1
|
Zhang Y, Liang Y, Gu Y. The dopaminergic system and Alzheimer's disease. Neural Regen Res 2025; 20:2495-2512. [PMID: 39314145 PMCID: PMC11801300 DOI: 10.4103/nrr.nrr-d-24-00230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/21/2024] [Accepted: 07/31/2024] [Indexed: 09/25/2024] Open
Abstract
Alzheimer's disease is a common neurodegenerative disorder in older adults. Despite its prevalence, its pathogenesis remains unclear. In addition to the most widely accepted causes, which include excessive amyloid-beta aggregation, tau hyperphosphorylation, and deficiency of the neurotransmitter acetylcholine, numerous studies have shown that the dopaminergic system is also closely associated with the occurrence and development of this condition. Dopamine is a crucial catecholaminergic neurotransmitter in the human body. Dopamine-associated treatments, such as drugs that target dopamine receptor D and dopamine analogs, can improve cognitive function and alleviate psychiatric symptoms as well as ameliorate other clinical manifestations. However, therapeutics targeting the dopaminergic system are associated with various adverse reactions, such as addiction and exacerbation of cognitive impairment. This review summarizes the role of the dopaminergic system in the pathology of Alzheimer's disease, focusing on currently available dopamine-based therapies for this disorder and the common side effects associated with dopamine-related drugs. The aim of this review is to provide insights into the potential connections between the dopaminergic system and Alzheimer's disease, thus helping to clarify the mechanisms underlying the condition and exploring more effective therapeutic options.
Collapse
Affiliation(s)
- Yuhan Zhang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yuan Liang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Yixue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| |
Collapse
|
2
|
Joodi SA, Khattab MM, Ibrahim WW. Repurposing of cabergoline to improve cognitive decline in D-galactose-injected ovariectomized rats: Modulation of AKT/mTOR, GLT-1/P38-MAPK, and ERK1/2 signaling pathways. Toxicol Appl Pharmacol 2025; 500:117391. [PMID: 40349788 DOI: 10.1016/j.taap.2025.117391] [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: 01/25/2025] [Revised: 04/18/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Dopamine is involved in many physiological functions including reward phenomenon, motor, learning, and memory functions. Dopamine receptor agonists have been shown to reduce amyloid (Aβ) deposition, enhance memory, and improve cortical plasticity in experimental studies and Alzheimer's disease (AD) patients; however, the molecular mechanisms involved haven't been investigated yet. The target of this investigation was to elucidate the modulatory effects of cabergoline (CAB), a dopamine receptor agonist, against AD. Ovariectomized rats were injected with D-galactose (150 mg/kg/day, i.p) for ten weeks to exacerbate AD. CAB administration (1 mg/kg/day, i.p) for 28 days, beginning from the 7th week of D-galactose administration, attenuated the associated histopathological alterations and enhanced the spatial and recognition memory in Morris water maze and Novel object recognition tests, respectively. CAB decreased the hippocampal concentrations of Aβ42, p-tau, and β-secretase, while upregulating α-secretase. Moreover, CAB diminished nuclear factor-kappa β (NF-κβ), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and myeloperoxidase, while elevating brain-derived neurotrophic factor and phospho-cAMP response element binding protein. Further, CAB reduced the hippocampal phosphorylated forms of protein kinase B (AKT) and mammalian target of rapamycin (mTOR) contrary to elevating Beclin-1, resulting in autophagy induction, which participates in accelerating Aβ42 and p-tau aggregates clearance. Moreover, CAB increased the hippocampal glutamate transporter-1 (GLT-1) protein expression, promoting glutamate uptake that possibly reduced Ca2+ overload and consequently decreased the phosphorylated forms of P38-MAPK and ERK1/2. In conclusion, CAB improved cognitive decline of D-gal/OVX animals, restored hippocampal architecture, exerted neuroprotection, and enhanced autophagic machinery via modulating AKT/mTOR, GLT-1/P38-MAPK, and ERK1/2 pathways.
Collapse
Affiliation(s)
- Sheer A Joodi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
3
|
Morais RF, Sousa JM, Koba C, Andres L, Jesus T, Baldeiras I, Oliveira TG, Santana I. Differential involvement of neurotransmitter pathways in AD, bvFTD and MCI: Whole-brain MRI analysis. Neurobiol Dis 2025; 209:106897. [PMID: 40194635 DOI: 10.1016/j.nbd.2025.106897] [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: 11/19/2024] [Revised: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Neurodegenerative diseases, including Alzheimer's disease (AD), mild cognitive impairment (MCI), and frontotemporal dementia (FTD), are a growing public health challenge, with dementia incidence projected to triple in the coming decades. AD is associated with memory impairment, bvFTD with behavioral dysfunction, and MCI as a transitional stage between normal cognition and dementia. While structural brain changes have been widely studied, the role of neurotransmitter pathways remains underexplored. This study aims to correlate gray matter atrophy in AD, bvFTD, and MCI with neurotransmitter pathways to identify distinctive neurochemical impairments. METHODS We included 214 participants (89 CE, 74 bvFTD, 51 MCI) from a single-center cohort. MRI from 3 T scanners was segmented via FreeSurfer. Neurotransmitter maps were sourced from JuSpace. We performed volumetric and whole-brain correlation analyses to evaluate relationships between brain regional volumes (BRVs) and neurotransmitter pathways. Group differences were assessed with Kruskal-Wallis tests followed by post-hoc analyses. RESULTS Volumetric analysis showed expected atrophy patterns in each group. Correlation analysis indicated distinct neurotransmitter involvement: AD showed significant atrophy correlations with dopamine D2 and GABA A receptor distribution; bvFTD had significant negative correlations with the mu-opioid receptor; MCI exhibited early serotonergic dysregulation. CONCLUSIONS We identified distinct atrophy patterns linked to specific neurotransmitter systems, each showing unique neurochemical profiles. In AD, precuneus and inferior parietal lobules atrophy aligns with dopaminergic and GABAergic receptors, potentially impacting memory and executive functions. In bvFTD, medial orbitofrontal and temporal atrophy, is linked to mu-opioid receptor impairment, possibly contributing to behavioral symptoms. In MCI, early serotonergic dysregulation involving SERT occurs before detectable atrophy.
Collapse
Affiliation(s)
- Ricardo Félix Morais
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência (INESC TEC), Porto, Portugal; Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal; Neuroradiology Department, ULS São João, Porto, Portugal.
| | | | - Cemal Koba
- Sano Centre for Computational Medicine, Computational Neuroscience Team, Kraków, Poland
| | - Leon Andres
- Department of Statistics, National University of Colombia, Bogotá, Colombia
| | - Tiago Jesus
- Center Algoritmi, LASI, University of Minho, Braga, Portugal; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal; Neurology Department, ULS de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, ULS, Braga, Braga, Portugal
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Centre for Innovative Biomedicine and Biotechnology (CIBB), Universidade de Coimbra, Coimbra, Portugal; Neurology Department, ULS de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Islam MT, Al Shamsh Prottay A, Bhuia MS, Akbor MS, Chowdhury R, Ansari SA, Ansari IA, Islam MA, Tahim CM, Coutinho HDM. Memory-Enhancing Effects of Daidzin, Possibly Through Dopaminergic and AChEergic Dependent Pathways. J Nutr 2025:S0022-3166(25)00269-X. [PMID: 40324526 DOI: 10.1016/j.tjnut.2025.04.024] [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: 09/28/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND The soy isoflavone daidzin (DZN) possesses cognitive-enhancing effects in animals. OBJECTIVES However, the mechanism for this effect is yet to be discovered. METHODS For this, we investigate its memory-enhancing capacity using the mouse models of marble burying, dust removal, an open-field study, and in silico studies. Adult male Swiss albino mice were randomly assigned to different groups consisting of control (vehicle: 10 mL/kg), DZN 5, 10, and 20 mg/kg, dopamine (agonist: 22 mg/kg), galantamine (inhibitor: 3 mg/kg), and a combination of DZN-10 with standards. RESULTS DZN dose-dependently and significantly (P < 0.05) increased marble burying and removed dust while decreasing the total distance in the open-field test (OFT). DZN-10 enhanced dopamine's effect significantly (P < 0.05). In silico findings suggest that DZN has strong binding capacities of -10.3, -7.5, -9.8, and -9.2 kcal/mol to the acetylcholinesterase (AChE), D1, D3, and D5 receptors, respectively. CONCLUSIONS Taken together, DZN may exert its memory-enhancing ability by interacting with AChE and dopamine receptors.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh; Department of Pharmacy, Gopalganj Science and Technology University, Gopalganj, Bangladesh.
| | - Abdullah Al Shamsh Prottay
- Department of Pharmacy, Gopalganj Science and Technology University, Gopalganj, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Md Shimul Bhuia
- Department of Pharmacy, Gopalganj Science and Technology University, Gopalganj, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh.
| | - Md Showkot Akbor
- Department of Pharmacy, Gopalganj Science and Technology University, Gopalganj, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Gopalganj Science and Technology University, Gopalganj, Bangladesh; Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Md Amirul Islam
- Pharmacy Discipline, Khulna University, Khulna 9208, Bangladesh; Department of Pharmacy, East West University, Dhaka, Bangladesh
| | - Catarina Martins Tahim
- CECAPE College, Department of Odontology, Av. Padre Cícero, 3917 - São José, Juazeiro do Norte - CE, 63024-015, Brazil
| | | |
Collapse
|
5
|
Quintero-Espinosa DA, Velez-Pardo C, Jimenez-Del-Rio M. PF-06447475 Molecule Attenuates the Neuropathology of Familial Alzheimer's and Coexistent Parkinson's Disease Markers in PSEN1 I416T Dopaminergic-like Neurons. Molecules 2025; 30:2034. [PMID: 40363838 PMCID: PMC12074268 DOI: 10.3390/molecules30092034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Familial Alzheimer's disease (FAD) is a complex multifactorial disorder clinically characterized by cognitive impairment and memory loss. Pathologically, FAD is characterized by intracellular accumulation of the protein fragment Aβ42 (iAβ), hyperphosphorylated microtubule-associated protein TAU (p-TAU), and extensive degeneration of basal forebrain cholinergic neurons of the nucleus basalis of Meynert (NbM) and the medial septal nucleus (MSN), mainly caused by mutations in the amyloid precursor protein (APP), presenilin 1 (PSEN1), and PSEN2 gene. Since the dopaminergic system may contribute to FAD symptoms, alterations in the nigro-hippocampal pathway may be associated with cognitive impairment in FAD. Interestingly, p-α-synuclein (p-α-Syn), Aβ, and p-TAU have been found to coexist in vulnerable regions of postmortem AD brains. However, the mechanism by which Aβ, p-TAU, and α-Syn coexist in DAergic neurons in AD brains has not been determined. We generated PSEN1 I416T dopaminergic-like neurons (DALNs) from I416T menstrual stromal cells (MenSCs) in NeuroForsk 2.0 medium for 7 days and then cultured them in minimal culture medium (MCm) for another 4 days. On day 11, DALNs were analyzed for molecular and pathological markers by flow cytometry and fluorescence microscopy. We found that mutant DALNs showed increased accumulation of iAβ as well as increased phosphorylation of TAU at S202/T205 compared to WT DALNs. Thus, mutant DALNs exhibited typical pathological hallmarks of Alzheimer's disease. Furthermore, PSEN1 I416T DALNs showed concomitant signs of OS as evidenced by the appearance of oxidized sensor protein DJ-1 (i.e., DJ-1C106-SO3) and apoptotic markers TP53, pS63-c-JUN, PUMA, and cleavage caspase 3 (CC3). Notably, these DALNs exhibited PD-associated proteins such as intracellular accumulation of α-Syn (detected as aggregates of pS129-α-Syn) and phosphorylation of LRRK2 kinase at residue S935. In addition, mutant DALNs showed a 17.16- and 6.17-fold decrease in DA-induced Ca2+ flux, compared to WT DALNs. These observations suggest that iAβ and p-TAU, together with p-α-Syn, and p-LRRK2 kinase, may damage DAergic neurons and thereby contribute to the exacerbation of neuropathologic processes in FAD. Remarkably, the LRRK2 inhibitor PF-06447475 (PF-475) significantly reversed PSEN1 I416T-induced neuropathological markers in DAergic neurons. PF-465 inhibitor reduced iAβ, oxDJ-1C106-SO3, and p-TAU. In addition, this inhibitor reduced pS935-LRRK2, pS129-αSYN, pS63-c-JUN, and CC3. We conclude that the observed neuroprotective effects of PF-475 are due to direct inhibition of LRRK2 activity and that the LRRK2 protein is upstream of the molecular cascade of apoptosis and proteinopathy. Our results suggest that PF-475 is an effective neuroprotective agent against endogenous PSEN1 I416T-induced neurotoxicity in DALNs coexisting with Parkinson's disease markers. Therefore, PF-475 may be of great therapeutic value in FAD.
Collapse
Affiliation(s)
| | | | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Institute of Medical Research, Faculty of Medicine, University Research Headquarters, Calle 62#52-59, Building 1, Laboratory 411/412, Medellin 050010, Colombia; (D.A.Q.-E.); (C.V.-P.)
| |
Collapse
|
6
|
Sárkány Z, Damásio J, Macedo‐Ribeiro S, Martins PM. Association between the use of levodopa/carbidopa, Alzheimer's disease biomarkers, and cognitive decline among participants in the National Alzheimer's Coordinating Center Uniform Data Set. Alzheimers Dement 2025; 21:e70213. [PMID: 40356023 PMCID: PMC12069009 DOI: 10.1002/alz.70213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 05/15/2025]
Abstract
INTRODUCTION This retrospective study investigates whether exposure to levodopa/carbidopa (LA/CA) medication is associated with modified Alzheimer's disease (AD) trajectories. METHODS Multivariate analysis used cerebrospinal fluid (CSF) biomarker information included in the National Alzheimer's Coordinating Center Uniform Data Set for subjects with normal cognition (NC), mild cognitive impairment (MCI), and dementia (DE). Survival analyses examined the progression to MCI/DE and death events. RESULTS LA/CA use is associated with lower levels of CSF amyloid beta, phosphorylated-tau (p-tau) and total-tau. After adjusting for age, sex, and apolipoprotein E (APOE) ε4 allele presence, that effect was quantified by negative coefficients of the fitted linear mixed models: p-values < 0.01 in all cases except for p-tau in the MCI subgroup (p = 0.02). No similar effects were identified for other antiparkinsonians. Exposure to LA/CA decreased the progression from MCI to DE (p = 0.03). DISCUSSION The identified association between LA/CA exposure, AD biomarkers, and progression deserves further investigation in controlled clinical trials. HIGHLIGHTS LA/CA is associated with lower levels of CSF biomarkers for AD. This effect is not observed when other antiparkinsonian drugs are used. LA/CA is also associated with delayed progression to dementia by AD patients with MCI.
Collapse
Affiliation(s)
- Zsuzsa Sárkány
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC – Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Joana Damásio
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Centro de Genética Preditiva e Preventiva (CGPP)IBMC Universidade do PortoPortoPortugal
- Neurology DepartmentCentro Hospitalar Universitário de Santo António, ULS de Santo AntónioPortoPortugal
- ICBAS School of Medicine and Biomedical SciencesUniversidade do PortoPortoPortugal
| | - Sandra Macedo‐Ribeiro
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC – Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Pedro M. Martins
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC – Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| |
Collapse
|
7
|
La Barbera L, Krashia P, Nobili A. How dopamine tunes parvalbumin interneurons in the hippocampus: new experimental observations in Alzheimer's disease. Neural Regen Res 2025; 20:1405-1406. [PMID: 39075905 PMCID: PMC11624865 DOI: 10.4103/nrr.nrr-d-24-00322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/07/2024] [Accepted: 05/21/2024] [Indexed: 07/31/2024] Open
Affiliation(s)
- Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, Rome, Italy
| | - Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, Rome, Italy
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, Rome, Italy
| |
Collapse
|
8
|
Drumond-Bock AL, Blankenship HE, Pham KD, Carter KA, Freeman WM, Beckstead MJ. Parallel Gene Expression Changes in Ventral Midbrain Dopamine and GABA Neurons during Normal Aging. eNeuro 2025; 12:ENEURO.0107-25.2025. [PMID: 40360281 PMCID: PMC12121937 DOI: 10.1523/eneuro.0107-25.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/18/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
The consequences of aging can vary dramatically between different brain regions and cell types. In the ventral midbrain, dopaminergic neurons develop physiological deficits with normal aging that likely convey susceptibility to neurodegeneration. While nearby GABAergic neurons are thought to be more resilient, decreased GABA signaling in other areas nonetheless correlates with age-related cognitive decline and the development of degenerative diseases. Here, we used two novel cell type-specific translating ribosome affinity purification models to elucidate the impact of healthy brain aging on the molecular profiles of dopamine and GABA neurons in the ventral midbrain. By analyzing differential gene expression from young adult (7-10 months) and old (21-24 months) mice, we detected commonalities in the aging process in both neuronal types, including increased inflammatory responses and upregulation of pro-survival pathways. Both cell types also showed downregulation of genes involved in synaptic connectivity and plasticity. Intriguingly, genes involved in serotonergic synthesis were upregulated with age in GABA neurons and not dopamine-releasing cells. In contrast, dopaminergic neurons showed alterations in genes connected with mitochondrial function and calcium signaling, which were markedly downregulated in male mice. Sex differences were detected in both neuron types, but in general were more prominent in dopamine neurons. Multiple sex effects correlated with the differential prevalence for neurodegenerative diseases such as Parkinson's and Alzheimer's seen in humans. In summary, these results provide insight into the connection between non-pathological aging and susceptibility to neurodegenerative diseases involving the ventral midbrain, and identify molecular phenotypes that could underlie homeostatic maintenance during normal aging.
Collapse
Affiliation(s)
- Ana Luiza Drumond-Bock
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Harris E Blankenship
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
| | - Kevin D Pham
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Kelsey A Carter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
| | - Willard M Freeman
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma 73104
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104
- Department of Biochemistry and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma 73104
| |
Collapse
|
9
|
De Paolis ML, Loffredo G, Krashia P, La Barbera L, Nobili A, Cauzzi E, Babicola L, Di Segni M, Coccurello R, Puglisi-Allegra S, Latagliata EC, D'Amelio M. Repetitive prefrontal tDCS activates VTA dopaminergic neurons, resulting in attenuation of Alzheimer's Disease-like deficits in Tg2576 mice. Alzheimers Res Ther 2025; 17:94. [PMID: 40301905 PMCID: PMC12039073 DOI: 10.1186/s13195-025-01736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND Emerging evidence implicates early dysfunction of dopaminergic neurons in the Ventral Tegmental Area (VTA) as a key contributor to Alzheimer's Disease (AD) pathophysiology. Specifically, the VTA dopaminergic neurodegeneration and the consequent reduction of dopamine (DA) in mesocorticolimbic targets are associated with the onset of cognitive impairments and neuropsychiatric-like manifestations in AD animal models. Moreover, decreased midbrain volume and functional VTA disconnection are identified as predictors of accelerated progression from Mild Cognitive Impairment to AD-dementia in clinical populations. Given these findings, interventions capable of directly modulating VTA activity and augmenting DA release, despite the ongoing neurodegeneration, may hold therapeutic potential for mitigating DA-related deficits in AD. This study aims at evaluating the therapeutic potential of prefrontal transcranial Direct Current Stimulation (tDCS) in the Tg2576 mouse model of AD, exhibiting early VTA dopaminergic neurodegeneration. METHODS Repeated tDCS was applied to assess its ability to activate VTA DA neurons. We also evaluated tDCS effects on synaptic plasticity, cognitive and non-cognitive behaviours and AD-related pathology. Hippocampal DA release and Nucleus Accumbens (NAc) DA transporter (DAT) expression were measured. With immunohistochemistry we examined microglial density and morphological complexity at different disease stages. Additionally, intracellular amyloid-β (Aβ) levels and plaque burden were evaluated to determine the impact of tDCS on AD pathology. RESULTS Prefrontal tDCS enhanced the activity of VTA dopaminergic neurons, leading to increased hippocampal DA release and higher DAT levels in the NAc. The enhanced DA outflow is associated with restored CA3-CA1 synaptic plasticity and improvements in recognition memory and motivational behaviours. tDCS reduced microglial numbers and morphological complexity in Tg2576 mice at both pre-plaque stage (7-months) and at an advanced stage characterized by plaque accumulation (12-months). Notably, tDCS also decreased Aβ plaque burden, although no changes in intracellular Aβ levels were observed in younger Tg2576 mice. CONCLUSIONS These findings highlight the multifaceted therapeutic potential of prefrontal tDCS in targeting key AD pathophysiological hallmarks, including dopaminergic dysfunction, synaptic impairments, neuroinflammation and plaque deposition. As a non-invasive neuromodulatory approach, prefrontal tDCS emerges as a promising early intervention strategy to complement existing AD treatments, with the potential to improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Gilda Loffredo
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | - Lucy Babicola
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- Department of Psychology, Sapienza University of Rome, P.Le Aldo Moro, 5, 00185, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, IRCCS Eugenio Medea, Via Don Luigi Monza, 20, 23842, Bosisio Parini, Italy
| | - Roberto Coccurello
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy
- National Research Council (CNR), Institute for Complex System (ISC), Via Dei Taurini, 19, 00185, Rome, Italy
| | - Stefano Puglisi-Allegra
- Istituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Neuromed, Via Atinense, 18, 86077, Pozzilli, Italy
| | - Emanuele Claudio Latagliata
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
- Department of Psychology, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Via Alvaro del Portillo, 21, 00128, Rome, Italy.
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso Di Fiorano, 64, 00143, Rome, Italy.
| |
Collapse
|
10
|
Ostlund SB, Chen G, Kosheleff A, Lueptow LM, Zhuravka I, Frautschy SA, Lam HA, Maidment NT. Early emergence of motivational and hedonic feeding deficits in the TgF344-AD rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1572956. [PMID: 40357232 PMCID: PMC12066702 DOI: 10.3389/fnagi.2025.1572956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is characterized by progressive cognitive decline and has a long prodromal phase during which subclinical cognitive deficits and neuropsychiatric symptoms may begin to emerge. Apathy, defined as a lack of motivation or volition, is increasingly recognized as a core feature and a potentially early marker of AD. Despite its significance, apathy-like behavior has been underexplored in transgenic models of AD. Methods We performed a longitudinal analysis of apathy-like behavior using the well-established TgF344-AD rat model. We compared male and female TgF344-AD and wildtype rats on hedonic (palatable food intake) and motivational (progressive ratio) assays during early (3-4 months), intermediate (6-7 months), and later (9-10 months) stages of adulthood. Results We found that female TgF344-AD rats exhibited early and persistent deficits in motivational and hedonic feeding, emerging at 3-4 months and 6-7 months, respectively. During a battery of cognitive tests conducted after 12-14 months of age, TgF344-AD rats were impaired in spatial working memory but also showed wide-ranging deficits in exploratory behavior, which may also be indicative of an apathy-like loss of investigatory drive. Conclusion Our findings highlight the TgF344-AD rat as a valuable model for studying early apathy-like behavior in AD and underscore the need to consider sex differences in AD research to better understand the prodromal phase of this disease.
Collapse
Affiliation(s)
- Sean B. Ostlund
- Department of Anesthesiology and Perioperative Care, Irvine School of Medicine, Irvine Center for Addiction Neuroscience (ICAN), University of California, Irvine, Irvine, CA, United States
| | - Grace Chen
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alisa Kosheleff
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay M Lueptow
- Behavioral Testing Core, Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Irina Zhuravka
- Behavioral Testing Core, Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sally A. Frautschy
- Departments of Neurology and Medicine, Geriatric Research Education and Clinical Center, Veterans Greater Los Angeles HealthCare System, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hoa A. Lam
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nigel T. Maidment
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Tortolani D, Decandia D, Giacovazzo G, Scipioni L, Panuccio A, Ciaramellano F, Eugelio F, Fanti F, Latagliata EC, La Barbera L, Cutuli D, Compagnone D, D’Amelio M, Coccurello R, Oddi S, Petrosini L, Maccarrone M. Chronic palmitoylethanolamide administration via slow-release subcutaneous pellets promotes neuroprotection and mitigates neuroinflammation in the Tg2576 mouse model of Alzheimer's disease. Front Cell Neurosci 2025; 19:1571428. [PMID: 40313591 PMCID: PMC12043567 DOI: 10.3389/fncel.2025.1571428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive and non-cognitive decline associated with neuropathological hallmarks, including neuroinflammation. Palmitoylethanolamide (PEA), an endogenous lipid with anti-inflammatory and neuroprotective properties, has emerged as a promising therapeutic agent in managing AD. This study investigated the therapeutic effects of chronic (6-months) PEA administration via subcutaneous pellet in Tg2576 mice, a validated model of AD. The impact of PEA on amyloid precursor protein (APP) processing, astrocytic activation, microglial reactivity and neuroinflammation, nitrosative stress, dendritic spine density in hippocampal CA1 pyramidal neurons, and cognitive performance was assessed. Chronic PEA treatment of Tg2576 mice increased the expression of the α-secretase ADAM9 and reduced astrogliosis. Furthermore, PEA attenuated microglia reactivity, downregulated pro-inflammatory (CXCL13, MCP-1, GCSF) and upregulated anti-inflammatory (CXC3CL1 and IL-9) cytokine expression. Chronic PEA administration also decreased protein nitrosylation, downregulated calcineurin expression, restored dendritic spine density, and improved cognitive functions. Chronic PEA administration offers a promising therapeutic approach for AD by mitigating neuroinflammation, oxidative stress, and synaptic dysfunction, ultimately leading to cognitive function restoration.
Collapse
Affiliation(s)
- Daniel Tortolani
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Davide Decandia
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Lucia Scipioni
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Anna Panuccio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | | | - Fabiola Eugelio
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Livia La Barbera
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Debora Cutuli
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Marcello D’Amelio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
| | - Sergio Oddi
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Laura Petrosini
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
12
|
Qi Y, Jang D, Ryu J, Bai T, Shin Y, Gu W, Iyer A, Li G, Ma H, Liou JY, van der Meer M, Qiang Y, Fang H. Stabilized carbon coating on microelectrodes for scalable and interoperable neurotransmitter sensing. Nat Commun 2025; 16:3300. [PMID: 40195312 PMCID: PMC11977211 DOI: 10.1038/s41467-025-58388-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
Real-time monitoring of neurotransmitters is essential in driving basic neuroscience understandings and creating treatments for various brain disorders. However, current neurotransmitter sensing devices are highly limited in their spatiotemporal resolution and ability to integrate with neuronal recording. Here, we introduce a unique carbon coating approach to achieve high-performance voltammetry electrodes with extraordinary scalability and interoperability. Surprisingly, we discovered that mild annealing drastically improves the electrochemical stability of graphene-based carbon coating, enabling the transformation of conventional neuroelectrodes into fast-scan-cyclic-voltammetry-stable carbon sensors. We successfully validated sub-second detection of nanomolar dopamine in vivo using carbon-coated microelectrodes (CCMs) in rodents and demonstrated arrays of one hundred CCMs with high yield and uniformity. Furthermore, we developed a dual-modal neural probe that integrates the CCM with electrophysiological recording sites, allowing us to demonstrate that dopamine fluctuation in the ventral striatum of awake rats strongly correlates with the high gamma power in the brain with sub-second-level precision. Together, these advances pave the way for spatiotemporally scalable and multiplexed brain interfacing, with also broad applicability in electrochemical-related diagnostic and interventional approaches.
Collapse
Affiliation(s)
- Yongli Qi
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Dongyeol Jang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Jaehyeon Ryu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Tianyu Bai
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Yieljae Shin
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Wen Gu
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, USA
- Lingang Laboratory, Shanghai, China
| | - Aditya Iyer
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Gen Li
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Hongtao Ma
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY, USA
| | - Jyun-You Liou
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Yi Qiang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | - Hui Fang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
13
|
Zhang L, Liu Y, Wang X, Wu H, Xie J, Liu Y. Treadmill exercise ameliorates hippocampal synaptic injury and recognition memory deficits by TREM2 in AD rat model. Brain Res Bull 2025; 223:111280. [PMID: 40015348 DOI: 10.1016/j.brainresbull.2025.111280] [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: 06/14/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE The impairment of cognitive function has been associated with Alzheimer's disease (AD). Exercise exerts a positive modulatory effect on cognition by reducing synapse injury. However, limited in vivo evidence is available to validate the neuroprotective effect of TREM2 on synaptic function in this phenomenon. Here, we aim to explore whether physical exercise pretreatment alters Aβ-induced recognition memory impairment in structural synaptic plasticity within the hippocampus in AD rats. METHODS:: In study 1, fifty-two Sprague-Dawley (SD) rats were randomly divided into following four groups: control group (C group, n = 13), Alzheimer's disease group (AD group, n = 13), 4 weeks of physical exercise and Alzheimer's disease group (Exe+AD group, n = 13), 4 weeks of physical exercise and blank group (Exercise group, n = 13). Four weeks of treadmill exercise intervention was performed, and AD model were established by intra-cerebroventricular injection (ICV) injection of Aβ1-42 protein. After 3 weeks, we also conducted a novel object test to evaluate recognition memory in the behavior assessment. Golgi staining and transmission electron microscopy were used to evaluate the morphology and synaptic ultrastructure of neurons. Western blotting was used to measure the expression of hippocampal synaptic proteins. Extracellular neurotransmitters in the hippocampus were detected by microdialysis coupled with high-performance liquid chromatography. In study 2, 33 SD rats were randomly divided into three groups: 4 weeks of physical exercise and Alzheimer's disease group (Exe+AD group, n = 11), AAV-Control and physical exercise and Alzheimer's disease group (AAV-Control+Exe+AD group, n = 11), AAV-TREM2 and physical exercise and Alzheimer's disease group (AAV-TREM2 +Exe+AD group, n = 11). Stereotactic intracerebral injection in the bilateral hippocampus was performed to achieve microglial TREM2 down-expression by using adeno-associated virus (AAV) with CD68 promoter. After 4 weeks treadmill exercise and 3 weeks Aβ injection, all rats received behavior test and molecular experiment, which the same with experiment 2. RESULTS Novel recognition index in novel object recognition test significantly decreased, and western blot demonstrate that hippocampal TREM2 protein is significantly decreased (P < 0.001). But physical exercise reversed this phenomenon(P < 0.001). In addition, compared with Con group, the neuron from Exe+AD group exhibited a more complex branching pattern (P < 0.05). And impaired synaptic ultrastructure was observed in AD group. Hippocampal synaptic-related protein (SYX, SYP, GAP43, PSD95) and neurotransmitter (DA, Glu, GABA) was also significantly decreased (P < 0.01) in AD group. But the neuroprotection effect can be found in Exe+AD group, which are associated with the inhibition of synaptic injury by activate hippocampal TREM2 (P < 0.05). However, when blockade of hippocampal TREM2 reduced brain protective effect of exercise in AD rat model, including increased the damage of neuronal dendritic complexity, synaptic ultrastructure, and the decrease of hippocampal synapses-related protein, typical neurotransmitter. CONCLUSION Treadmill exercise facilitated recognition memory acquisition via TREM2-mediated structural synaptic plasticity of the hippocampus in an AD rat model.
Collapse
Affiliation(s)
- Linlin Zhang
- Department of physical education, Henan normal university, Xinxiang 453007, China; Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Yanzhong Liu
- School of physical education and health, Henan University of China Medicine, Zhengzhou, China
| | - Xin Wang
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Hao Wu
- Comprehensive Key Laboratory of Sports Ability Evaluation and Research of the General Administration of Sport of China, Beijing Key Laboratory of Sports Function Assessment and Technical Analysis, Capital University of Physical Education and Sports, Beijing 100191, China
| | - Jiahui Xie
- Department of Physical Education and Research, Fuzhou University, Fuzhou 350108, China.
| | - Yiping Liu
- Provincial University Key Laboratory of Sport and Health Science, School of Physical Education and Sport Sciences, Fujian Normal University, Fuzhou 350007, China.
| |
Collapse
|
14
|
Xiong Y, Guo J, Yu W, Zeng D, Song C, Zhou L, Anatolyevna NL, Baranenko D, Xiao D, Zhou Y, Lu W. Molecular Mechanism of Microgravity-Induced Intestinal Flora Dysbiosis on the Abnormalities of Liver and Brain Metabolism. Int J Mol Sci 2025; 26:3094. [PMID: 40243802 PMCID: PMC11988970 DOI: 10.3390/ijms26073094] [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: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Space flight has many adverse effects on the physiological functions of astronauts. Certain similarities have been observed in some physiological processes of rodents and astronauts in space, although there are also differences. These similarities make rodents helpful models for initial investigations into space-induced physiological changes. This study uses a 3D-Clinostat to simulate microgravity and explores the role of microgravity in space flight-induced liver and brain abnormalities by comparing changes in the gut microbiota, serum metabolites, and the function and physiological biochemistry of liver and brain tissues between the simulated microgravity (SMG) group mice and the wild type (WT) group mice. The study, based on hematoxylin-eosin (HE) staining, 16S sequencing technology, and non-targeted metabolomics analysis, shows that the gut tissue morphology of the SMG group mice is abnormal, and the structure of the gut microbiota and the serum metabolite profile are imbalanced. Furthermore, using PICRUST 2 technology, we have predicted the functions of the gut microbiota and serum metabolites, and the results indicate that the liver metabolism and functions (including lipid metabolism, amino acid metabolism, and sugar metabolism, etc.) of the SMG group mice are disrupted, and the brain tissue metabolism and functions (including neurotransmitters and hormone secretion, etc.) are abnormal, suggesting a close relationship between microgravity and liver metabolic dysfunction and brain dysfunction. Additionally, the high similarity in the structure of the gut microbiota and serum metabolite profile between the fecal microbiota transplant (FMT) group mice and the SMG group mice, and the physiological and biochemical differences in liver and brain tissues compared to the WT group mice, suggest that microgravity induces imbalances in the gut microbiota, which in turn triggers abnormalities in liver and brain metabolism and function. Finally, through MetaMapp analysis and Pearson correlation analysis, we found that valeric acid, a metabolite of gut microbiota, is more likely to be the key metabolite that relates to microgravity-induced gut microbiota abnormalities, disorders of amino acid and lipid metabolism, and further induced metabolic or functional disorders in the liver and brain. This study has significant practical application value for deepening the understanding of the adaptability of living organisms in the space environment.
Collapse
Affiliation(s)
- Yi Xiong
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; (Y.X.)
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Jianguo Guo
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Wenchen Yu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China; (Y.X.)
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Deyong Zeng
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Chenchen Song
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Li Zhou
- National Human Diseases Animal Model Resource Center, National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Comparative Medicine, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Nadtochii Liudmila Anatolyevna
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
- School of Life Sciences, International Research Centre Biotechnologies of the Third Millennium, ITMO University, St. Petersburg 197101, Russia
| | - Denis Baranenko
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
- School of Life Sciences, International Research Centre Biotechnologies of the Third Millennium, ITMO University, St. Petersburg 197101, Russia
| | - Dan Xiao
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Yingyu Zhou
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Weihong Lu
- National and Local Joint Engineering Laboratory for Synthesis, Harbin Institute of Technology, Harbin 150001, China
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| |
Collapse
|
15
|
Zhang X, Yan F, He XJ, Chen Y, Gu R, Dong X, Wei Y, Bai L, Bai J. Thioredoxin-1 Downregulation in the SNpc Exacerbates the Cognitive Impairment Induced by MPTP. Antioxid Redox Signal 2025. [PMID: 40135707 DOI: 10.1089/ars.2024.0630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Aims: Parkinson's disease (PD) is characterized by dopaminergic (DAergic) neuron degeneration in the substantia nigra pars compacta (SNpc). Thioredoxin-1 (Trx-1) is a redox protein that protects neurons from various injuries. Our study revealed that Trx-1 overexpression improved the learning and memory impairments induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, the role of the specific transmission of signals from the SNpc to the hippocampus regulated by Trx-1 in cognition deficits associated with PD is still unknown. Results: We observed that Trx-1 downregulation in the SNpc aggravated cognitive dysfunction induced by MPTP. Importantly, we observed that the SNpc directly projects to the hippocampus. We found that the loss of DAergic neurons in the SNpc induced by MPTP resulted in a decrease in dopamine D1 receptor (D1R) expression in the hippocampus, which was promoted by Trx-1 downregulation in the SNpc. The levels of phosphorylated extracellular signal-regulated kinase (p-ERK1/2), phosphorylated cAMP-response element binding protein (p-CREB), brain-derived neurotrophic factor (BDNF), and postsynaptic density protein 95 (PSD95) in the hippocampus were decreased by MPTP and further decreased by Trx-1 downregulation in the SNpc. Finally, the number of synapses in the hippocampus was decreased by MPTP in the hippocampus and further reduced by Trx-1 downregulation in the SNpc. Innovation: Trx-1 downregulation accelerated the loss of DAergic neurons in the SNpc, leading to a decrease in the number dopaminergic projections to the hippocampus, subsequently inhibiting the D1R-ERK1/2-CREB-BDNF pathway in the hippocampus, and ultimately impairing hippocampus-dependent cognition. Conclusions: These results indicate that a decrease in Trx-1 level in the SNpc plays a critical regulatory role in cognitive dysfunction in individuals with PD by decreasing the hippocampal D1R signaling pathway. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Xianwen Zhang
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Fang Yan
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xiong Jie He
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Yali Chen
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Rou Gu
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Xianghuan Dong
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Yonghang Wei
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Liping Bai
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| | - Jie Bai
- Medical Faculty, Laboratory of Molecular Neurobiology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
16
|
Fuchsberger T, Stockwell I, Woods M, Brzosko Z, Greger IH, Paulsen O. Dopamine increases protein synthesis in hippocampal neurons enabling dopamine-dependent LTP. eLife 2025; 13:RP100822. [PMID: 40063079 PMCID: PMC11893101 DOI: 10.7554/elife.100822] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
The reward and novelty-related neuromodulator dopamine plays an important role in hippocampal long-term memory, which is thought to involve protein-synthesis-dependent synaptic plasticity. However, the direct effects of dopamine on protein synthesis, and the functional implications of newly synthesised proteins for synaptic plasticity, have not yet been investigated. We have previously reported that timing-dependent synaptic depression (t-LTD) can be converted into potentiation by dopamine application during synaptic stimulation (Brzosko et al., 2015) or postsynaptic burst activation (Fuchsberger et al., 2022). Here, we show that dopamine increases protein synthesis in mouse hippocampal CA1 neurons, enabling dopamine-dependent long-term potentiation (DA-LTP), which is mediated via the Ca2+-sensitive adenylate cyclase (AC) subtypes 1/8, cAMP, and cAMP-dependent protein kinase (PKA). We found that neuronal activity is required for the dopamine-induced increase in protein synthesis. Furthermore, dopamine induced a protein-synthesis-dependent increase in the AMPA receptor subunit GluA1, but not GluA2. We found that DA-LTP is absent in GluA1 knock-out mice and that it requires calcium-permeable AMPA receptors. Taken together, our results suggest that dopamine together with neuronal activity controls synthesis of plasticity-related proteins, including GluA1, which enable DA-LTP via a signalling pathway distinct from that of conventional LTP.
Collapse
Affiliation(s)
- Tanja Fuchsberger
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Matty Woods
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Zuzanna Brzosko
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, Physiological Laboratory, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
17
|
Liu S, Li X, Jiao S, Zheng Y, Xia L, Figueredo YN, Liu K, Jin M. Phenyl salicylate induces neurotoxicity and early Alzheimer's disease-like symptoms through ndrg1-regulated myelin damage, increasing bace1 in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 965:178664. [PMID: 39893810 DOI: 10.1016/j.scitotenv.2025.178664] [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: 08/09/2024] [Revised: 12/10/2024] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Phenyl salicylate, an important industrial raw material, is widely used in plastics, cosmetics, and pharmaceuticals. However, little is known about its neurotoxicity on wildlife. Here, we exposed zebrafish embryos at 4 hours post-fertilization (hpf) to 0.025, 0.05, 0.1, 0.25, 0.5, and 1.0 mg/L of phenyl salicylate up to 144 hpf and found its developmental- and neuro-toxicity. Specifically, a dose-dependent increase in mortality and malformation in zebrafish were revealed. Phenyl salicylate also adversely affected the development of monoaminergic neurons, cerebral blood vessels, and the blood-brain barrier (BBB), as well as induced cerebral hemorrhages and locomotion change. RNA-sequencing results combined with verification data showed that phenyl salicylate downregulated the expression of the N-myc downstream regulated gene-1 (ndrg1), caused myelin damage in zebrafish, and then increased expression of beta-secretase 1 (bace1), which ultimately led to early Alzheimer's disease (AD)-like symptoms, including BBB leakage, bleeding in the brain, and upregulation of the glial fibrillary acidic protein gene (gfap) and cholinergic system-related gene (chrna7a). In conclusion, phenyl salicylate exposure triggered developmental toxicity and neurotoxicity in zebrafish, which has a potential risk for the development of AD. Given the effects of phenyl salicylate exposure to ecosystem, the safety usage limit should be treated with caution.
Collapse
Affiliation(s)
- Siyu Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Xinjia Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Shouqing Jiao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Yuanteng Zheng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Lijie Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Yanier Nuñez Figueredo
- Centro de Investigación y Desarrollo de Medicamentos (CIDEM), Ave 26, No.1605, e/Boyeros y Puentes Grandes, La Habana CP10600, Cuba
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan 250103, Shandong Province, People's Republic of China.
| |
Collapse
|
18
|
Song J, Cho E, Lee H, Lee S, Kim S, Kim J. Development of Neurodegenerative Disease Diagnosis and Monitoring from Traditional to Digital Biomarkers. BIOSENSORS 2025; 15:102. [PMID: 39997004 PMCID: PMC11852611 DOI: 10.3390/bios15020102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
Monitoring and assessing the progression of symptoms in neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are critical for improving patient outcomes. Traditional biomarkers, such as cerebrospinal fluid analysis and brain imaging, are widely used to investigate the underlying mechanisms of disease and enable early diagnosis. In contrast, digital biomarkers derived from phenotypic changes-such as EEG, eye movement, gait, and speech analysis-offer a noninvasive and accessible alternative. Leveraging portable and widely available devices, such as smartphones and wearable sensors, digital biomarkers are emerging as a promising tool for ND diagnosis and monitoring. This review highlights the comprehensive developments in digital biomarkers, emphasizing their unique advantages and integration potential alongside traditional biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinsik Kim
- Department of Biomedical Engineering, College of Life Science and Biotechnology, Dongguk University, Seoul 04620, Republic of Korea; (J.S.); (E.C.); (H.L.); (S.L.); (S.K.)
| |
Collapse
|
19
|
Liu Y, Xu X, Wu X, Yang G, Luo J, Liang X, Chen J, Li Y. TMF Attenuates Cognitive Impairment and Neuroinflammation by Inhibiting the MAPK/NF-κB Pathway in Alzheimer's Disease: A Multi-Omics Analysis. Mar Drugs 2025; 23:74. [PMID: 39997198 PMCID: PMC11857128 DOI: 10.3390/md23020074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
The rising prevalence of Alzheimer's disease (AD) underscores the urgent need for novel therapeutic agents derived from natural sources. Among flavonoids, 3',4',5,7-tetramethoxyflavone (TMF), a structural analog of luteolin, has gained attention for its favorable pharmacokinetics and potential neuroprotective properties. Despite the significant neuroprotective effects and favorable pharmacokinetics of TMF, its efficacy and mechanism of action in AD remain unclear. This study explored TMF's pharmacological effects in AD models, highlighting its ability to improve memory and cognitive deficits in APP/PS1 mice. TMF reduced Aβ plaques, NFTs formation, and glial activation while suppressing neuroinflammation through the MAPK/NF-κB pathway. Further analysis in LPS-induced BV2 cells revealed TMF's ability to reduce microglial activation. These findings highlight the anti-neuroinflammatory activity of TMF, suggesting its potential as a treatment for AD.
Collapse
Affiliation(s)
- Yonglin Liu
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| | - Xi Xu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Xiaoming Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Guodong Yang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Jiaxin Luo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Xinli Liang
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| | - Jie Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
| | - Yiguang Li
- National Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang 330096, China; (Y.L.); (X.L.)
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (X.X.); (X.W.); (G.Y.); (J.L.)
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang 330103, China
| |
Collapse
|
20
|
Zaccone C, Nobili A, D’Amelio M. Decoding Alzheimer's disease: acetylcholine and dopamine pathway disruptions as early markers of cognitive decline. Brain Commun 2025; 7:fcaf057. [PMID: 39980741 PMCID: PMC11840162 DOI: 10.1093/braincomms/fcaf057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
This scientific commentary refers to 'Changes in neurotransmitter-related functional connectivity along the Alzheimer's disease continuum', by Manca et al. (https://doi.org/10.1093/braincomms/fcaf008).
Collapse
Affiliation(s)
- Claudio Zaccone
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Marcello D’Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| |
Collapse
|
21
|
Chib S, Dutta BJ, Chalotra R, Abubakar M, Kumar P, Singh TG, Singh R. Role of Flavonoids in Mitigating the Pathological Complexities and Treatment Hurdles in Alzheimer's Disease. Phytother Res 2025; 39:747-775. [PMID: 39660432 DOI: 10.1002/ptr.8406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
With the passage of time, people step toward old age and become more prone to several diseases associated with the age. One such is Alzheimer's disease (AD) which results into neuronal damage and dementia with the progression of age. The existing therapeutics has been hindered by various enkindles like less eminent between remote populations, affordability issues and toxicity profiles. Moreover, lack of suitable therapeutic option further worsens the quality of life in older population. Developing an efficient therapeutic intervention to cure AD is still a challenge for medical fraternity. Recently, alternative approaches attain the attention of researchers to focus on plant-based therapy in mitigating AD. In this context, flavonoids gained centrality as a feasible treatment in modifying various neurological deficits. This review mainly focuses on the pathological facets and economic burden of AD. Furthermore, we have explored the possible mechanism of flavonoids with the preclinical and clinical aspects for curing AD. Flavonoids being potential therapeutic, target the pathogenic factors of AD such as oxidative stress, inflammation, metal toxicity, Aβ accumulation, modulate neurotransmission and insulin signaling. In this review, we emphasized on potential neuroprotective effects of flavonoids in AD pathology, with focus on both experimental and clinical findings. While preclinical studies suggest promising therapeutic benefits, clinical data remains limited and inconclusive. Thus, further high-quality clinical trials are necessary to validate the efficacy of flavonoids in AD. The study aim is to promote the plant-based therapies and encourage people to add flavonoids to regular diet to avail the beneficial effects in preventive therapy for AD.
Collapse
Affiliation(s)
- Shivani Chib
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Bhaskar Jyoti Dutta
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Rishabh Chalotra
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Md Abubakar
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | | | - Randhir Singh
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
22
|
Akyuz E, Arulsamy A, Aslan FS, Sarisözen B, Guney B, Hekimoglu A, Yilmaz BN, Retinasamy T, Shaikh MF. An Expanded Narrative Review of Neurotransmitters on Alzheimer's Disease: The Role of Therapeutic Interventions on Neurotransmission. Mol Neurobiol 2025; 62:1631-1674. [PMID: 39012443 PMCID: PMC11772559 DOI: 10.1007/s12035-024-04333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. The accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles are the key players responsible for the pathogenesis of the disease. The accumulation of Aβ plaques and tau affect the balance in chemical neurotransmitters in the brain. Thus, the current review examined the role of neurotransmitters in the pathogenesis of Alzheimer's disease and discusses the alterations in the neurochemical activity and cross talk with their receptors and transporters. In the presence of Aβ plaques and neurofibrillary tangles, changes may occur in the expression of neuronal receptors which in turn triggers excessive release of glutamate into the synaptic cleft contributing to cell death and neuronal damage. The GABAergic system may also be affected by AD pathology in a similar way. In addition, decreased receptors in the cholinergic system and dysfunction in the dopamine neurotransmission of AD pathology may also contribute to the damage to cognitive function. Moreover, the presence of deficiencies in noradrenergic neurons within the locus coeruleus in AD suggests that noradrenergic stimulation could be useful in addressing its pathophysiology. The regulation of melatonin, known for its effectiveness in enhancing cognitive function and preventing Aβ accumulation, along with the involvement of the serotonergic system and histaminergic system in cognition and memory, becomes remarkable for promoting neurotransmission in AD. Additionally, nitric oxide and adenosine-based therapeutic approaches play a protective role in AD by preventing neuroinflammation. Overall, neurotransmitter-based therapeutic strategies emerge as pivotal for addressing neurotransmitter homeostasis and neurotransmission in the context of AD. This review discussed the potential for neurotransmitter-based drugs to be effective in slowing and correcting the neurodegenerative processes in AD by targeting the neurochemical imbalance in the brain. Therefore, neurotransmitter-based drugs could serve as a future therapeutic strategy to tackle AD.
Collapse
Affiliation(s)
- Enes Akyuz
- Department of Biophysics, International School of Medicine, University of Health Sciences, Istanbul, Turkey
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
| | | | - Bugra Sarisözen
- School of Medicine, Tekirdağ Namık Kemal University, Tekirdağ, Turkey
| | - Beyzanur Guney
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | | | - Beyza Nur Yilmaz
- International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Bandar Sunway, Selangor, Malaysia.
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, New South Wales, 2800, Australia.
| |
Collapse
|
23
|
Miao HT, Wang J, Shao JJ, Song RX, Li WG, Sun JK, Jia SY, Zhang DX, Li XM, Zhao JY, Zhang LM. Astrocytic NLRP3 cKO mitigates depression-like behaviors induced by mild TBI in mice. Neurobiol Dis 2025; 205:106785. [PMID: 39793767 DOI: 10.1016/j.nbd.2024.106785] [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: 08/13/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Reports indicate that depression is a common mental health issue following traumatic brain injury (TBI). Our prior research suggests that Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-related neuroinflammation, modulated by glial cells such as astrocytes, is likely to play a crucial role in the progression of anxiety and cognitive dysfunction. However, there is limited understanding of the potential of astrocytic NLRP3 in treating depression under mild TBI condition. This study aimed to determine whether astrocytic NLRP3 knockout (KO) could mitigate depressive-like behaviors following mild TBI and explore potential variations in such behaviors between genders post-mild TBI. METHODS Mild TBI was induced in mice using Feeney's weight-drop method. Behavioral assessments included neurological severity scores (NSS), social interaction test (SI), tail suspension test (TST), and forced swimming test (FST). Pathological changes were evaluated through immunofluorescence and local field potential (LFP) recordings at various time points post-injury. RESULTS Our findings indicated that astrocyte-specific NLRP3 KO decreased cleaved caspase-1 colocalized with astrocytes, decreased pathogenic astrocytes and increased Postsynaptic density protein 95 (PSD95) intensity, and significantly alleviated mild TBI-induced depression-like behaviors. It also led to the upregulation of protective astrocytes and apoptosis-associated factors, including cleaved caspase-3 post-mild TBI. Additionally, astrocyte-specific NLRP3 deletion resulting in improved θ and γ power and θ-γ phase coupling in the social interaction test (SI). Notably, under mild TBI conditions, astrocyte-specific NLRP3 exhibited greater neuroprotective effects in female knockout mice compared to males. CONCLUSION Astrocyte NLRP3 knockout demonstrated a protective mechanism in mice subjected to mild TBI, possibly attributed to the inhibition of pyroptosis through the NLRP3 signaling pathway in astrocytes.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China; Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jun Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jing-Jing Shao
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Wen-Guang Li
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Jian-Kai Sun
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Ming Li
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jian-Yong Zhao
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,.
| |
Collapse
|
24
|
Calvin-Dunn KN, Mcneela A, Leisgang Osse A, Bhasin G, Ridenour M, Kinney JW, Hyman JM. Electrophysiological insights into Alzheimer's disease: A review of human and animal studies. Neurosci Biobehav Rev 2025; 169:105987. [PMID: 39732222 DOI: 10.1016/j.neubiorev.2024.105987] [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: 01/22/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
This review highlights the crucial role of neuroelectrophysiology in illuminating the mechanisms underlying Alzheimer's disease (AD) pathogenesis and progression, emphasizing its potential to inform the development of effective treatments. Electrophysiological techniques provide unparalleled precision in exploring the intricate networks affected by AD, offering insights into the synaptic dysfunction, network alterations, and oscillatory abnormalities that characterize the disease. We discuss a range of electrophysiological methods, from non-invasive clinical techniques like electroencephalography and magnetoencephalography to invasive recordings in animal models. By drawing on findings from these studies, we demonstrate how electrophysiological research has deepened our understanding of AD-related network disruptions, paving the way for targeted therapeutic interventions. Moreover, we underscore the potential of electrophysiological modalities to play a pivotal role in evaluating treatment efficacy. Integrating electrophysiological data with clinical neuroimaging and longitudinal studies holds promise for a more comprehensive understanding of AD, enabling early detection and the development of personalized treatment strategies. This expanded research landscape offers new avenues for unraveling the complexities of AD and advancing therapeutic approaches.
Collapse
Affiliation(s)
- Kirsten N Calvin-Dunn
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, United States.
| | - Adam Mcneela
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States
| | - A Leisgang Osse
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - G Bhasin
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| | - M Ridenour
- Department of Psychology, University of Nevada, Las Vegas, United States
| | - J W Kinney
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - J M Hyman
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| |
Collapse
|
25
|
Bobkova NV, Chuvakova LN, Kovalev VI, Zhdanova DY, Chaplygina AV, Rezvykh AP, Evgen'ev MB. A Mouse Model of Sporadic Alzheimer's Disease with Elements of Major Depression. Mol Neurobiol 2025; 62:1337-1358. [PMID: 38980563 DOI: 10.1007/s12035-024-04346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
After olfactory bulbectomy, animals are often used as a model of major depression or sporadic Alzheimer's disease and, hence, the status of this model is still disputable. To elucidate the nature of alterations in the expression of the genome after the operation, we analyzed transcriptomes of the cortex, hippocampus, and cerebellum of the olfactory bulbectomized (OBX) mice. Analysis of the functional significance of genes in the brain of OBX mice indicates that the balance of the GABA/glutamatergic systems is disturbed with hyperactivation of the latter in the hippocampus, leading to the development of excitotoxicity and induction of apoptosis in the background of severe mitochondrial dysfunction and astrogliosis. On top of this, the synthesis of neurotrophic factors decreases leading to the disruption of the cytoskeleton of neurons, an increase in the level of intracellular calcium, and the activation of tau protein hyperphosphorylation. Moreover, the acetylcholinergic system is deficient in the background of the hyperactivation of acetylcholinesterase. Importantly, the activity of the dopaminergic, endorphin, and opiate systems in OBX mice decreases, leading to hormonal dysfunction. On the other hand, genes responsible for the regulation of circadian rhythms, cell migration, and innate immunity are activated in OBX animals. All this takes place in the background of a drastic downregulation of ribosomal protein genes in the brain. The obtained results indicate that OBX mice represent a model of Alzheimer's disease with elements of major depression.
Collapse
Affiliation(s)
- N V Bobkova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - L N Chuvakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - V I Kovalev
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - D Y Zhdanova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A V Chaplygina
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A P Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - M B Evgen'ev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
| |
Collapse
|
26
|
Zeng HX, Qin SJ, Andersson J, Li SP, Zeng QG, Li JH, Wu QZ, Meng WJ, Oudin A, Kanninen KM, Jalava P, Dong GH, Zeng XW. The emerging roles of particulate matter-changed non-coding RNAs in the pathogenesis of Alzheimer's disease: A comprehensive in silico analysis and review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125440. [PMID: 39631655 DOI: 10.1016/j.envpol.2024.125440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Research on epigenetic‒environmental interactions in the development of Alzheimer's disease (AD) has accelerated rapidly in recent decades. Numerous studies have demonstrated the contribution of ambient particulate matter (PM) to the onset of AD. Emerging evidence indicates that non-coding RNAs (ncRNAs), including long non-coding RNAs, circular RNAs, and microRNAs, play a role in the pathophysiology of AD. In this review, we provide an overview of PM-altered ncRNAs in the brain, with emphasis on their potential roles in the pathogenesis of AD. These results suggest that these PM-altered ncRNAs are involved in the regulation of amyloid-beta pathology, microtubule-associated protein Tau pathology, synaptic dysfunction, damage to the blood‒brain barrier, microglial dysfunction, dysmyelination, and neuronal loss. In addition, we utilized in silico analysis to explore the biological functions of PM-altered ncRNAs in the development of AD. This review summarizes the knowns and unknowns of PM-altered ncRNAs in AD pathogenesis and discusses the current dilemma regarding PM-altered ncRNAs as promising biomarkers of AD. Altogether, this is the first thorough review of the connection between PM exposure and ncRNAs in AD pathogenesis, which may offer novel insights into the prevention, diagnosis, and treatment of AD associated with ambient PM exposure.
Collapse
Affiliation(s)
- Hui-Xian Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang-Jian Qin
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | | | - Shen-Pan Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Guo Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia-Hui Li
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qi-Zhen Wu
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wen-Jie Meng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Anna Oudin
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Katja M Kanninen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Pasi Jalava
- Department of Environmental and Biological Science, University of Eastern Finland, Kuopio, Finland
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Wen Zeng
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
27
|
Ji Y, Yang C, Pang X, Yan Y, Wu Y, Geng Z, Hu W, Hu P, Wu X, Wang K. Repetitive transcranial magnetic stimulation in Alzheimer's disease: effects on neural and synaptic rehabilitation. Neural Regen Res 2025; 20:326-342. [PMID: 38819037 PMCID: PMC11317939 DOI: 10.4103/nrr.nrr-d-23-01201] [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: 07/19/2023] [Revised: 10/23/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Alzheimer's disease is a neurodegenerative disease resulting from deficits in synaptic transmission and homeostasis. The Alzheimer's disease brain tends to be hyperexcitable and hypersynchronized, thereby causing neurodegeneration and ultimately disrupting the operational abilities in daily life, leaving patients incapacitated. Repetitive transcranial magnetic stimulation is a cost-effective, neuro-modulatory technique used for multiple neurological conditions. Over the past two decades, it has been widely used to predict cognitive decline; identify pathophysiological markers; promote neuroplasticity; and assess brain excitability, plasticity, and connectivity. It has also been applied to patients with dementia, because it can yield facilitatory effects on cognition and promote brain recovery after a neurological insult. However, its therapeutic effectiveness at the molecular and synaptic levels has not been elucidated because of a limited number of studies. This study aimed to characterize the neurobiological changes following repetitive transcranial magnetic stimulation treatment, evaluate its effects on synaptic plasticity, and identify the associated mechanisms. This review essentially focuses on changes in the pathology, amyloidogenesis, and clearance pathways, given that amyloid deposition is a major hypothesis in the pathogenesis of Alzheimer's disease. Apoptotic mechanisms associated with repetitive transcranial magnetic stimulation procedures and different pathways mediating gene transcription, which are closely related to the neural regeneration process, are also highlighted. Finally, we discuss the outcomes of animal studies in which neuroplasticity is modulated and assessed at the structural and functional levels by using repetitive transcranial magnetic stimulation, with the aim to highlight future directions for better clinical translations.
Collapse
Affiliation(s)
- Yi Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Chaoyi Yang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Xuerui Pang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yibing Yan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Yue Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhi Geng
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Wenjie Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei, Anhui Province, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, Anhui Province, China
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, Anhui Province, China
- Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
28
|
Chang X, Tse AM, Fayzullina M, Albanese A, Kim M, Wang CF, Zheng Z, Joshi RV, Williams CK, Magaki SD, Vinters HV, Jones JO, Haworth IS, Seidler PM. Monoaminergic neurotransmitters are bimodal effectors of tau aggregation. SCIENCE ADVANCES 2025; 11:eadr8055. [PMID: 39888993 PMCID: PMC11784839 DOI: 10.1126/sciadv.adr8055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/31/2024] [Indexed: 02/02/2025]
Abstract
Neurotransmitters (NTs) mediate trans-synaptic signaling, and disturbances in their levels are linked to aging and brain disorders. Here, we ascribe an additional function for NTs in mediating intracellular protein aggregation by interaction with cytosolic protein fibrils. Cell-based seeding experiments revealed monoaminergic NTs as inhibitors of tau. Seeding is a disease-relevant mechanism involving catalysis by fibrils, leading to the aggregation of proteins in Alzheimer's disease and other neurodegenerative diseases. Chemotyping small molecules with varied backbone structures revealed determinants of aggregation inhibitors and catalysts. Among those identified were monoaminergic NTs. Dose titrations revealed bimodal effects indicative of fibril disaggregation, with aggregation catalysis occurring at low ratios of NTs and inhibited seeding ensuing at higher concentrations. Bimodal effects by NTs extend from in vitro systems to dopaminergic neurons, suggesting that pharmacotherapies that modify intracellular NT levels could shape the neuronal protein aggregation environment.
Collapse
Affiliation(s)
- Xinmin Chang
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Amanda M. Tse
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Marina Fayzullina
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Angela Albanese
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Department of Medical Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Minchan Kim
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Conner F. Wang
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Zipeng Zheng
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Ruchira V. Joshi
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher K. Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shino D. Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Harry V. Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Brain Research Institute, David Geffen UCLA School of Medicine, Los Angeles, CA 90095, USA
| | - Jeremy O. Jones
- Simulations Plus Inc., 42505 10th Street West, Lancaster, CA 93534-7059, USA
| | - Ian S. Haworth
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul M. Seidler
- Department of Pharmacology and Pharmaceutical Sciences, USC Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
29
|
Pilotto A, Galli A, Sala A, Caminiti SP, Presotto L, Liguori C, Mercuri NB, Premi E, Garibotto V, Frisoni G, Chiaravalloti A, Schillaci O, D'Amelio M, Paghera B, Lucchini S, Bertagna F, Perani D, Padovani A. Dopaminergic deficits along the spectrum of Alzheimer's disease. Mol Psychiatry 2025:10.1038/s41380-025-02913-5. [PMID: 39890920 DOI: 10.1038/s41380-025-02913-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Both post-mortem and in vivo data argue for dopamine dysfunction in patients with Alzheimer's Disease (AD). However, the timing and regional progression of dopaminergic systems alterations in AD are still debated. The aim of the study was to investigate in vivo the pattern of dopaminergic changes and connectivity using DAT-SPECT imaging in patients across the AD spectrum. Fifty-nine AD patients (n = 21 AD-MCI; n = 38 AD-DEM) and a control group (CG) of n = 45 age- and sex-matched individuals entered the study and underwent 123I-FP-CIT dopaminergic imaging. The occipital binding was used as reference region to obtain single-subject binding in different brain regions. Between-group differences in 123I-FP-CIT binding in both mesolimbic and nigrostriatal dopaminergic pathways were assessed using an ANCOVA test, adjusting for the effect of center of imaging acquisition, age, and sex. Regions resulting from the voxel-wise direct comparison between AD-MCI and AD-DEM were considered as a seed of interest for a voxel-wise interregional correlation analysis. Both AD-MCI and AD-DEM patients showed dopaminergic depletion within the basal ganglia, whereas cortico-limbic regions (namely hippocampus, amygdala, anterior and middle cingulate, frontal cortex and thalamus) resulted impaired only in the dementia phase. The brain voxel-wise interregional correlation analysis showed a progressive pattern of disruption of caudate/thalamus dopaminergic connectivity to hippocampus and amygdala from AD-MCI to AD-DEM stages. This study indicates basal ganglia dopaminergic alterations and connectivity disruption in the nigrostriatal and mesolimbic systems already in early stage AD, extending to several cortico-limbic regions in dementia phases.
Collapse
Affiliation(s)
- Andrea Pilotto
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy.
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy.
- Neurology Unit, Department of continuity of care and frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy.
- Neurobiorepository and Laboratory of advanced biological markers, University of Brescia and ASST Spedali Civili Hospital, Brescia, Italy.
| | - Alice Galli
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
| | - Arianna Sala
- Coma Science group- University of Liege, Liège, Belgium
| | | | - Luca Presotto
- Department of Physics "G. Occhialini", University of Milano-Bicocca, Milan, Italy
| | - Claudio Liguori
- Neurophysiology Unit, Sleep and Epilepsy Center- University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurophysiology Unit, Sleep and Epilepsy Center- University of Rome Tor Vergata, Rome, Italy
| | - Enrico Premi
- Stroke Unit, ASST Spedali Civili Brescia Hospital, Brescia, Italy
| | - Valentina Garibotto
- Department of Radiology and Medical Informatics- Geneva University Hospital, Geneva, Switzerland
| | - Giovanni Frisoni
- Department of Psychiatry- Geneva University Hospital, Geneva, Switzerland
| | | | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Barbara Paghera
- Nuclear Medicine Unit- University of Brescia, Brescia, Italy
| | - Silvia Lucchini
- Nuclear Medicine Unit- University of Brescia, Brescia, Italy
| | | | - Daniela Perani
- University Vita-Salute San Raffaele, IRCCS San Raffaele Hospital, Milan, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Unit, University of Brescia, Brescia, Italy
- Laboratory of Digital Neurology and Biosensors, University of Brescia, Brescia, Italy
- Neurology Unit, Department of continuity of care and frailty, ASST Spedali Civili Brescia Hospital, Brescia, Italy
- Brain Health Center, University of Brescia, Brescia, Italy
| |
Collapse
|
30
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2025; 21:01300535-990000000-00688. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
ABSTRACT For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
31
|
Yoo J, Lee J, Ahn B, Han J, Lim MH. Multi-target-directed therapeutic strategies for Alzheimer's disease: controlling amyloid-β aggregation, metal ion homeostasis, and enzyme inhibition. Chem Sci 2025; 16:2105-2135. [PMID: 39810997 PMCID: PMC11726323 DOI: 10.1039/d4sc06762b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative dementia, marked by progressive cognitive decline and memory impairment. Despite advances in therapeutic research, single-target-directed treatments often fall short in addressing the complex, multifactorial nature of AD. This arises from various pathological features, including amyloid-β (Aβ) aggregate deposition, metal ion dysregulation, oxidative stress, impaired neurotransmission, neuroinflammation, mitochondrial dysfunction, and neuronal cell death. This review illustrates their interrelationships, with a particular emphasis on the interplay among Aβ, metal ions, and AD-related enzymes, such as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), matrix metalloproteinase 9 (MMP9), lysyl oxidase-like 2 (LOXL2), acetylcholinesterase (AChE), and monoamine oxidase B (MAOB). We further underscore the potential of therapeutic strategies that simultaneously inhibit Aβ aggregation and address other pathogenic mechanisms. These approaches offer a more comprehensive and effective method for combating AD, overcoming the limitations of conventional therapies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jimin Lee
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Byeongha Ahn
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
32
|
Vignoli A, Bellomo G, Paoletti FP, Luchinat C, Tenori L, Parnetti L. Studying Alzheimer's disease through an integrative serum metabolomic and lipoproteomic approach. J Transl Med 2025; 23:119. [PMID: 39871333 PMCID: PMC11773822 DOI: 10.1186/s12967-025-06148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most frequent neurodegenerative disorder worldwide. The great variability in disease evolution and the incomplete understanding of the molecular mechanisms underlying AD make it difficult to predict when a patient will convert from prodromal stage to dementia. We hypothesize that metabolic alterations present at the level of the brain could be reflected at a systemic level in blood serum of patients, and that these alterations could be used as prognostic biomarkers. METHODS This pilot study proposes a serum investigation via nuclear magnetic resonance (NMR) spectroscopy in a consecutive series of AD patients including 57 patients affected by Alzheimer's disease at dementia stage (AD-dem) and 45 patients with mild cognitive impairment (MCI) due to AD (MCI-AD). As control group, we considered 31 subjects with mild cognitive impairment in whom AD and other neurodegenerative disorders were excluded (MCI). A panel of 26 metabolites and 112 lipoprotein-related parameters was quantified and the logistic LASSO regression algorithm was employed to identify the optimal combination of metabolites-lipoproteins and their ratios to discriminate the groups of interest. RESULTS In the training set, our model classified AD-dem and MCI with an accuracy of 81.7%. These results were reproduced in the validation set (accuracy 75.0%). Evolution of MCI-AD patients was evaluated over time. Patients who displayed a decrease in MMSE < 1.5 point per year were considered at lower progression rate: we obtained a division in 18 MCI-AD at lower progression rate (MCI-AD LR) and 27 at higher progression rate (MCI-AD HR). The model calculated using 4 metabolic features identified MCI-AD LR and MCI-AD HR with an accuracy of 73.3%. CONCLUSIONS The identification of potential novel peripheral biomarkers of Alzheimer's disease, as proposed in this study, opens a new prospect for an innovative and minimally invasive method to identify AD in its very early stages. We proposed a novel approach able to sub-stratify MCI-AD patients identifying those associated with a faster rate of clinical progression.
Collapse
Affiliation(s)
- Alessia Vignoli
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, 50019, Italy.
- Magnetic Resonance Center (CERM/CIRMMP), University of Florence, Sesto Fiorentino, 50019, Italy.
| | - Giovanni Bellomo
- Center for Memory Disturbances, Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, 06129, Italy.
| | - Federico Paolini Paoletti
- Center for Memory Disturbances, Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, 06129, Italy
| | | | - Leonardo Tenori
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, 50019, Italy
- Magnetic Resonance Center (CERM/CIRMMP), University of Florence, Sesto Fiorentino, 50019, Italy
| | - Lucilla Parnetti
- Center for Memory Disturbances, Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, 06129, Italy
| |
Collapse
|
33
|
Haag L, Lancini E, Yakupov R, Ziegler G, Yi YJ, Lüsebrink F, Glanz W, Peters O, Spruth EJ, Altenstein S, Priller J, Schneider LS, Wang X, Preis L, Brosseron F, Roy-Kluth N, Fliessbach K, Wagner M, Wolfsgruber S, Kleineidam L, Ramirez A, Spottke A, Jessen F, Wiltfang J, Schneider A, Hansen N, Rostamzadeh A, Buerger K, Ewers M, Perneczky R, Janowitz D, Rauchmann BS, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Heneka M, Dechent P, Hetzer S, Scheffler K, Düzel E, Betts MJ, Hämmerer D. CSF biomarkers are differentially linked to brain areas high and low in noradrenaline, dopamine and serotonin across the Alzheimer's disease spectrum. Brain Commun 2025; 7:fcaf031. [PMID: 39926613 PMCID: PMC11806415 DOI: 10.1093/braincomms/fcaf031] [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: 06/28/2023] [Revised: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/11/2025] Open
Abstract
Neurotransmitter systems of noradrenaline, dopamine, serotonin and acetylcholine are implicated in cognitive functions such as memory, learning and attention and are known to be altered in neurodegenerative diseases like Alzheimer's disease. Specific brain structures involved in these systems, e.g. the locus coeruleus, the main source of noradrenaline in the cortex, are in fact affected earliest by Alzheimer's disease tau pathology. Preserved volumetric neurotransmitter specific brain areas could therefore be an important neural resource for cognitive reserve in aging. The aim of this study was to determine whether volumes of brain areas known to be high in neurotransmitter receptors are relatively preserved in individuals with lower levels of Alzheimer's disease pathology. Based on the Human Protein Atlas for neurotransmitter receptor distribution, we distinguished between 'areas high and low' in noradrenaline, dopamine, serotonin and acetylcholine and assessed associations of atrophy in those areas with CSF amyloid-ß 42/40, CSF phosphorylated tau protein and cognitive function across healthy controls (n = 122), individuals with subjective cognitive decline (n = 156), mild cognitive impairment or mild Alzheimer's disease dementia (n = 126) using structural equation modelling. CSF pathology markers were inversely correlated and showed a stronger association with disease severity, suggesting distinguishable interrelatedness of these biomarkers depending on the stage of Alzheimer's disease dementia. Across groups, amyloid pathology was linked to atrophy in areas high as well as low in neurotransmitter receptor densities, while tau pathology did not show any significant link to brain area volumes for any of the neurotransmitters. Within disease severity groups, individuals with more amyloid pathology showed more atrophy only in 'areas high in noradrenaline', whereas for dopamine tau pathology was linked to higher volumes in areas low in receptor density possibly indicating compensatory mechanisms. Furthermore, individuals with more tau pathology showed a selective decrease in memory function while amyloid pathology was related to a decline in executive function and language capacity as well as memory function. In summary, our analyses highlight the benefits of investigating disease-relevant factors in Alzheimer's disease using a multivariate multigroup approach. Assessing multivariate dependencies in different disease stages and across individuals revealed selective links of pathologies, cognitive decline and atrophy in particular for areas modulated by noradrenaline, dopamine and serotonin.
Collapse
Affiliation(s)
- Lena Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Elisa Lancini
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Renat Yakupov
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Yeo-Jin Yi
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Falk Lüsebrink
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, 12200 Berlin, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Humboldt University Berlin, 10117 Berlin, Germany
| | - Slawek Altenstein
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Humboldt University Berlin, 10117 Berlin, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Humboldt University Berlin, 10117 Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Luisa Sophie Schneider
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, 12200 Berlin, Germany
| | - Xiao Wang
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, 12200 Berlin, Germany
| | - Lukas Preis
- Department of Psychiatry and Psychotherapy, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt—Universität zu Berlin, 12200 Berlin, Germany
| | - Frederic Brosseron
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
| | - Nina Roy-Kluth
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
| | - Klaus Fliessbach
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Deptartment of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Deptartment of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Steffen Wolfsgruber
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Deptartment of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Deptartment of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
| | - Alfredo Ramirez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Deptartment of Neurodegenerative Disease and Geriatric Psychiatry/Psychiatry, University of Bonn Medical Center, 53127 Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
- Department of Psychiatry & Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53127 Bonn, Germany
- Department of Neurology, University of Bonn, 53127 Bonn, Germany
| | - Frank Jessen
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), 37075 Goettingen, Germany
- Department of Psychiatry, University of Cologne, Medical Faculty, 50924 Cologne, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), 37075 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, 37075 Goettingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, 3810-198 Aveiro, Portugal
| | - Anja Schneider
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, 37075 Goettingen, Germany
| | - Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen, University of Goettingen, 37075 Goettingen, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, University of Cologne, Medical Faculty, 50924 Cologne, Germany
| | - Katharina Buerger
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Michael Ewers
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London W12 0BZ, UK
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, 80336 Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
- Department of Neuroradiology, University Hospital LMU, 81377 Munich, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Doreen Goerss
- German Center for Neurodegenerative Diseases (DZNE), 18147 Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, 18147 Rostock, Germany
| | - Christoph Laske
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
| | - Matthias H Munk
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
| | - Michael Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Stefan Hetzer
- Berlin Center for Advanced Neuroimaging, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, 72076 Tübingen, Germany
| | - Emrah Düzel
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Matthew J Betts
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, 39120 Magdeburg, Germany
| | - Dorothea Hämmerer
- Institute of Cognitive Neurology and Dementia Research (IKND), 39120 Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences, University of Magdeburg, 39120 Magdeburg, Germany
- Department of Psychology, University of Innsbruck, 6020 Innsbruck, Austria
- Institute of Cognitive Neuroscience, University College London, London WC1E 6BT, UK
| |
Collapse
|
34
|
Oddi S, Scipioni L, Totaro A, Giacovazzo G, Ciaramellano F, Tortolani D, Leuti A, Businaro R, Armeli F, Bilkei-Gorzo A, Coccurello R, Zimmer A, Maccarrone M. Fatty-acid amide hydrolase inhibition mitigates Alzheimer's disease progression in mouse models of amyloidosis. FEBS J 2025. [PMID: 39822137 DOI: 10.1111/febs.17403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/17/2024] [Accepted: 11/22/2024] [Indexed: 01/19/2025]
Abstract
The endocannabinoid N-arachidonoylethanolamine (AEA) is a pro-homeostatic bioactive lipid known for its anti-inflammatory, anti-oxidative, immunomodulatory, and neuroprotective properties, which may contrast/mitigate Alzheimer's disease (AD) pathology. This study explores the therapeutic potential of targeting fatty acid amide hydrolase (FAAH), the major enzyme degrading AEA, in mouse models of amyloidosis (APP/PS1 and Tg2576). Enhancing AEA signaling by genetic deletion of FAAH delayed cognitive deficits in APP/PS1 mice and improved cognitive symptoms in 12-month-old AD-like mice. Chronic pharmacological FAAH inhibition fully reverted neurocognitive decline, attenuated neuroinflammation, and promoted neuroprotective mechanisms in Tg2576 mice. Additionally, pharmacological FAAH inhibition robustly suppressed β-amyloid production and accumulation, associated with decreased expression of β-site amyloid precursor protein cleaving enzyme 1 (BACE1), possibly through a cannabinoid receptor 1-dependent epigenetic mechanism. These findings improve our understanding of AEA signaling in AD pathogenesis and provide proof of concept that selective targeting of FAAH activity could be a promising therapeutic strategy against AD.
Collapse
Affiliation(s)
- Sergio Oddi
- Department of Veterinary Medicine, University of Teramo, Italy
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Lucia Scipioni
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Antonio Totaro
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Giacomo Giacovazzo
- Department of Veterinary Medicine, University of Teramo, Italy
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Francesca Ciaramellano
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Daniel Tortolani
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Alessandro Leuti
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
- Department of Medicine, Campus Bio-Medico University of Rome, Italy
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Federica Armeli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Andras Bilkei-Gorzo
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Germany
| | - Roberto Coccurello
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
- National Research Council (CNR), Institute for Complex System (ISC), Rome, Italy
| | - Andreas Zimmer
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, Germany
| | - Mauro Maccarrone
- European Center for Brain Research/Institute for Research and Health Care (IRCCS) Santa Lucia Foundation, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| |
Collapse
|
35
|
Anupriya ES, Chen R, Kalski D, Palmer J, Shen M. Dual-channel nano-carbon-liquid/liquid junction electrodes for multi-modal analysis: redox-active (dopamine) and non-redox-active (acetylcholine). Analyst 2025; 150:414-424. [PMID: 39688537 DOI: 10.1039/d4an01153h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
We present here a dual-channel nanoelectrode to detect both redox-active and non-redox-active analytes. The dual-channel nanoelectrode was developed from theta nanopipette. We developed one channel of the theta nanopipette to be a carbon nanoelectrode and the other channel to be a nano interface between two immiscible electrolyte solutions (nanoITIES) electrode, producing a nano-carbon-ITIES platform. The carbon nanoelectrode channel was developed by carbon deposition via pyrolysis followed by focused ion beam milling to measure redox-active analytes. The nanoITIES electrode channel was developed to detect non-redox-active analytes. The nano-carbon-ITIES electrodes were characterized using electrochemistry, scanning electron microscopy and transmission electron microscopy. Dopamine (a redox-active analyte) and acetylcholine (a non-redox-active analyte) were measured on the dual-channel nano-carbon-ITIES platform using the carbon nanoelectrode and the nanoITIES electrode, respectively. Using cyclic voltammetry, the diffusion-limited current of dopamine and acetylcholine detection on the nano-carbon-ITIES electrode increased linearly with increasing their concentrations. Using chronoamperometry (current versus time), we showed that the nano-carbon-ITIES electrode detected acetylcholine and dopamine at the same time. The introduced first-ever dual-functional nano-carbon-ITIES electrodes expand the current literature in multi-channel electrodes for multi-purpose analysis, which is an emerging area of research. Developing the analytical capability for the simultaneous detection of acetylcholine and dopamine is a critical step towards understanding diseases and disorders where both dopamine and acetylcholine are involved.
Collapse
Affiliation(s)
- Edappalil Satheesan Anupriya
- Department of Chemistry, The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois
| | - Ran Chen
- Department of Chemistry, The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| | - Daniel Kalski
- Department of Chemistry, The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| | - Jordynn Palmer
- Department of Chemistry, The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
| | - Mei Shen
- Department of Chemistry, The Beckman Institute for Advanced Science and Technology, University of Illinois Urbana Champaign, Urbana, IL 61801, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois
| |
Collapse
|
36
|
Manca R, De Marco M, Soininen H, Ruffini L, Venneri A. Changes in neurotransmitter-related functional connectivity along the Alzheimer's disease continuum. Brain Commun 2025; 7:fcaf008. [PMID: 39980737 PMCID: PMC11840171 DOI: 10.1093/braincomms/fcaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
Alzheimer's disease may be associated with early dopamine dysfunction. However, its effects on neurofunctional alterations in the neurotransmission pathways remain elusive. In this study, positron emission tomography atlases and functional MRI data for 86 older adults with mild cognitive impairment Alzheimer's disease (MCI), 58 with mild Alzheimer's disease-dementia and 76 cognitively unimpaired were combined to investigate connectivity alterations associated with the dopaminergic and cholinergic systems. A cross-sectional design was used to compare neurotransmitter-related functional connectivity across groups and associations between functional connectivity and cognitive performance. The findings show that the Alzheimer's disease dementia group showed a decline in mesocorticolimbic dopamine-related connectivity in the precuneus but heightened connectivity in the thalamus, whereas the Alzheimer's disease-MCI group showed a decline in nigrostriatal connectivity in the left temporal areas. Acetylcholine-related connectivity decline was observed in both Alzheimer's disease-MCI and Alzheimer's disease-dementia primarily in the temporo-parietal areas. Episodic memory scores correlated positively with acetylcholine- and dopamine-related connectivity in the temporo-parietal cortex and negatively with dopamine-related functional connectivity in the fronto-thalamic areas. This study shows that connectivity alterations in acetylcholine and dopamine functional pathways parallel cognitive decline in Alzheimer's disease and might be a clinically relevant marker in early Alzheimer's disease.
Collapse
Affiliation(s)
- Riccardo Manca
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Matteo De Marco
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
| | - Hilkka Soininen
- Institute of Clinical Medicine, Neurology, University of Eastern Finland, 70210 Kuopio, Finland
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria of Parma, 43126 Parma, Italy
| | - Annalena Venneri
- Department of Life Sciences, Brunel University of London, UB8 3PH London, UK
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| |
Collapse
|
37
|
Ali N, Beheshti A, Hampikian G. Space exploration and risk of Parkinson's disease: a perspective review. NPJ Microgravity 2025; 11:1. [PMID: 39753605 PMCID: PMC11698718 DOI: 10.1038/s41526-024-00457-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025] Open
Abstract
Systemic mitochondrial dysfunction, dopamine loss, sustained structural changes in the basal ganglia including reduced tyrosine hydroxylase, and altered gait- these effects observed in space-flown animals and astronauts mirrors Parkinson's disease (PD). Evidence of mitochondrial changes in space-flown human cells, examined through the lens of PD, suggests that spaceflight-induced PD-like molecular changes are important to monitor during deep space exploration. These changes, may potentially elevate the risk of PD in astronauts.
Collapse
Affiliation(s)
- Nilufar Ali
- Department of Biological Science, Boise State University, Boise, ID, 83725, USA.
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Greg Hampikian
- Department of Biological Science, Boise State University, Boise, ID, 83725, USA
| |
Collapse
|
38
|
Velázquez-Delgado C, Hernández-Ortiz E, Landa-Navarro L, Tapia-Rodríguez M, Moreno-Castilla P, Bermúdez-Rattoni F. Repeated exposure to novelty promotes resilience against the amyloid-beta effect through dopaminergic stimulation. Psychopharmacology (Berl) 2025; 242:85-100. [PMID: 39145803 PMCID: PMC11742894 DOI: 10.1007/s00213-024-06650-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
RATIONALE The accumulation of beta-amyloid peptide (Aβ) in the forebrain leads to cognitive dysfunction and neurodegeneration in Alzheimer's disease. Studies have shown that individuals with a consistently cognitively active lifestyle are less vulnerable to Aβ toxicity. Recent research has demonstrated that intrahippocampal Aβ can impact catecholaminergic release and spatial memory. Interestingly, exposure to novelty stimuli has been found to stimulate the release of catecholamines in the hippocampus. However, it remains uncertain whether repeated enhancing catecholamine activity can effectively alleviate cognitive impairment in individuals with Alzheimer's disease. OBJECTIVES Our primary aim was to investigate whether repeated exposure to novelty could enable cognitive resilience against Aβ. This protection could be achieved by modulating catecholaminergic activity within the hippocampus. METHODS To investigate this hypothesis, we subjected mice to three different conditions-standard housing (SH), repeated novelty (Nov), or daily social interaction (Soc) for one month. We then infused saline solution (SS) or Aβ (Aβ1-42) oligomers intrahippocampally and measured spatial memory retrieval in a Morris Water Maze (MWM). Stereological analysis and extracellular baseline dopamine levels using in vivo microdialysis were assessed in independent groups of mice. RESULTS The mice that received Aβ1-42 intrahippocampal infusions and remained in SH or Soc conditions showed impaired spatial memory retrieval. In contrast, animals subjected to the Nov protocol demonstrated remarkable resilience, showing strong spatial memory expression even after Aβ1-42 intrahippocampal infusion. The stereological analysis indicated that the Aβ1-42 infusion reduced the tyrosine hydroxylase axonal length in SH or Soc mice compared to the Nov group. Accordingly, the hippocampal extracellular dopamine levels increased significantly in the Nov groups. CONCLUSIONS These compelling results demonstrate the potential for repeated novelty exposure to strengthen the dopaminergic system and mitigate the toxic effects of Aβ1-42. They also highlight new and promising therapeutic avenues for treating and preventing AD, especially in its early stages.
Collapse
Affiliation(s)
- Cintia Velázquez-Delgado
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Eduardo Hernández-Ortiz
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Lucia Landa-Navarro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Miguel Tapia-Rodríguez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Perla Moreno-Castilla
- Laboratory of Cognitive Resilience, Center of Aging Research (CIE), Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV, Mexico City, Mexico.
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
39
|
Stanzione A, Melchiori FM, Costa A, Leonardi C, Scalici F, Caltagirone C, Carlesimo GA. Dopaminergic Treatment and Episodic Memory in Parkinson's Disease: A Meta-analysis of the Literature. Neuropsychol Rev 2024:10.1007/s11065-024-09656-0. [PMID: 39708069 DOI: 10.1007/s11065-024-09656-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
To date, few studies have focused on the benefits of dopaminergic treatment on episodic memory functions in patients affected by Parkinson's disease (PD). We conducted a meta-analysis to determine the effects of pharmacological therapy with dopamine in alleviating episodic memory deficits in Parkinson's patients. A secondary aim was to evaluate the role of dopamine in episodic memory circuits and thus in different memory systems. We conducted a comprehensive literature search in PubMed (1971-2022) to find studies that met specific inclusion criteria. The studies had to provide sufficient data (means and standard deviations) to evaluate performance on neuropsychological tests of episodic memory. A total of k = 36 measures were included in the analysis. A statistically significant difference suggested better performance following dopaminergic therapy assumption (ON condition) than following dopaminergic withdrawal (OFF condition), specifically the estimated pooled effect calculated through a random-effects restricted maximum likelihood model was log ratio of means (RoM) = 0.047 (p = 0.011). The back-transformed RoM, indicating a 4.8% improvement, provides an interpretable measure of the effect size, as it reflects the multiplicative change in performance associated with the ON condition. A meta-regression analysis was also performed to assess the influence of specific memory tasks and relevant covariates/factors on the overall meta-analytic effect: four memory contrasts (verbal/visual, immediate/delayed, recall/recognition, word-list/short-story), age of participants, years of education, severity of illness, duration of illness in years, country of study, proportion of women in the sample, type of medication, counterbalancing. Word list/short story and proportion of women in the sample were the only two statistically significant predictors in the model, both associated with a positive higher pooled effect size. The present study revealed a significant overall difference between the results obtained in the ON and OFF conditions. We also found a significantly greater pharmacological effect in the recall of short stories than word lists, which supports the hypothesis of a beneficial effect of dopamine on the hippocampal circuit rather than on prefrontal cortical areas.
Collapse
Affiliation(s)
- Agostino Stanzione
- Laboratory of Neuropsychology of Memory, Department of Clinical Neuroscience and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179, Rome, Italy.
| | | | - Alberto Costa
- Laboratory of Neuropsychology of Memory, Department of Clinical Neuroscience and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179, Rome, Italy
- Niccolò Cusano University, Rome, Italy
| | - Carla Leonardi
- Laboratory of Neuropsychology of Memory, Department of Clinical Neuroscience and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179, Rome, Italy
- Niccolò Cusano University, Rome, Italy
| | - Francesco Scalici
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
- Centre for Space BioMedicine, University of Rome Tor Vergata, Rome, Italy
- Laboratory of Neuromotor Physiology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Caltagirone
- Laboratory of Neuropsychology of Memory, Department of Clinical Neuroscience and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179, Rome, Italy
| | - Giovanni Augusto Carlesimo
- Laboratory of Neuropsychology of Memory, Department of Clinical Neuroscience and Neurorehabilitation, IRCCS Santa Lucia Foundation, Via Ardeatina 306, 00179, Rome, Italy
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| |
Collapse
|
40
|
Yang H, Tan H, Wen H, Xin P, Liu Y, Deng Z, Xu Y, Gao F, Zhang L, Ye Z, Zhang Z, Chen Y, Wang Y, Sun J, Lam JWY, Zhao Z, Kwok RTK, Qiu Z, Tang BZ. Recent Progress in Nanomedicine for the Diagnosis and Treatment of Alzheimer's Diseases. ACS NANO 2024; 18:33792-33826. [PMID: 39625718 DOI: 10.1021/acsnano.4c11966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that causes memory loss and progressive and permanent deterioration of cognitive function. The most challenging issue in combating AD is its complicated pathogenesis, which includes the deposition of amyloid β (Aβ) plaques, intracellular hyperphosphorylated tau protein, neurofibrillary tangles (NFT), etc. Despite rapid advancements in mechanistic research and drug development for AD, the currently developed drugs only improve cognitive ability and temporarily relieve symptoms but cannot prevent the development of AD. Moreover, the blood-brain barrier (BBB) creates a huge barrier to drug delivery in the brain. Therefore, effective diagnostic tools and treatments are urgently needed. In recent years, nanomedicine has provided opportunities to overcome the challenges and limitations associated with traditional diagnostics or treatments. Various types of nanoparticles (NPs) play an essential role in nanomedicine for the diagnosis and treatment of AD, acting as drug carriers to improve targeting and bioavailability across/bypass the BBB or acting as drugs directly on AD lesions. This review categorizes different types of NPs and summarizes their applications in nanomedicine for the diagnosis and treatment of AD. It also discusses the challenges associated with clinical applications and explores the latest developments and prospects of nanomedicine for AD.
Collapse
Affiliation(s)
- Han Yang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong China
| | - Haozhe Tan
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Haifei Wen
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Peikun Xin
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Yanling Liu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Ziwei Deng
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Yanning Xu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Feng Gao
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Liping Zhang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Ziyue Ye
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Zicong Zhang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Yunhao Chen
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Yueze Wang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Jianwei Sun
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong China
| | - Jacky W Y Lam
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong China
| | - Zheng Zhao
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Ryan T K Kwok
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong China
| | - Zijie Qiu
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, P.R. China
- Department of Chemistry, the Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Division of Life Science, State Key Laboratory of Molecular Neuroscience, and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong China
| |
Collapse
|
41
|
Paoletti I, Coccurello R. Irisin: A Multifaceted Hormone Bridging Exercise and Disease Pathophysiology. Int J Mol Sci 2024; 25:13480. [PMID: 39769243 PMCID: PMC11676223 DOI: 10.3390/ijms252413480] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
The fibronectin domain-containing protein 5 (FNDC5), or irisin, is an adipo-myokine hormone produced during exercise, which shows therapeutic potential for conditions like metabolic disorders, osteoporosis, sarcopenia, obesity, type 2 diabetes, and neurodegenerative diseases, including Alzheimer's disease (AD). This review explores its potential across various pathophysiological processes that are often considered independent. Elevated in healthy states but reduced in diseases, irisin improves muscle-adipose communication, insulin sensitivity, and metabolic balance by enhancing mitochondrial function and reducing oxidative stress. It promotes osteogenesis and mitigates bone loss in osteoporosis and sarcopenia. Irisin exhibits anti-inflammatory effects by inhibiting NF-κB signaling and countering insulin resistance. In the brain, it reduces amyloid-β toxicity, inflammation, and oxidative stress, enhancing brain-derived neurotrophic factor (BDNF) signaling, which improves cognition and synaptic health in AD models. It also regulates dopamine pathways, potentially alleviating neuropsychiatric symptoms like depression and apathy. By linking physical activity to systemic health, irisin emphasizes its role in the muscle-bone-brain axis. Its multifaceted benefits highlight its potential as a therapeutic target for AD and related disorders, with applications in prevention, in treatment, and as a complement to exercise strategies.
Collapse
Affiliation(s)
- Ilaria Paoletti
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Research Council (C.N.R.), 00185 Rome, Italy
| |
Collapse
|
42
|
Hiraga T, Hata T, Soya S, Shimoda R, Takahashi K, Soya M, Inoue K, Johansen JP, Okamoto M, Soya H. Light-exercise-induced dopaminergic and noradrenergic stimulation in the dorsal hippocampus: Using a rat physiological exercise model. FASEB J 2024; 38:e70215. [PMID: 39668509 PMCID: PMC11638517 DOI: 10.1096/fj.202400418rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024]
Abstract
Exercise activates the dorsal hippocampus that triggers synaptic and cellar plasticity and ultimately promotes memory formation. For decades, these benefits have been explored using demanding and stress-response-inducing exercise at moderate-to-vigorous intensities. In contrast, our translational research with animals and humans has focused on light-intensity exercise (light exercise) below the lactate threshold (LT), which almost anyone can safely perform with minimal stress. We found that even light exercise can stimulate hippocampal activity and enhance memory performance. Although the circuit mechanism of this boost remains unclear, arousal promotion even with light exercise implies the involvement of the ascending monoaminergic system that is essential to modulate hippocampal activity and impact memory. To test this hypothesis, we employed our physiological exercise model based on the LT of rats and immunohistochemically assessed the neuronal activation of the dorsal hippocampal sub-regions and brainstem monoaminergic neurons. Also, we monitored the extracellular concentration of monoamines in the dorsal hippocampus using in vivo microdialysis. We found that even light exercise increased neuronal activity in the dorsal hippocampal sub-regions and elevated the extracellular concentrations of noradrenaline and dopamine. Furthermore, we found that tyrosine hydroxylase-positive neurons in the locus coeruleus (LC) and the ventral tegmental area (VTA) were activated even by light exercise and were both positively correlated with the dorsal hippocampal activation. In conclusion, our findings demonstrate that light exercise stimulates dorsal hippocampal neurons, which are associated with LC-noradrenergic and VTA-dopaminergic activation. This shed light on the circuit mechanisms responsible for hippocampal neural activation during exercise, consequently enhancing memory function.
Collapse
Affiliation(s)
- Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Toshiaki Hata
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Shingo Soya
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaJapan
- Department of Molecular Behavioral Physiology, Institute of MedicineUniversity of TsukubaTsukubaJapan
| | - Ryo Shimoda
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Kanako Takahashi
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Mariko Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Department of Anatomy and Neuroscience, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Koshiro Inoue
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Center for Education in Liberal Arts and SciencesHealth Sciences University of HokkaidoIshikariJapan
| | - Joshua P. Johansen
- Laboratory for Neural Circuitry of MemoryRIKEN Center for Brain ScienceSaitamaJapan
| | - Masahiro Okamoto
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
- Division of Sport Neuroscience, Kokoro Division, Advanced Research Initiative for Human High Performance (ARIHHP), Institute of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| |
Collapse
|
43
|
Albuz Ö, Acir I, Haşimoğlu O, Suskun M, Hocaoğlu E, Yayla V. Cranial volume measurement with artificial intelligence and cognitive scales in patients with clinically isolated syndrome. Front Neurol 2024; 15:1500140. [PMID: 39722699 PMCID: PMC11668644 DOI: 10.3389/fneur.2024.1500140] [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: 09/22/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Objective We aimed to investigate the relationship between volumetric measurements of specific brain regions which were measured with artificial intelligence (AI) and various neuropsychological tests in patients with clinically isolated syndrome. Materials and methods A total of 28 patients diagnosed with CIS were included in the study. The patients were administered Öktem Verbal Memory Processes Test, Symbol Digit Modalities Test (SDMT), Backward-Forward Digit Span Test, Stroop Test, Trail Making Test, Controlled Oral Word Association Test (COWAT), Brief Visuospatial Memory Test, Judgement of Line Orientation Test, Beck Depression Scale, Beck Anxiety Scale and Fatigue Severity Scale. Artificial intelligence assisted BrainLab Elements™ Atlas-Based Automatic Segmentation program was used for calculating volumes. The measured volumes were compared with the reference database. In addition, neuropsychological test performances and volumetric measurements of the patients were compared. Results Of the patients included in the study, 78.6% were female and 21.4% were male, with an average age of 33 years. Verbal Memory Processes Test, SDMT, Backward-Forward Digit Span, JLOT, and Stroop Test showed significant correlations with multiple anatomical regions, particularly the anterior thalamic nucleus, which was associated with the highest number of cognitive tests. The JLOT exhibited the strongest correlation with six different brain regions (p < 0.001). Conclusion The Judgement of Line Orientation and Stroop Tests, correlated with multiple brain regions, especially the anterior thalamic nucleus, underscoring the importance of these tests in assessing cognitive function in CIS.
Collapse
Affiliation(s)
- Özlem Albuz
- Bakırköy Dr. Sadi Konuk Eğitim ve Araştırma Hastanesi, Istanbul, Türkiye
| | - Ibrahim Acir
- Bakırköy Dr. Sadi Konuk Eğitim ve Araştırma Hastanesi, Istanbul, Türkiye
| | - Ozan Haşimoğlu
- Basaksehir Cam and Sakura City Hospital, Istanbul, Türkiye
| | - Melis Suskun
- Bakırköy Dr. Sadi Konuk Eğitim ve Araştırma Hastanesi, Istanbul, Türkiye
| | - Elif Hocaoğlu
- Bakırköy Dr. Sadi Konuk Eğitim ve Araştırma Hastanesi, Istanbul, Türkiye
| | - Vildan Yayla
- Bakırköy Dr. Sadi Konuk Eğitim ve Araştırma Hastanesi, Istanbul, Türkiye
| |
Collapse
|
44
|
Zhong Z, Dong H, Zhou S, Lin C, Huang P, Li X, Zhang J, Xie J, Wu Y, Li P. Caffeine's Neuroprotective Effect on Memory Impairment: Suppression of Adenosine A 2A Receptor and Enhancement of Tyrosine Hydroxylase in Dopaminergic Neurons Under Hypobaric Hypoxia Conditions. CNS Neurosci Ther 2024; 30:e70134. [PMID: 39670604 PMCID: PMC11638883 DOI: 10.1111/cns.70134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
AIMS Chronic hypobaric hypoxia frequently results in memory deficits, with severe cases showing marked alterations in dopamine levels and its metabolites. This research explores caffeine's modulation of the adenosine A2A receptor (A2AR) and its regulatory effects on tyrosine hydroxylase (TH), aiming to restore dopamine homeostasis and mitigate memory impairments associated with hypoxia. The goal is to identify novel preventive strategies against cognitive decline induced by hypoxia. METHODS Network pharmacological analysis was employed to predict the interactions between caffeine, cognitive function, and hypobaric hypoxia-related disorders. The novel object recognition and Y-maze tests were utilized to assess caffeine's impact on memory deficits under hypobaric hypoxia conditions in male mice. LC-MS/MS analysis was subsequently conducted to examine the variations in dopamine and its metabolites within the midbrain. Molecular docking further confirmed the binding affinities between A2AR and caffeine, as well as TH and caffeine. Additionally, immunofluorescence and protein-protein docking were employed to elucidate the interaction between A2AR and TH. RESULTS The findings highlight the pivotal role of adenosine receptors and dopamine-related pathways in the interplay between caffeine, cognition, and hypobaric hypoxia-related disorders. Behavioral tests demonstrated that caffeine effectively alleviated memory impairments caused by chronic hypobaric hypoxia. LC-MS/MS results revealed significant differences in dopamine, metanephrine, and 3-hydroxyanthranilic acid levels following caffeine treatment for hypoxia-induced cognitive deficits. Molecular docking confirmed the high affinity between A2AR and caffeine, as well as TH and caffeine, while immunofluorescence and protein-protein docking provided insights into the A2AR-TH interaction and its modulation during hypobaric hypoxia. CONCLUSIONS Caffeine exhibits potent neuroprotective effects against chronic high-altitude-induced cognitive impairments, potentially through its action on A2AR, leading to enhanced TH expression and subsequent release of dopamine and its related neurotransmitters.
Collapse
Affiliation(s)
- Zhifeng Zhong
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Huaping Dong
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Simin Zhou
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Chaoqun Lin
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Pei Huang
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Xiaoxu Li
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Jijian Zhang
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Jiaxin Xie
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Yu Wu
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| | - Peng Li
- Department of High Altitude Operational Medicine, College of High Altitude Military MedicineArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of ChinaArmy Medical UniversityChongqingPeople's Republic of China
- Key Laboratory of High Altitude MedicineArmy Medical UniversityChongqingPeople's Republic of China
| |
Collapse
|
45
|
Wang Z, Arnold JC. Cannabinoids and healthy ageing: the potential for extending healthspan and lifespan in preclinical models with an emphasis on Caenorhabditis elegans. GeroScience 2024; 46:5643-5661. [PMID: 38696056 PMCID: PMC11493940 DOI: 10.1007/s11357-024-01162-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 10/23/2024] Open
Abstract
There is a significant global upsurge in the number and proportion of older persons in the population. With this comes an increasing prevalence of age-related conditions which pose a major challenge to healthcare systems. The development of anti-ageing treatments may help meet this challenge by targeting the ageing process which is a common denominator to many health problems. Cannabis-like compounds (cannabinoids) are reported to improve quality of life and general well-being in human trials, and there is increasing preclinical research highlighting that they have anti-ageing activity. Moreover, preclinical evidence suggests that endogenous cannabinoids regulate ageing processes. Here, we review the anti-ageing effects of the cannabinoids in various model systems, including the most extensively studied nematode model, Caenorhabditis elegans. These studies highlight that the cannabinoids lengthen healthspan and lifespan, with emerging evidence that they may also hinder the development of cellular senescence. The non-psychoactive cannabinoid cannabidiol (CBD) shows particular promise, with mechanistic studies demonstrating it may work through autophagy induction and activation of antioxidative systems. Furthermore, CBD improves healthspan parameters such as diminishing age-related behavioural dysfunction in models of both healthy and accelerated ageing. Translation into mammalian systems provides an important next step. Moreover, looking beyond CBD, future studies could probe the multitude of other cannabis constituents for their anti-ageing activity.
Collapse
Affiliation(s)
- Zhizhen Wang
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Jonathon C Arnold
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.
- Discipline of Pharmacology, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
46
|
Kumar S, Mitra R, Ayyannan SR. Design, synthesis and evaluation of benzothiazole-derived phenyl thioacetamides as dual inhibitors of monoamine oxidases and cholinesterases. Mol Divers 2024:10.1007/s11030-024-11031-3. [PMID: 39520616 DOI: 10.1007/s11030-024-11031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
A series of rationally designed benzothiazole-derived thioacetamides was synthesized and investigated for monoamine oxidases (MAO-A and MAO-B) and cholinesterases (AChE and BChE) inhibition properties. The tested compounds 18-31 inhibited MAO-A and MAO-B in the micromolar to nanomolar range and AChE in the submicromolar range. Compound 28 was identified as the most potent MAO-A inhibitor with an IC50 = 0.030 ± 0.008 µM, whereas compound 30 showed the highest potency towards MAO-B and AChE with IC50 values of 0.015 ± 0.007 µM and 0.114 ± 0.003 µM, respectively. Further, compound 30 inhibited BChE at an IC50 value of 4.125 ± 0.143 µM. Among all screened molecules, compound 30 emerged as the lead dual MAO-B and AChE inhibitor that blocked these enzymes in a competitive-reversible and mixed-reversible mode, respectively. Selected compounds have displayed iron-chelation and antioxidant properties. Further, computational assessment of ligand binding affinity and pharmacokinetic parameters of all new compounds and molecular dynamic simulation of compound 30 with MAO-B and AChE were carried out to understand ligand efficiency, pharmacokinetic, and virtual molecular interaction profile, respectively. The in silico ADMET prediction studies revealed a few undesired pharmacokinetic attributes of our compounds. The attempted virtual lead-based library synthesis and subsequent biological investigation produced a new benzothiazole-bearing dual MAO-B and AChE inhibitor as a prospective MTDL candidate for treating neurological disorders.
Collapse
Affiliation(s)
- Sandeep Kumar
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Rangan Mitra
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Senthil Raja Ayyannan
- Pharmaceutical Chemistry Research Laboratory II, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India.
| |
Collapse
|
47
|
Blankenship HE, Carter KA, Pham KD, Cassidy NT, Markiewicz AN, Thellmann MI, Sharpe AL, Freeman WM, Beckstead MJ. VTA dopamine neurons are hyperexcitable in 3xTg-AD mice due to casein kinase 2-dependent SK channel dysfunction. Nat Commun 2024; 15:9673. [PMID: 39516200 PMCID: PMC11549218 DOI: 10.1038/s41467-024-53891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) patients exhibit neuropsychiatric symptoms that extend beyond classical cognitive deficits, suggesting involvement of subcortical areas. Here, we investigated the role of midbrain dopamine (DA) neurons in AD using the amyloid + tau-driven 3xTg-AD mouse model. We found deficits in reward-based operant learning in AD mice, suggesting possible VTA DA neuron dysregulation. Physiological assessment revealed hyperexcitability and disrupted firing in DA neurons caused by reduced activity of small-conductance calcium-activated potassium (SK) channels. RNA sequencing from contents of single patch-clamped DA neurons (Patch-seq) identified up-regulation of the SK channel modulator casein kinase 2 (CK2), which we corroborated by immunohistochemical protein analysis. Pharmacological inhibition of CK2 restored SK channel activity and normal firing patterns in 3xTg-AD mice. These findings identify a mechanism of ion channel dysregulation in VTA DA neurons that could contribute to behavioral abnormalities in AD, paving the way for novel treatment strategies.
Collapse
Affiliation(s)
- Harris E Blankenship
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelsey A Carter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kevin D Pham
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Nina T Cassidy
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Andrea N Markiewicz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael I Thellmann
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
48
|
Barzegar Behrooz A, Aghanoori MR, Nazari M, Latifi-Navid H, Vosoughian F, Anjomani M, Lotfi J, Ahmadiani A, Eliassi A, Nabavizadeh F, Soleimani E, Ghavami S, Khodagholi F, Fahanik-Babaei J. 40 Hz light preserves synaptic plasticity and mitochondrial function in Alzheimer's disease model. Sci Rep 2024; 14:26949. [PMID: 39506052 PMCID: PMC11541745 DOI: 10.1038/s41598-024-78528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Its causes are not fully understood, but it is now known that factors like mitochondrial dysfunction, oxidative stress, and compromised ion channels contribute to its onset and progression. Flickering light therapy has shown promise in AD treatment, though its mechanisms remain unclear. In this study, we used a rat model of streptozotocin (STZ)-induced AD to evaluate the effects of 40 Hz flickering light therapy. Rats received intracerebroventricular (ICV) STZ injections, and 7 days after, they were exposed to 40 Hz flickering light for 15 min daily over seven days. Cognitive and memory functions were assessed using Morris water maze, novel object recognition, and passive avoidance tests. STZ-induced AD rats exhibited cognitive decline, elevated reactive oxygen species, amyloid beta accumulation, decreased serotonin and dopamine levels, and impaired mitochondrial function. However, light therapy prevented these effects, preserving cognitive function and synaptic plasticity. Additionally, flickering light restored mitochondrial metabolites and normalized ATP-insensitive mitochondrial calcium-sensitive potassium (mitoBKCa) channel activity, which was otherwise downregulated in AD rats. Our findings suggest that 40 Hz flickering light therapy could be a promising treatment for neurodegenerative disorders like AD by preserving synaptic and mitochondrial function.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Mohamad-Reza Aghanoori
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary & Alberta Children's Hospital Research Institute, Calgary, AB, T2N 4N1, Canada
| | - Maryam Nazari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hamid Latifi-Navid
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Fatemeh Vosoughian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojdeh Anjomani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jabar Lotfi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Eliassi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Soleimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, 41-800, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, Canada
- Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Fahanik-Babaei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Crawford JL, Berry AS. Examining resilience to Alzheimer's disease through the lens of monoaminergic neuromodulator systems. Trends Neurosci 2024; 47:892-903. [PMID: 39368845 PMCID: PMC11563896 DOI: 10.1016/j.tins.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 10/07/2024]
Abstract
The monoaminergic nuclei are thought to be some of the earliest sites of Alzheimer's disease (AD) pathology in the brain, with tau-containing pretangles appearing in these nuclei decades before the onset of clinical impairments. It has increasingly been recognized that monoamine systems represent a critical target of investigation towards understanding the progression of AD and designing early detection and treatment approaches. This review synthesizes evidence across animal studies, human neuropathology, and state-of-the-art neuroimaging and daily life assessment methods in humans, which demonstrate robust relationships between monoamine systems and AD pathophysiology and behavior. Further, the review highlights the promise of multimethod, multisystem approaches to studying monoaminergic mechanisms of resilience to AD pathology.
Collapse
Affiliation(s)
| | - Anne S Berry
- Department of Psychology, Brandeis University, Waltham, MA, USA.
| |
Collapse
|
50
|
Krisanova N, Pozdnyakova N, Pastukhov A, Dudarenko M, Tarasenko A, Borysov A, Driuk M, Tolochko A, Bezkrovnyi O, Paliienko K, Sivko R, Gnatyuk O, Dovbeshko G, Borisova T. Synergistic neurological threat from Сu and wood smoke particulate matter. Food Chem Toxicol 2024; 193:115009. [PMID: 39304082 DOI: 10.1016/j.fct.2024.115009] [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: 08/08/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Trace metal Cu and carbonaceous airborn particulate matter (PM) are dangerous neuropollutants. Here, the ability of Cu2+ to modulate the neurotoxicity caused by water-suspended wood smoke PM preparations (SPs) and vice versa was examined using presynaptic rat cortex nerve terminals. Interaction of Cu2+ and SPs, changes of particle size and surface properties were shown in the presence of Cu2+ using microscopy, DLS, and IR spectroscopy. In nerve terminals, Cu2+ and SPs per se elevated the ambient levels of excitatory and inhibitory neurotransmitters L-[14C]glutamate and [3H]GABA, respectively. During combined application, Cu2+ significantly enhanced a SPs-induced increase in the ambient levels of both neurotransmitters, thereby demonstrating a cumulative synergistic effect and significant interference in the neurotoxic threat associated with Cu2+and SPs. In fluorimetric measurements, Cu2+ and SPs also demonstrated cumulative synergistic effects on the membrane potential, mitochondrial potential, synaptic vesicle acidification and ROS generation. Therefore, synergistic effects of Cu2+ and SPs on the most crucial presynaptic characteristics and neurohazard of multiple pollutants through excitatory/inhibitory imbalance, disruption of the membrane and mitochondrial potential, vesicle acidification and ROS generation were revealed. Increased expansion and burden of neuropathology may result from underestimation of synergistic interference of the neurotoxic effects of Cu2+ and carbonaceous smoke PM.
Collapse
Affiliation(s)
- Nataliya Krisanova
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine; Optical Spectroscopy Division, Institute of Low Temperature and Structure Research Polish Academy of Sciences, Okólna 2, Wrocław, 50-422, Poland
| | - Natalia Pozdnyakova
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Artem Pastukhov
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Marina Dudarenko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Alla Tarasenko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Arsenii Borysov
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Mykola Driuk
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Anatoliy Tolochko
- Department of Physics of Biological Systems, Institute of Physics, National Academy of Sciences of Ukraine, 46 Nauky Ave, Kyiv, 03680, Ukraine
| | - Oleksii Bezkrovnyi
- Optical Spectroscopy Division, Institute of Low Temperature and Structure Research Polish Academy of Sciences, Okólna 2, Wrocław, 50-422, Poland
| | - Konstantin Paliienko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Roman Sivko
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine
| | - Olena Gnatyuk
- Department of Physics of Biological Systems, Institute of Physics, National Academy of Sciences of Ukraine, 46 Nauky Ave, Kyiv, 03680, Ukraine; Optical Spectroscopy Division, Institute of Low Temperature and Structure Research Polish Academy of Sciences, Okólna 2, Wrocław, 50-422, Poland
| | - Galyna Dovbeshko
- Department of Physics of Biological Systems, Institute of Physics, National Academy of Sciences of Ukraine, 46 Nauky Ave, Kyiv, 03680, Ukraine; Optical Spectroscopy Division, Institute of Low Temperature and Structure Research Polish Academy of Sciences, Okólna 2, Wrocław, 50-422, Poland
| | - Tatiana Borisova
- Department of Neurochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, 9 Leontovicha str, Kyiv, 01054, Ukraine; Optical Spectroscopy Division, Institute of Low Temperature and Structure Research Polish Academy of Sciences, Okólna 2, Wrocław, 50-422, Poland.
| |
Collapse
|