = 1642.64, p <0.001 and in TARCC /serum F(1) = 3059.96, p <0.001]. Afflicted cases were more likely to convert to AD in the next four years [by Cox's F, ADNI /plasma: F (252, 268) = 3.74 p < 0.001; TARCC /serum: F (160, 134) = 3.03, p < 0.001 (in TARCC's entire sample), F (110, 90) = 4.92, p <0.001 in NHW, and F(50, 44) = 2.13, p = 0.006 in MA]. The proportions converting were similar among afflicted NHW in both cohorts /biofluids but MA exhibited a lower risk (7% in TARCC /serum at 48 months). CONCLUSIONS AND RELEVANCE Our inflammation-specific psychometric classifier selects individuals with pre-specified biomarker profiles and predicts conversion to "AD" across cohorts, biofluids, and ethnicities. This algorithm might be applied to any dementia-related biomarker making the psychometric estimation of individual biomarker effects feasible without biomarker assessment. Our approach also distinguishes individuals resilient to individual biomarker effects allowing for more accurate prediction and precision intervention.
Collapse
Affiliation(s)
- Donald R. Royall
- Department of Psychiatry and Behavioral Science, The University of Texas Health Science Center, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
- Department of Family and Community Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
- The Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Disease, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Raymond F. Palmer
- Department of Family and Community Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| |
Collapse
|
40
|
Uleman JF, Quax R, Melis RJF, Hoekstra AG, Olde Rikkert MGM. The need for systems thinking to advance Alzheimer's disease research. Psychiatry Res 2024; 333:115741. [PMID: 38277813 DOI: 10.1016/j.psychres.2024.115741] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/08/2023] [Accepted: 01/12/2024] [Indexed: 01/28/2024]
Abstract
Despite extensive research efforts to mechanistically understand late-onset Alzheimer's disease (LOAD) and other complex mental health disorders, curative treatments remain elusive. We emphasize the multiscale multicausality inherent to LOAD, highlighting the interplay between interconnected pathophysiological processes and risk factors. Systems thinking methods, such as causal loop diagrams and systems dynamic models, offer powerful means to capture and study this complexity. Recent studies developed and validated a causal loop diagram and system dynamics model using multiple longitudinal data sets, enabling the simulation of personalized interventions on various modifiable risk factors in LOAD. The results indicate that targeting factors like sleep disturbance and depressive symptoms could be promising and yield synergistic benefits. Furthermore, personalized interventions showed significant potential, with top-ranked intervention strategies differing significantly across individuals. We argue that systems thinking approaches can open new prospects for multifactorial precision medicine. In future research, systems thinking may also guide structured, model-driven data collection on the multiple interactions in LOAD's complex multicausality, facilitating theory development and possibly resulting in effective prevention and treatment options.
Collapse
Affiliation(s)
- Jeroen F Uleman
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark; Department of Geriatric Medicine, Radboudumc Alzheimer Center, Donders Institute for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Rick Quax
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - René J F Melis
- Department of Geriatric Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alfons G Hoekstra
- Computational Science Lab, Informatics Institute, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Marcel G M Olde Rikkert
- Department of Geriatric Medicine, Radboudumc Alzheimer Center, Donders Institute for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
41
|
Pase MP, Himali JJ, Puerta R, Beiser AS, Gonzales MM, Satizabal CL, Yang Q, Aparicio HJ, Kojis DJ, Decarli CS, Lopez OL, Longstreth W, Gudnason V, Mosley TH, Bis JC, Fohner A, Psaty BM, Boada M, García-González P, Valero S, Marquié M, Tracy R, Launer LJ, Ruiz A, Fornage M, Seshadri S. Association of Plasma YKL-40 With MRI, CSF, and Cognitive Markers of Brain Health and Dementia. Neurology 2024; 102:e208075. [PMID: 38290090 PMCID: PMC11383876 DOI: 10.1212/wnl.0000000000208075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Higher YKL-40 levels in the CSF are a known biomarker of brain inflammation. We explored the utility of plasma YKL-40 as a biomarker for accelerated brain aging and dementia risk. METHODS We performed cross-sectional and prospective analyses of 4 community-based cohorts in the United States or Europe: the Age, Gene/Environment Susceptibility-Reykjavik Study, Atherosclerosis Risk in the Communities study, Coronary Artery Risk Development in Young Adults study, and Framingham Heart Study (FHS). YKL-40 was measured from stored plasma by a single laboratory using Mesoscale Discovery with levels log transformed and standardized within each cohort. Outcomes included MRI total brain volume, hippocampal volume, and white matter hyperintensity volume (WMHV) as a percentage of intracranial volume, a general cognitive composite derived from neuropsychological testing (SD units [SDU]), and the risk of incident dementia. We sought to replicate associations with dementia in the clinic-based ACE csf cohort, which also had YKL-40 measured from the CSF. RESULTS Meta-analyses of MRI outcomes included 6,558 dementia-free participants, and for analysis of cognition, 6,670. The blood draw preceded MRI/cognitive assessment by up to 10.6 years across cohorts. The mean ages ranged from 50 to 76 years, with 39%-48% male individuals. In random-effects meta-analysis of study estimates, each SDU increase in log-transformed YKL-40 levels was associated with smaller total brain volume (β = -0.33; 95% CI -0.45 to -0.22; p < 0.0001) and poorer cognition (β = -0.04; 95% CI -0.07 to -0.02; p < 0.01), following adjustments for demographic variables. YKL-40 levels did not associate with hippocampal volume or WMHV. In the FHS, each SDU increase in log YKL-40 levels was associated with a 64% increase in incident dementia risk over a median of 5.8 years of follow-up, following adjustments for demographic variables (hazard ratio 1.64; 95% CI 1.25-2.16; p < 0.001). In the ACE csf cohort, plasma and CSF YKL-40 were correlated (r = 0.31), and both were associated with conversion from mild cognitive impairment to dementia, independent of amyloid, tau, and neurodegeneration status. DISCUSSION Higher plasma YKL-40 levels were associated with lower brain volume, poorer cognition, and incident dementia. Plasma YKL-40 may be useful for studying the association of inflammation and its treatment on dementia risk.
Collapse
Affiliation(s)
- Matthew P Pase
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Jayandra J Himali
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Raquel Puerta
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alexa S Beiser
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mitzi M Gonzales
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Claudia L Satizabal
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Qiong Yang
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Hugo J Aparicio
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Daniel J Kojis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Charles S Decarli
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Oscar L Lopez
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Will Longstreth
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Vilmundur Gudnason
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Thomas H Mosley
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Joshua C Bis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alison Fohner
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Bruce M Psaty
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mercè Boada
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Pablo García-González
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sergi Valero
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Marta Marquié
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Russell Tracy
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Lenore J Launer
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Agustín Ruiz
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Myriam Fornage
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sudha Seshadri
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| |
Collapse
|
42
|
Ferrante M, Boccato T, Toschi N. Enabling uncertainty estimation in neural networks through weight perturbation for improved Alzheimer's disease classification. Front Neuroinform 2024; 18:1346723. [PMID: 38380126 PMCID: PMC10876844 DOI: 10.3389/fninf.2024.1346723] [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: 11/29/2023] [Accepted: 01/15/2024] [Indexed: 02/22/2024] Open
Abstract
Background The willingness to trust predictions formulated by automatic algorithms is key in a wide range of domains. However, a vast number of deep architectures are only able to formulate predictions without associated uncertainty. Purpose In this study, we propose a method to convert a standard neural network into a Bayesian neural network and estimate the variability of predictions by sampling different networks similar to the original one at each forward pass. Methods We combine our method with a tunable rejection-based approach that employs only the fraction of the data, i.e., the share that the model can classify with an uncertainty below a user-set threshold. We test our model in a large cohort of brain images from patients with Alzheimer's disease and healthy controls, discriminating the former and latter classes based on morphometric images exclusively. Results We demonstrate how combining estimated uncertainty with a rejection-based approach increases classification accuracy from 0.86 to 0.95 while retaining 75% of the test set. In addition, the model can select the cases to be recommended for, e.g., expert human evaluation due to excessive uncertainty. Importantly, our framework circumvents additional workload during the training phase by using our network "turned into Bayesian" to implicitly investigate the loss landscape in the neighborhood of each test sample in order to determine the reliability of the predictions. Conclusion We believe that being able to estimate the uncertainty of a prediction, along with tools that can modulate the behavior of the network to a degree of confidence that the user is informed about (and comfortable with), can represent a crucial step in the direction of user compliance and easier integration of deep learning tools into everyday tasks currently performed by human operators.
Collapse
Affiliation(s)
- Matteo Ferrante
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Tommaso Boccato
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Dyer AH, McNulty H, Caffrey A, Gordon S, Laird E, Hoey L, Hughes CF, Ward M, Strain JJ, O'Kane M, Tracey F, Molloy AM, Cunningham C, McCarroll K. Low-Grade systemic inflammation is associated with domain-specific cognitive performance and cognitive decline in older adults: Data from the TUDA study. Neurobiol Aging 2024; 134:94-105. [PMID: 38043161 DOI: 10.1016/j.neurobiolaging.2023.11.008] [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: 08/11/2023] [Revised: 11/12/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023]
Abstract
Studies examining the relationships between chronic inflammation, cognitive function and cognitive decline in older adults have yielded conflicting results. In a large cohort of older adults free from established dementia (n = 3270; 73.1 ± 7.9 years; 68.4% female), we evaluated the cross-sectional and longitudinal relationships between serum cytokines (IL-6, IL-10, TNF-α) and both global and domain-specific cognitive performance (Repeatable Battery for Assessment of Neuropsychological Status [RBANS]). Higher IL-6 (OR: 1.33; 1.06, 1.66, p = 0.01), TNF-α (OR: 1.35; 1.09, 1.67, p = 0.01) and IL-6:IL-10 Ratio (OR: 1.43; 1.17, 1.74, p = 0.001) were cross-sectionally associated with impaired global RBANS performance. For specific cognitive domains, greatest effect sizes were observed between higher TNF-α levels and poorer visual-spatial and attention performance. In a subset of participants (n = 725; 69.8 ± 5.5 years; 67.0% female) with repeat assessment performed at a median of 5.4 years, only higher baseline IL-6:IL-10 ratio was associated with impaired incident overall, immediate memory and visual-spatial performance. Associations were stronger in females, but not modified by age or APOE genotype.
Collapse
Affiliation(s)
- Adam H Dyer
- Department of Age-Related Healthcare, Tallaght University Hospital, United Kingdom; Department of Medical Gerontology, School of Medicine, Trinity College Dublin, United Kingdom.
| | - Helene McNulty
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Aoife Caffrey
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Shane Gordon
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Eamon Laird
- Department of Physical Education and Sport Science, University of Limerick, United Kingdom
| | - Leane Hoey
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Catherine F Hughes
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Mary Ward
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - J J Strain
- The Nutrition Innovation Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Maurice O'Kane
- Clinical Chemistry Laboratory, Altnagelvin Hospital, Western Health and Social Care Trust, Londonderry, Northern Ireland, United Kingdom
| | - Fergal Tracey
- Causeway Hospital, Northern Health and Social Care Trust, Coleraine, Northern Ireland, United Kingdom
| | | | - Conal Cunningham
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, United Kingdom; Mercer's Institute for Successful Ageing, St James's Hospital, Dublin, Ireland
| | - Kevin McCarroll
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, United Kingdom; Mercer's Institute for Successful Ageing, St James's Hospital, Dublin, Ireland
| |
Collapse
|
44
|
Maheshwari S, Singh A, Ansari VA, Mahmood T, Wasim R, Akhtar J, Verma A. Navigating the dementia landscape: Biomarkers and emerging therapies. Ageing Res Rev 2024; 94:102193. [PMID: 38215913 DOI: 10.1016/j.arr.2024.102193] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
The field of dementia research has witnessed significant developments in our understanding of neurodegenerative disorders, with a particular focus on Alzheimer's disease (AD) and Frontotemporal Dementia (FTD). Dementia, a collection of symptoms arising from the degeneration of brain cells, presents a significant healthcare challenge, especially as its prevalence escalates with age. This abstract delves into the complexities of these disorders, the role of biomarkers in their diagnosis and monitoring, as well as emerging neurophysiological insights. In the context of AD, anti-amyloid therapy has gained prominence, aiming to reduce the accumulation of amyloid-beta (Aβ) plaques in the brain, a hallmark of the disease. Notably, Leqembi recently received full FDA approval, marking a significant breakthrough in AD treatment. Additionally, ongoing phase 3 clinical trials are investigating novel therapies, including Masitinib and NE3107, focusing on cognitive and functional improvements in AD patients. In the realm of FTD, research has unveiled distinct neuropathological features, including the involvement of proteins like TDP-43 and progranulin, providing valuable insights into the diagnosis and management of this heterogeneous condition. Biomarkers, including neurofilaments and various tau fragments, have shown promise in enhancing diagnostic accuracy. Neurophysiological techniques, such as transcranial magnetic stimulation (TMS), have contributed to our understanding of AD and FTD. TMS has uncovered unique neurophysiological signatures, highlighting impaired plasticity, hyperexcitability, and altered connectivity in AD, while FTD displays differences in neurotransmitter systems, particularly GABAergic and glutamatergic circuits. Lastly, ongoing clinical trials in anti-amyloid therapy for AD, such as Simufilam, Solanezumab, Gantenerumab, and Remternetug, offer hope for individuals affected by this devastating disease, with the potential to alter the course of cognitive decline. These advancements collectively illuminate the evolving landscape of dementia research and the pursuit of effective treatments for these challenging conditions.
Collapse
Affiliation(s)
- Shubhrat Maheshwari
- Faculty of Pharmaceutical Sciences Rama University Mandhana, Bithoor Road, Kanpur, Uttar Pradesh 209217, India; Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| | - Aditya Singh
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Vaseem Ahamad Ansari
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Tarique Mahmood
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Rufaida Wasim
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Juber Akhtar
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Amita Verma
- Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| |
Collapse
|
45
|
Savall ASP, de Mello JD, Fidelis EM, Comis-Neto AA, Nepomuceno MR, Pacheco CDO, Haas SE, Pinton S. Nanoencapsulated Curcumin: Enhanced Efficacy in Reversing Memory Loss in An Alzheimer Disease Model. Brain Sci 2024; 14:130. [PMID: 38391705 PMCID: PMC10886961 DOI: 10.3390/brainsci14020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Investigating new drugs or formulations that target Alzheimer disease (AD) is critical for advancing therapeutic interventions. Therefore, this study aimed to assess the effectiveness of nanoencapsulated curcumin (NC Curc) in alleviating memory impairment, oxidative stress, and neuroinflammation in a validated AD model. Male Wistar rats were given bilateral intracerebroventricular injections of either saline or streptozotocin (STZ) (3 mg/3 µL/site) to establish the AD model (day 0). On day 22, daily oral administrations of curcumin (6 mg/kg), NC Curc (6 mg/kg), or a vehicle (unloaded NC) were initiated and continued for 14 days. NC Curc significantly reversed memory deficits in object recognition and inhibitory avoidance tests induced by STZ. Both formulations of curcumin attenuated elevated acetylcholinesterase activity caused by STZ. Importantly, NC Curc alone effectively mitigated STZ-induced oxidative stress. Additionally, NC Curc treatment normalized GFAP levels, suggesting a potential reduction in neuroinflammation in STZ-treated rats. Our findings indicate that NC Curc improves memory in an AD rat model, highlighting its enhanced therapeutic effects compared to unencapsulated curcumin. This research significantly contributes to understanding the therapeutic and neurorestorative potential of NC Curc in AD, particularly in reversing pathophysiological changes.
Collapse
Affiliation(s)
- Anne Suély Pinto Savall
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Jhuly Dorneles de Mello
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Eduarda Monteiro Fidelis
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Antonio Alvenir Comis-Neto
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Maria Regina Nepomuceno
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Camila de Oliveira Pacheco
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Sandra Elisa Haas
- Laboratory of Pharmacology and Pharmacometrics, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| | - Simone Pinton
- Research Group on Biochemistry and Toxicology in Eukaryotes, Federal University of Pampa, Campus Uruguaiana, Uruguaiana 97500-970, RS, Brazil
| |
Collapse
|
46
|
Muñoz-Grajales C, Barraclough ML, Diaz-Martinez JP, Su J, Bingham K, Kakvan M, Kretzmann RP, Tartaglia MC, Ruttan L, Choi MY, Appenzeller S, Marzouk S, Bonilla D, Katz P, Beaton D, Green R, Gladman DD, Wither J, Touma Z. Serum S100A8/A9 and MMP-9 levels are elevated in systemic lupus erythematosus patients with cognitive impairment. Front Immunol 2024; 14:1326751. [PMID: 38332909 PMCID: PMC10851148 DOI: 10.3389/fimmu.2023.1326751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024] Open
Abstract
Objective Cognitive impairment (CI) is one of the most common manifestations of Neuropsychiatric Systemic Lupus Erythematosus (NPSLE). Despite its frequency, we have a limited understanding of the underlying immune mechanisms, resulting in a lack of pathways to target. This study aims to bridge this gap by investigating differences in serum analyte levels in SLE patients based on their cognitive performance, independently from the attribution to SLE, and exploring the potential for various serum analytes to differentiate between SLE patients with and without CI. Methods Two hundred ninety individuals aged 18-65 years who met the 2019-EULAR/ACR classification criteria for SLE were included. Cognitive function was measured utilizing the adapted ACR-Neuropsychological Battery (ACR-NB). CI was defined as a z-score of ≤-1.5 in two or more domains. The serum levels of nine analytes were measured using ELISA. The data were randomly partitioned into a training (70%) and a test (30%) sets. Differences in the analyte levels between patients with and without CI were determined; and their ability to discriminate CI from non-CI was evaluated. Results Of 290 patients, 40% (n=116) had CI. Serum levels of S100A8/A9 and MMP-9, were significantly higher in patients with CI (p=0.006 and p=0.036, respectively). For most domains of the ACR-NB, patients with CI had higher S100A8/A9 serum levels than those without. Similarly, S100A8/A9 had a negative relationship with multiple CI tests and the highest AUC (0.74, 95%CI: 0.66-0.88) to differentiate between patients with and without CI. Conclusion In this large cohort of well-characterized SLE patients, serum S100A8/A9 and MMP-9 were elevated in patients with CI. S100A8/A9 had the greatest discriminatory ability in differentiating between patients with and without CI.
Collapse
Affiliation(s)
- Carolina Muñoz-Grajales
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Michelle L. Barraclough
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- National Institute for Health and Care Research (NIHR), Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Juan P. Diaz-Martinez
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Jiandong Su
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Kathleen Bingham
- Centre for Mental Health, University Health Network, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Mahta Kakvan
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Roberta Pozzi Kretzmann
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- Department of Medicine, Division of Neurology, University of Toronto Krembil Neurosciences Centre, Toronto, ON, Canada
| | - Lesley Ruttan
- Department of Psychology, University Health Network-Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - May Y. Choi
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Simone Appenzeller
- School of Medical Science, Department of Orthopedics, Rheumatology and Traumatology, University of Campinas, São Paulo, Brazil
| | - Sherief Marzouk
- Centre for Mental Health, University Health Network, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Dennisse Bonilla
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Patricia Katz
- Division of Rheumatology, Department of Medicine, and Institute for Health Policy Studies, University of California, San Francisco, Novato, CA, United States
| | - Dorcas Beaton
- Institute for Work and Health, University of Toronto, Toronto, ON, Canada
| | - Robin Green
- Department of Psychology, University Health Network-Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Dafna D. Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | - Joan Wither
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, ON, Canada
| | - Zahi Touma
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- University of Toronto Lupus Clinic, Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
47
|
Lin M, Yu JX, Zhang WX, Lao FX, Huang HC. Roles of TREM2 in the Pathological Mechanism and the Therapeutic Strategies of Alzheimer's Disease. J Prev Alzheimers Dis 2024; 11:1682-1695. [PMID: 39559879 PMCID: PMC11573818 DOI: 10.14283/jpad.2024.164] [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: 11/20/2024]
Abstract
Alzheimer's disease (AD) is an age-related degenerative disease, which is characteristic by the deposition of senile plaques (SP) outside the cells, the neurofibrillary tangles (NFTs) inside the neurons, and the loss of synapse and neurons. Neuroinflammation may play an important role in the pathogenesis of AD. Microglia are the immune cells in the central nervous system. However, microglia might become disease-related microglia (DAMs) when stimulated by the external environment. DAMs have been shown to be involved in a series of events of AD development including Aβ accumulation and tau phosphorylation. The triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane receptor that is mainly expressed by microglia in the central nervous system (CNS). TREM2 plays an important role in the physiological function of microglia, and the dyshomeostasis of TREM2 is related to the development of late-onset AD. This article summarized the latest advances in TREM2 biology and its impact on the roles of microglia in AD development, with a particular emphasis on the structure, ligands, signal transduction, and the agonistic antibodies of TREM2 for AD treatment. We further discussed the survival, migration, phagocytosis, inflammation, and cellular metabolism of microglia, as well as the role of sTREM2 in neuroprotection and as a biomarker for AD. It provides a reference for further research on the molecular mechanism of microglial TREM2 in the occurrence and development of AD and on the therapeutic strategies targeted on the microglial TREM2.
Collapse
Affiliation(s)
- M Lin
- Dr. Han-Chang Huang, Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100023, China. No. 18, Fatou third block, Chaoyang District, Beijing, China, E-mail: , Phone: +8610-52072057
| | | | | | | | | |
Collapse
|
48
|
Rathod SS, Agrawal YO. Phytocannabinoids as Potential Multitargeting Neuroprotectants in Alzheimer's Disease. Curr Drug Res Rev 2024; 16:94-110. [PMID: 37132109 DOI: 10.2174/2589977515666230502104021] [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: 10/20/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 05/04/2023]
Abstract
The Endocannabinoid System (ECS) is a well-studied system that influences a variety of physiological activities. It is evident that the ECS plays a significant role in metabolic activities and also has some neuroprotective properties. In this review, we emphasize several plant-derived cannabinoids such as β-caryophyllene (BCP), Cannabichromene (CBC), Cannabigerol (CBG), Cannabidiol (CBD), and Cannabinol (CBN), which are known to have distinctive modulation abilities of ECS. In Alzheimer's disease (AD), the activation of ECS may provide neuroprotection by modulating certain neuronal circuitry pathways through complex molecular cascades. The present article also discusses the implications of cannabinoid receptors (CB1 and CB2) as well as cannabinoid enzymes (FAAH and MAGL) modulators in AD. Specifically, CBR1 or CB2R modulations result in reduced inflammatory cytokines such as IL-2 and IL-6, as well as a reduction in microglial activation, which contribute to an inflammatory response in neurons. Furthermore, naturally occurring cannabinoid metabolic enzymes (FAAH and MAGL) inhibit the NLRP3 inflammasome complex, which may offer significant neuroprotection. In this review, we explored the multi-targeted neuroprotective properties of phytocannabinoids and their possible modulations, which could offer significant benefits in limiting AD.
Collapse
Affiliation(s)
- Sumit S Rathod
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
- Shri Vile Parle Kelavani Mandal's, Institute of Pharmacy, Dhule, Dist. Dhule, 424001, Maharashtra, India
| | - Yogeeta O Agrawal
- Department of Pharmacy, R.C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dist. Dhule, 425405, Maharashtra, India
| |
Collapse
|
49
|
Trabert M, Seifert R. Critical analysis of ginkgo preparations: comparison of approved drugs and dietary supplements marketed in Germany. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:451-461. [PMID: 37470803 PMCID: PMC10771617 DOI: 10.1007/s00210-023-02602-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 06/24/2023] [Indexed: 07/21/2023]
Abstract
Demographic change is taking place in the population of western industrialized countries, and the population is aging constantly. As a result, the mortality rate of patients due to dementia is rising steadily. To counteract this, the relevance of neuroprotective agents is increasing. Preparations from the medicinal tree species Ginkgo biloba ("gingko") are becoming increasingly popular. In this study, 63 ginkgo preparations marketed in Germany were analyzed. The following data were collected from the package inserts of the preparations: Country of manufacture, approval as a drug, compliance to target values of flavone glycosides, compliance to target values of terpene lactones, compliance to target values of ginkgolic acids, dosage per unit in milligrams (mg), duration of use, interactions with other drugs, contraindications, adverse effects and daily defined dose costs. In the next step, these data were compared in the following form: Total preparations versus preparations with drug approval versus dietary supplements. Almost without exception, the results indicate a pharmaceutical reliability of the preparations with drug approval and a dubious reliability of the preparations marketed as dietary supplements. Thus, ginkgo preparations marketed as dietary supplements appear to have an economic rather than a medical focus. We discuss the evidence of efficacy, and other criteria mentioned above, to evaluate the adequacy of the costs for the statutory health insurance that pay for preparations with drug approval in Germany. From the analysis of our results it is very doubtful that ginkgo biloba extract preparations of the food industry have any health benefit. It must be evaluated whether prohibition of selling ginkgo biloba extract as a dietary supplement is an option.
Collapse
Affiliation(s)
- Milan Trabert
- Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625, Hanover, Germany
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625, Hanover, Germany.
| |
Collapse
|
50
|
Sharma R. Innovative Genoceuticals in Human Gene Therapy Solutions: Challenges and Safe Clinical Trials of Orphan Gene Therapy Products. Curr Gene Ther 2024; 24:46-72. [PMID: 37702177 DOI: 10.2174/1566523223666230911120922] [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: 11/03/2022] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 09/14/2023]
Abstract
The success of gene therapy attempts is controversial and inconclusive. Currently, it is popular among the public, the scientific community, and manufacturers of Gene Therapy Medical Products. In the absence of any remedy or treatment options available for untreatable inborn metabolic orphan or genetic diseases, cancer, or brain diseases, gene therapy treatment by genoceuticals and T-cells for gene editing and recovery remains the preferred choice as the last hope. A new concept of "Genoceutical Gene Therapy" by using orphan 'nucleic acid-based therapy' aims to introduce scientific principles of treating acquired tissue damage and rare diseases. These Orphan Genoceuticals provide new scope for the 'genodrug' development and evaluation of genoceuticals and gene products for ideal 'gene therapy' use in humans with marketing authorization application (MAA). This perspective study focuses on the quality control, safety, and efficacy requirements of using 'nucleic acid-based and human cell-based new gene therapy' genoceutical products to set scientific advice on genoceutical-based 'orphan genodrug' design for clinical trials as per Western and European guidelines. The ethical Western FDA and European EMA guidelines suggest stringent legal and technical requirements on genoceutical medical products or orphan genodrug use for other countries to frame their own guidelines. The introduction section proposes lessknown 'orphan drug-like' properties of modified RNA/DNA, human cell origin gene therapy medical products, and their transgene products. The clinical trial section explores the genoceutical sources, FDA/EMA approvals for genoceutical efficacy criteria with challenges, and ethical guidelines relating to gene therapy of specific rare metabolic, cancer and neurological diseases. The safety evaluation of approved genoceuticals or orphan drugs is highlighted with basic principles and 'genovigilance' requirements (to observe any adverse effects, side effects, developed signs/symptoms) to establish their therapeutic use. Current European Union and Food and Drug Administration guidelines continuously administer fast-track regulatory legal framework from time to time, and they monitor the success of gene therapy medical product efficacy and safety. Moreover, new ethical guidelines on 'orphan drug-like genoceuticals' are updated for biodistribution of the vector, genokinetics studies of the transgene product, requirements for efficacy studies in industries for market authorization, and clinical safety endpoints with their specific concerns in clinical trials or public use.
Collapse
Affiliation(s)
- Rakesh Sharma
- Surgery NMR Lab, Plastic Surgery Research, Massachusetts General Hospital, Boston, MA 02114, USA
- CCSU, Government Medical College, Saharanpur, 247232 India
| |
Collapse
|