1
|
Saggu S, Bai A, Aida M, Rehman H, Pless A, Ware D, Deak F, Jiao K, Wang Q. Monoamine alterations in Alzheimer's disease and their implications in comorbid neuropsychiatric symptoms. GeroScience 2024:10.1007/s11357-024-01359-x. [PMID: 39331291 DOI: 10.1007/s11357-024-01359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by relentless cognitive decline and the emergence of profoundly disruptive neuropsychiatric symptoms. As the disease progresses, it unveils a formidable array of neuropsychiatric manifestations, including debilitating depression, anxiety, agitation, and distressing episodes of psychosis. The intricate web of the monoaminergic system, governed by serotonin, dopamine, and norepinephrine, significantly influences our mood, cognition, and behavior. Emerging evidence suggests that dysregulation and degeneration of this system occur early in AD, leading to notable alterations in these critical neurotransmitters' levels, metabolism, and receptor function. However, how the degeneration of monoaminergic neurons and subsequent compensatory changes contribute to the presentation of neuropsychiatric symptoms observed in Alzheimer's disease remains elusive. This review synthesizes current findings on monoamine alterations in AD and explores how these changes contribute to the neuropsychiatric symptomatology of the disease. By elucidating the biological underpinnings of AD-related psychiatric symptoms, we aim to underscore the complexity and inform innovative approaches for treating neuropsychiatric symptoms in AD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Ava Bai
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Mae Aida
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
2
|
Pasquini L, Jenabi M, Graham M, Peck KK, Schöder H, Holodny AI, Krebs S. Tumors Affect the Metabolic Connectivity of the Human Brain Measured by 18 F-FDG PET. Clin Nucl Med 2024; 49:822-829. [PMID: 38693648 PMCID: PMC11300165 DOI: 10.1097/rlu.0000000000005227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
PURPOSE 18 F-FDG PET captures the relationship between glucose metabolism and synaptic activity, allowing for modeling brain function through metabolic connectivity. We investigated tumor-induced modifications of brain metabolic connectivity. PATIENTS AND METHODS Forty-three patients with left hemispheric tumors and 18 F-FDG PET/MRI were retrospectively recruited. We included 37 healthy controls (HCs) from the database CERMEP-IDB-MRXFDG. We analyzed the whole brain and right versus left hemispheres connectivity in patients and HC, frontal versus temporal tumors, active tumors versus radiation necrosis, and patients with high Karnofsky performance score (KPS = 100) versus low KPS (KPS < 70). Results were compared with 2-sided t test ( P < 0.05). RESULTS Twenty high-grade glioma, 4 low-grade glioma, and 19 metastases were included. The patients' whole-brain network displayed lower connectivity metrics compared with HC ( P < 0.001), except assortativity and betweenness centrality ( P = 0.001). The patients' left hemispheres showed decreased similarity, and lower connectivity metrics compared with the right ( P < 0.01), with the exception of betweenness centrality ( P = 0.002). HC did not show significant hemispheric differences. Frontal tumors showed higher connectivity metrics ( P < 0.001) than temporal tumors, but lower betweenness centrality ( P = 4.5 -7 ). Patients with high KPS showed higher distance local efficiency ( P = 0.01), rich club coefficient ( P = 0.0048), clustering coefficient ( P = 0.00032), betweenness centrality ( P = 0.008), and similarity ( P = 0.0027) compared with low KPS. Patients with active tumor(s) (14/43) demonstrated significantly lower connectivity metrics compared with necroses. CONCLUSIONS Tumors cause reorganization of metabolic brain networks, characterized by formation of new connections and decreased centrality. Patients with frontal tumors retained a more efficient, centralized, and segregated network than patients with temporal tumors. Stronger metabolic connectivity was associated with higher KPS.
Collapse
Affiliation(s)
- Luca Pasquini
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mehrnaz Jenabi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maya Graham
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- The Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kyung K. Peck
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Medical College of Cornell University, New York, NY
| | - Andrei I. Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Medical College of Cornell University, New York, NY
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Radiology, Weill Medical College of Cornell University, New York, NY
| |
Collapse
|
3
|
Hanania JU, Reimers E, Bevington CWJ, Sossi V. PET-based brain molecular connectivity in neurodegenerative disease. Curr Opin Neurol 2024; 37:353-360. [PMID: 38813843 DOI: 10.1097/wco.0000000000001283] [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: 05/31/2024]
Abstract
PURPOSE OF REVIEW Molecular imaging has traditionally been used and interpreted primarily in the context of localized and relatively static neurochemical processes. New understanding of brain function and development of novel molecular imaging protocols and analysis methods highlights the relevance of molecular networks that co-exist and interact with functional and structural networks. Although the concept and evidence of disease-specific metabolic brain patterns has existed for some time, only recently has such an approach been applied in the neurotransmitter domain and in the context of multitracer and multimodal studies. This review briefly summarizes initial findings and highlights emerging applications enabled by this new approach. RECENT FINDINGS Connectivity based approaches applied to molecular and multimodal imaging have uncovered molecular networks with neurodegeneration-related alterations to metabolism and neurotransmission that uniquely relate to clinical findings; better disease stratification paradigms; an improved understanding of the relationships between neurochemical and functional networks and their related alterations, although the directionality of these relationships are still unresolved; and a new understanding of the molecular underpinning of disease-related alteration in resting-state brain activity. SUMMARY Connectivity approaches are poised to greatly enhance the information that can be extracted from molecular imaging. While currently mostly contributing to enhancing understanding of brain function, they are highly likely to contribute to the identification of specific biomarkers that will improve disease management and clinical care.
Collapse
Affiliation(s)
| | - Erik Reimers
- Department of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
4
|
Wang Z, Yang X, Li H, Wang S, Liu Z, Wang Y, Zhang X, Chen Y, Xu Q, Xu J, Wang Z, Wang J. Bidirectional two-sample Mendelian randomization analyses support causal relationships between structural and diffusion imaging-derived phenotypes and the risk of major neurodegenerative diseases. Transl Psychiatry 2024; 14:215. [PMID: 38806463 PMCID: PMC11133432 DOI: 10.1038/s41398-024-02939-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
Previous observational investigations suggest that structural and diffusion imaging-derived phenotypes (IDPs) are associated with major neurodegenerative diseases; however, whether these associations are causal remains largely uncertain. Herein we conducted bidirectional two-sample Mendelian randomization analyses to infer the causal relationships between structural and diffusion IDPs and major neurodegenerative diseases using common genetic variants-single nucleotide polymorphism (SNPs) as instrumental variables. Summary statistics of genome-wide association study (GWAS) for structural and diffusion IDPs were obtained from 33,224 individuals in the UK Biobank cohort. Summary statistics of GWAS for seven major neurodegenerative diseases were obtained from the largest GWAS for each disease to date. The forward MR analyses identified significant or suggestively statistical causal effects of genetically predicted three structural IDPs on Alzheimer's disease (AD), frontotemporal dementia (FTD), and multiple sclerosis. For example, the reduction in the surface area of the left superior temporal gyrus was associated with a higher risk of AD. The reverse MR analyses identified significantly or suggestively statistical causal effects of genetically predicted AD, Lewy body dementia (LBD), and FTD on nine structural and diffusion IDPs. For example, LBD was associated with increased mean diffusivity in the right superior longitudinal fasciculus and AD was associated with decreased gray matter volume in the right ventral striatum. Our findings might contribute to shedding light on the prediction and therapeutic intervention for the major neurodegenerative diseases at the neuroimaging level.
Collapse
Affiliation(s)
- Zirui Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xuan Yang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Department of Radiology, Jining No.1 People's Hospital, Jining, Shandong, 272000, China
| | - Haonan Li
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Siqi Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Zhixuan Liu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yaoyi Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xingyu Zhang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yayuan Chen
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Junping Wang
- Department of Radiology, Tianjin Key Lab of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
5
|
Spoleti E, La Barbera L, Cauzzi E, De Paolis ML, Saba L, Marino R, Sciamanna G, Di Lazzaro V, Keller F, Nobili A, Krashia P, D'Amelio M. Dopamine neuron degeneration in the Ventral Tegmental Area causes hippocampal hyperexcitability in experimental Alzheimer's Disease. Mol Psychiatry 2024; 29:1265-1280. [PMID: 38228889 PMCID: PMC11189820 DOI: 10.1038/s41380-024-02408-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/17/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024]
Abstract
Early and progressive dysfunctions of the dopaminergic system from the Ventral Tegmental Area (VTA) have been described in Alzheimer's Disease (AD). During the long pre-symptomatic phase, alterations in the function of Parvalbumin interneurons (PV-INs) are also observed, resulting in cortical hyperexcitability represented by subclinical epilepsy and aberrant gamma-oscillations. However, it is unknown whether the dopaminergic deficits contribute to brain hyperexcitability in AD. Here, using the Tg2576 mouse model of AD, we prove that reduced hippocampal dopaminergic innervation, due to VTA dopamine neuron degeneration, impairs PV-IN firing and gamma-waves, weakens the inhibition of pyramidal neurons and induces hippocampal hyperexcitability via lower D2-receptor-mediated activation of the CREB-pathway. These alterations coincide with reduced PV-IN numbers and Perineuronal Net density. Importantly, L-DOPA and the selective D2-receptor agonist quinpirole rescue p-CREB levels and improve the PV-IN-mediated inhibition, thus reducing hyperexcitability. Moreover, similarly to quinpirole, sumanirole - another D2-receptor agonist and a known anticonvulsant - not only increases p-CREB levels in PV-INs but also restores gamma-oscillations in Tg2576 mice. Conversely, blocking the dopaminergic transmission with sulpiride (a D2-like receptor antagonist) in WT mice reduces p-CREB levels in PV-INs, mimicking what occurs in Tg2576. Overall, these findings support the hypothesis that the VTA dopaminergic system integrity plays a key role in hippocampal PV-IN function and survival, disclosing a relevant contribution of the reduced dopaminergic tone to aberrant gamma-waves, hippocampal hyperexcitability and epileptiform activity in early AD.
Collapse
Affiliation(s)
- Elena Spoleti
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Livia La Barbera
- 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
| | - Emma Cauzzi
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Maria Luisa De Paolis
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Luana Saba
- 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
| | - Ramona Marino
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
| | - Giuseppe Sciamanna
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- UniCamillus International University of Health Sciences, 00131, Rome, Italy
| | - Vincenzo Di Lazzaro
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128, Rome, Italy
- Operative Research Unit of Neurology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128, Rome, Italy
| | - Flavio Keller
- 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
| | - Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128, 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
|
6
|
Ruch F, Gnörich J, Wind K, Köhler M, Zatcepin A, Wiedemann T, Gildehaus FJ, Lindner S, Boening G, von Ungern-Sternberg B, Beyer L, Herms J, Bartenstein P, Brendel M, Eckenweber F. Validity and value of metabolic connectivity in mouse models of β-amyloid and tauopathy. Neuroimage 2024; 286:120513. [PMID: 38191101 DOI: 10.1016/j.neuroimage.2024.120513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/25/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Among functional imaging methods, metabolic connectivity (MC) is increasingly used for investigation of regional network changes to examine the pathophysiology of neurodegenerative diseases such as Alzheimer's disease (AD) or movement disorders. Hitherto, MC was mostly used in clinical studies, but only a few studies demonstrated the usefulness of MC in the rodent brain. The goal of the current work was to analyze and validate metabolic regional network alterations in three different mouse models of neurodegenerative diseases (β-amyloid and tau) by use of 2-deoxy-2-[18F]fluoro-d-glucose positron emission tomography (FDG-PET) imaging. We compared the results of FDG-µPET MC with conventional VOI-based analysis and behavioral assessment in the Morris water maze (MWM). The impact of awake versus anesthesia conditions on MC read-outs was studied and the robustness of MC data deriving from different scanners was tested. MC proved to be an accurate and robust indicator of functional connectivity loss when sample sizes ≥12 were considered. MC readouts were robust across scanners and in awake/ anesthesia conditions. MC loss was observed throughout all brain regions in tauopathy mice, whereas β-amyloid indicated MC loss mainly in spatial learning areas and subcortical networks. This study established a methodological basis for the utilization of MC in different β-amyloid and tau mouse models. MC has the potential to serve as a read-out of pathological changes within neuronal networks in these models.
Collapse
Affiliation(s)
- François Ruch
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Karin Wind
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Mara Köhler
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Wiedemann
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Franz-Joseph Gildehaus
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Guido Boening
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Center of Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Florian Eckenweber
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Possemato E, La Barbera L, Nobili A, Krashia P, D'Amelio M. The role of dopamine in NLRP3 inflammasome inhibition: Implications for neurodegenerative diseases. Ageing Res Rev 2023; 87:101907. [PMID: 36893920 DOI: 10.1016/j.arr.2023.101907] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/10/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023]
Abstract
In the Central Nervous System (CNS), neuroinflammation orchestrated by microglia and astrocytes is an innate immune response to counteract stressful and dangerous insults. One of the most important and best characterized players in the neuroinflammatory response is the NLRP3 inflammasome, a multiproteic complex composed by NOD-like receptor family Pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC) and pro-caspase-1. Different stimuli mediate NLRP3 activation, resulting in the NLRP3 inflammasome assembly and the pro-inflammatory cytokine (IL-1β and IL-18) maturation and secretion. The persistent and uncontrolled NLRP3 inflammasome activation has a leading role during the pathophysiology of neuroinflammation in age-related neurodegenerative diseases such as Parkinson's (PD) and Alzheimer's (AD). The neurotransmitter dopamine (DA) is one of the players that negatively modulate NLRP3 inflammasome activation through DA receptors expressed in both microglia and astrocytes. This review summarizes recent findings linking the role of DA in the modulation of NLRP3-mediated neuroinflammation in PD and AD, where early deficits of the dopaminergic system are well characterized. Highlighting the relationship between DA, its glial receptors and the NLRP3-mediated neuroinflammation can provide insights to novel diagnostic strategies in early disease phases and new pharmacological tools to delay the progression of these diseases.
Collapse
Affiliation(s)
- Elena Possemato
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Livia La Barbera
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro 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 Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Paraskevi Krashia
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano, 64, 00143 Rome, Italy; Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Álvaro del Portillo, 21, 00128 Rome, Italy.
| |
Collapse
|
8
|
La Barbera L, D'Amelio M. Alzheimer's Disease and Sex-Dependent Alterations in the Striatum: A Lesson from a Mouse Model. J Alzheimers Dis 2023; 94:1377-1380. [PMID: 37522213 DOI: 10.3233/jad-230681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
In the last years, many clinical studies highlighted sex-specific differences in the pathophysiology of Alzheimer's disease (AD). The recent paper published in the Journal of Alzheimer's Disease shows the influence of sex on amyloid-β plaque deposition, behavior, and dopaminergic signaling in the 5xFAD mouse model of AD, with worse alterations in female mice. This commentary focuses on the importance of recognizing sex as a key variable to consider for a more precise clinical practice, with the challenge to develop sex-specific therapeutic interventions in neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Livia La Barbera
- Università Campus Bio-Medico di Roma, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marcello D'Amelio
- Università Campus Bio-Medico di Roma, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
9
|
Guo Z, Liu K, Li J, Zhu H, Chen B, Liu X. Disrupted topological organization of functional brain networks in Alzheimer's disease patients with depressive symptoms. BMC Psychiatry 2022; 22:810. [PMID: 36539729 PMCID: PMC9764564 DOI: 10.1186/s12888-022-04450-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Depression is a common symptom of Alzheimer's disease (AD), but the underlying neural mechanism is unknown. The aim of this study was to explore the topological properties of AD patients with depressive symptoms (D-AD) using graph theoretical analysis. METHODS We obtained 3-Tesla rsfMRI data from 24 D-AD patients, 20 non-depressed AD patients (nD-AD), and 20 normal controls (NC). Resting state networks were identified using graph theory analysis. ANOVA with a two-sample t-test post hoc analysis in GRETNA was used to assess the topological measurements. RESULTS Our results demonstrate that the three groups show characteristic properties of a small-world network. NCs showed significantly larger global and local efficiency than D-AD and nD-AD patients. Compared with nD-AD patients, D-AD patients showed decreased nodal centrality in the pallidum, putamen, and right superior temporal gyrus. They also showed increased nodal centrality in the right superior parietal gyrus, the medial orbital portion of the right superior frontal gyrus, and the orbital portion of the right superior frontal gyrus. Compared with nD-AD patients, NC showed decreased nodal betweenness in the right superior temporal gyrus, and increased nodal betweenness in medial orbital part of the right superior frontal gyrus. CONCLUSIONS These results indicate that D-AD is associated with alterations of topological structure. Our study provides new insights into the brain mechanisms underlying D-AD.
Collapse
Affiliation(s)
- Zhongwei Guo
- grid.417168.d0000 0004 4666 9789Tongde Hospital of Zhejiang Province, Zhejiang Provincial Health Commission, Hangzhou, 310012 China
| | - Kun Liu
- grid.417384.d0000 0004 1764 2632The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027 China
| | - Jiapeng Li
- grid.417168.d0000 0004 4666 9789Tongde Hospital of Zhejiang Province, Zhejiang Provincial Health Commission, Hangzhou, 310012 China
| | - Haokai Zhu
- grid.268505.c0000 0000 8744 8924The Second Clinical Medical College, Zhejiang Chinese Medicine University, Hangzhou, 310000 China
| | - Bo Chen
- Tongde Hospital of Zhejiang Province, Zhejiang Provincial Health Commission, Hangzhou, 310012, China.
| | - Xiaozheng Liu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
10
|
Caligiore D, Giocondo F, Silvetti M. The Neurodegenerative Elderly Syndrome (NES) hypothesis: Alzheimer and Parkinson are two faces of the same disease. IBRO Neurosci Rep 2022; 13:330-343. [PMID: 36247524 PMCID: PMC9554826 DOI: 10.1016/j.ibneur.2022.09.007] [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: 03/14/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) and Parkinson's disease (PD) share monoamine and alpha-synuclein (αSyn) dysfunctions, often beginning years before clinical manifestations onset. The triggers for these impairments and the causes leading these early neurodegenerative processes to become AD or PD remain unclear. We address these issues by proposing a radically new perspective to frame AD and PD: they are different manifestations of one only disease we call "Neurodegenerative Elderly Syndrome (NES)". NES goes through three phases. The seeding stage, which starts years before clinical signs, and where the part of the brain-body affected by the initial αSyn and monoamine dysfunctions, influences the future possible progression of NES towards PD or AD. The compensatory stage, where the clinical symptoms are still silent thanks to compensatory mechanisms keeping monoamine concentrations homeostasis. The bifurcation stage, where NES becomes AD or PD. We present recent literature supporting NES and discuss how this hypothesis could radically change the comprehension of AD and PD comorbidities and the design of novel system-level diagnostic and therapeutic actions.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Via Sebino 32, Rome 00199, Italy
| | - Flora Giocondo
- Laboratory of Embodied Natural and Artificial Intelligence, Institute of Cognitive Sciences and Technologies, National Research Council (LENAI-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| | - Massimo Silvetti
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Via San Martino della Battaglia 44, Rome 00185, Italy
| |
Collapse
|
11
|
La Barbera L, Nobili A, Cauzzi E, Paoletti I, Federici M, Saba L, Giacomet C, Marino R, Krashia P, Melone M, Keller F, Mercuri NB, Viscomi MT, Conti F, D’Amelio M. Upregulation of Ca 2+-binding proteins contributes to VTA dopamine neuron survival in the early phases of Alzheimer's disease in Tg2576 mice. Mol Neurodegener 2022; 17:76. [PMID: 36434727 PMCID: PMC9700939 DOI: 10.1186/s13024-022-00580-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 10/31/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Recent clinical and experimental studies have highlighted the involvement of Ventral Tegmental Area (VTA) dopamine (DA) neurons for the early pathogenesis of Alzheimer's Disease (AD). We have previously described a progressive and selective degeneration of these neurons in the Tg2576 mouse model of AD, long before amyloid-beta plaque formation. The degenerative process in DA neurons is associated with an autophagy flux impairment, whose rescue can prevent neuronal loss. Impairments in autophagy can be the basis for accumulation of damaged mitochondria, leading to disturbance in calcium (Ca2+) homeostasis, and to functional and structural deterioration of DA neurons. METHODS In Tg2576 mice, we performed amperometric recordings of DA levels and analysis of dopaminergic fibers in the Nucleus Accumbens - a major component of the ventral striatum precociously affected in AD patients - together with retrograde tracing, to identify the most vulnerable DA neuron subpopulations in the VTA. Then, we focused on these neurons to analyze mitochondrial integrity and Apoptosis-inducing factor (AIF) localization by electron and confocal microscopy, respectively. Stereological cell count was also used to evaluate degeneration of DA neuron subpopulations containing the Ca2+-binding proteins Calbindin-D28K and Calretinin. The expression levels for these proteins were analyzed by western blot and confocal microscopy. Lastly, using electrophysiology and microfluorometry we analyzed VTA DA neuron intrinsic properties and cytosolic free Ca2+ levels. RESULTS We found a progressive degeneration of mesolimbic DA neurons projecting to the ventral striatum, located in the paranigral nucleus and parabrachial pigmented subnucleus of the VTA. At the onset of degeneration (3 months of age), the vulnerable DA neurons in the Tg2576 accumulate damaged mitochondria, while AIF translocates from the mitochondria to the nucleus. Although we describe an age-dependent loss of the DA neurons expressing Calbindin-D28K or Calretinin, we observed that the remaining cells upregulate the levels of Ca2+-binding proteins, and the free cytosolic levels of Ca2+ in these neurons are significantly decreased. Coherently, TUNEL-stained Tg2576 DA neurons express lower levels of Calbindin-D28K when compared with non-apoptotic cells. CONCLUSION Overall, our results suggest that the overexpression of Ca2+-binding proteins in VTA DA neurons might be an attempt of cells to survive by increasing their ability to buffer free Ca2+. Exploring strategies to overexpress Ca2+-binding proteins could be fundamental to reduce neuronal suffering and improve cognitive and non-cognitive functions in AD.
Collapse
Affiliation(s)
- Livia La Barbera
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Annalisa Nobili
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Emma Cauzzi
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Paoletti
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mauro Federici
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Luana Saba
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Cecilia Giacomet
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ramona Marino
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Paraskevi Krashia
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.9657.d0000 0004 1757 5329Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Marcello Melone
- grid.7010.60000 0001 1017 3210Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020 Ancona, Italy ,Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020 Ancona, Italy
| | - Flavio Keller
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Nicola Biagio Mercuri
- grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy ,grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Maria Teresa Viscomi
- grid.8142.f0000 0001 0941 3192Department of Life Science and Public Health; Section of Histology and Embryology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy ,grid.414603.4Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy
| | - Fiorenzo Conti
- grid.7010.60000 0001 1017 3210Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020 Ancona, Italy ,Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020 Ancona, Italy ,grid.7010.60000 0001 1017 3210Foundation for Molecular Medicine, Università Politecnica delle Marche, 60020 Ancona, Italy
| | - Marcello D’Amelio
- grid.9657.d0000 0004 1757 5329Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy ,grid.417778.a0000 0001 0692 3437Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| |
Collapse
|
12
|
Comparative Metagenomics and Metabolomes Reveals Abnormal Metabolism Activity Is Associated with Gut Microbiota in Alzheimer's Disease Mice. Int J Mol Sci 2022; 23:ijms231911560. [PMID: 36232865 PMCID: PMC9569518 DOI: 10.3390/ijms231911560] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
A common symptom in Alzheimer's disease (AD) is cognitive decline, of which the potential pathogenesis remains unclear. In order to understand the mechanism of gut microbiota in AD, it is necessary to clarify the relationship between gut microbiota and metabolites. Behavioral tests, pathological examination, metagenomics, and metabolomics were applied to analyze the difference of gut microbiota and metabolome between APPswe/PS1ΔE9 (PAP) mice with cognitive decline and age-matched controls, and their possible correlations. Our results showed that PAP mice and health mice had different structures of the bacterial communities in the gut. The abundances and diversities of the bacterial communities in health mice were higher than in PAP mice by metagenomics analysis. The abundances of Libanicoccus massiliensis, Paraprevotella clara, and Lactobacillus amylovorus were significantly increased in PAP mice, while the abundances of Turicibacter sanguinis, Dubosiella newyorkensis, and Prevotella oris were greatly reduced. Furthermore, PAP mice possessed peculiar metabolic phenotypes in stool, serum, and hippocampus relative to WT mice, as is demonstrated by alterations in neurotransmitters metabolism, lipid metabolism, aromatic amino acids metabolism, energy metabolism, vitamin digestion and absorption, and bile metabolism. Microbiota-host metabolic correlation analysis suggests that abnormal metabolism in stool, serum, and hippocampus of PAP mice may be modulated by the gut microbiota, especially T. sanguinis, D. newyorkensis, and P. oris. Therefore, abnormal metabolism activity is associated with gut microbiota in Alzheimer's disease mice. Our results imply that modifying host metabolism through targeting gut microbiota may be a novel and viable strategy for the prevention and treatment of AD in the future.
Collapse
|
13
|
La Barbera L, Mauri E, D’Amelio M, Gori M. Functionalization strategies of polymeric nanoparticles for drug delivery in Alzheimer's disease: Current trends and future perspectives. Front Neurosci 2022; 16:939855. [PMID: 35992936 PMCID: PMC9387393 DOI: 10.3389/fnins.2022.939855] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, is a progressive and multifactorial neurodegenerative disorder whose primary causes are mostly unknown. Due to the increase in life expectancy of world population, including developing countries, AD, whose incidence rises dramatically with age, is at the forefront among neurodegenerative diseases. Moreover, a definitive cure is not yet within reach, imposing substantial medical and public health burdens at every latitude. Therefore, the effort to devise novel and effective therapeutic strategies is still of paramount importance. Genetic, functional, structural and biochemical studies all indicate that new and efficacious drug delivery strategies interfere at different levels with various cellular and molecular targets. Over the last few decades, therapeutic development of nanomedicine at preclinical stage has shown to progress at a fast pace, thus paving the way for its potential impact on human health in improving prevention, diagnosis, and treatment of age-related neurodegenerative disorders, including AD. Clinical translation of nano-based therapeutics, despite current limitations, may present important advantages and innovation to be exploited in the neuroscience field as well. In this state-of-the-art review article, we present the most promising applications of polymeric nanoparticle-mediated drug delivery for bypassing the blood-brain barrier of AD preclinical models and boost pharmacological safety and efficacy. In particular, novel strategic chemical functionalization of polymeric nanocarriers that could be successfully employed for treating AD are thoroughly described. Emphasis is also placed on nanotheranostics as both potential therapeutic and diagnostic tool for targeted treatments. Our review highlights the emerging role of nanomedicine in the management of AD, providing the readers with an overview of the nanostrategies currently available to develop future therapeutic applications against this chronic neurodegenerative disease.
Collapse
Affiliation(s)
- Livia La Barbera
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Santa Lucia Foundation, IRCSS, Rome, Italy
| | - Emanuele Mauri
- Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marcello D’Amelio
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Santa Lucia Foundation, IRCSS, Rome, Italy
| | - Manuele Gori
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Institute of Biochemistry and Cell Biology (IBBC) - National Research Council (CNR), Rome, Italy
| |
Collapse
|
14
|
Kim S, Nam Y, Kim HS, Jung H, Jeon SG, Hong SB, Moon M. Alteration of Neural Pathways and Its Implications in Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10040845. [PMID: 35453595 PMCID: PMC9025507 DOI: 10.3390/biomedicines10040845] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease accompanied by cognitive and behavioral symptoms. These AD-related manifestations result from the alteration of neural circuitry by aggregated forms of amyloid-β (Aβ) and hyperphosphorylated tau, which are neurotoxic. From a neuroscience perspective, identifying neural circuits that integrate various inputs and outputs to determine behaviors can provide insight into the principles of behavior. Therefore, it is crucial to understand the alterations in the neural circuits associated with AD-related behavioral and psychological symptoms. Interestingly, it is well known that the alteration of neural circuitry is prominent in the brains of patients with AD. Here, we selected specific regions in the AD brain that are associated with AD-related behavioral and psychological symptoms, and reviewed studies of healthy and altered efferent pathways to the target regions. Moreover, we propose that specific neural circuits that are altered in the AD brain can be potential targets for AD treatment. Furthermore, we provide therapeutic implications for targeting neuronal circuits through various therapeutic approaches and the appropriate timing of treatment for AD.
Collapse
Affiliation(s)
- Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Hyeon soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Haram Jung
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea; (S.K.); (Y.N.); (H.s.K.); (H.J.); (S.G.J.); (S.B.H.)
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
- Correspondence:
| |
Collapse
|
15
|
Krashia P, Spoleti E, D'Amelio M. The VTA dopaminergic system as diagnostic and therapeutical target for Alzheimer's disease. Front Psychiatry 2022; 13:1039725. [PMID: 36325523 PMCID: PMC9618946 DOI: 10.3389/fpsyt.2022.1039725] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Neuropsychiatric symptoms (NPS) occur in nearly all patients with Alzheimer's Disease (AD). Most frequently they appear since the mild cognitive impairment (MCI) stage preceding clinical AD, and have a prognostic importance. Unfortunately, these symptoms also worsen the daily functioning of patients, increase caregiver stress and accelerate the disease progression from MCI to AD. Apathy and depression are the most common of these NPS, and much attention has been given in recent years to understand the biological mechanisms related to their appearance in AD. Although for many decades these symptoms have been known to be related to abnormalities of the dopaminergic ventral tegmental area (VTA), a direct association between deficits in the VTA and NPS in AD has never been investigated. Fortunately, this scenario is changing since recent studies using preclinical models of AD, and clinical studies in MCI and AD patients demonstrated a number of functional, structural and metabolic alterations affecting the VTA dopaminergic neurons and their mesocorticolimbic targets. These findings appear early, since the MCI stage, and seem to correlate with the appearance of NPS. Here, we provide an overview of the recent evidence directly linking the dopaminergic VTA with NPS in AD and propose a setting in which the precocious identification of dopaminergic deficits can be a helpful biomarker for early diagnosis. In this scenario, treatments of patients with dopaminergic drugs might slow down the disease progression and delay the impairment of daily living activities.
Collapse
Affiliation(s)
- Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy.,Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Elena Spoleti
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, 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
| |
Collapse
|
16
|
Spoleti E, Krashia P, La Barbera L, Nobili A, Lupascu CA, Giacalone E, Keller F, Migliore M, Renzi M, D'Amelio M. Early derailment of firing properties in CA1 pyramidal cells of the ventral hippocampus in an Alzheimer's disease mouse model. Exp Neurol 2021; 350:113969. [PMID: 34973962 DOI: 10.1016/j.expneurol.2021.113969] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/06/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022]
Abstract
Gradual decline in cognitive and non-cognitive functions are considered clinical hallmarks of Alzheimer's Disease (AD). Post-mortem autoptic analysis shows the presence of amyloid β deposits, neuroinflammation and severe brain atrophy. However, brain circuit alterations and cellular derailments, assessed in very early stages of AD, still remain elusive. The understanding of these early alterations is crucial to tackle defective mechanisms. In a previous study we proved that the Tg2576 mouse model of AD displays functional deficits in the dorsal hippocampus and relevant behavioural AD-related alterations. We had shown that these deficits in Tg2576 mice correlate with the precocious degeneration of dopamine (DA) neurons in the Ventral Tegmental Area (VTA) and can be restored by L-DOPA treatment. Due to the distinct functionality and connectivity of dorsal versus ventral hippocampus, here we investigated neuronal excitability and synaptic functionality in the ventral CA1 hippocampal sub-region of Tg2576 mice. We found an age-dependent alteration of cell excitability and firing in pyramidal neurons starting at 3 months of age, that correlates with reduced levels in the ventral CA1 of tyrosine hydroxylase - the rate-limiting enzyme of DA synthesis. Additionally, at odds with the dorsal hippocampus, we found no alterations in basal glutamatergic transmission and long-term plasticity of ventral neurons in 8-month old Tg2576 mice compared to age-matched controls. Last, we used computational analysis to model the early derailments of firing properties observed and hypothesize that the neuronal alterations found could depend on dysfunctional sodium and potassium conductances, leading to anticipated depolarization-block of action potential firing. The present study depicts that impairment of cell excitability and homeostatic control of firing in ventral CA1 pyramidal neurons is a prodromal feature in Tg2576 AD mice.
Collapse
Affiliation(s)
- Elena Spoleti
- Faculty of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, Rome 00128, Italy
| | - Paraskevi Krashia
- Faculty of Medicine and Surgery, University Campus Bio-Medico, Rome 00128, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome 00143, Italy
| | - Livia La Barbera
- Faculty of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, Rome 00128, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome 00143, Italy
| | - Annalisa Nobili
- Faculty of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, Rome 00128, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome 00143, Italy
| | | | | | - Flavio Keller
- Faculty of Medicine and Surgery, University Campus Bio-Medico, Rome 00128, Italy
| | - Michele Migliore
- Institute of Biophysics, National Research Council, Palermo 90146, Italy
| | - Massimiliano Renzi
- Department of Physiology and Pharmacology, Sapienza University, Rome 00185, Italy.
| | - Marcello D'Amelio
- Faculty of Medicine and Surgery, University Campus Bio-Medico, Rome 00128, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome 00143, Italy.
| |
Collapse
|
17
|
Sala A, Caminiti SP, Presotto L, Pilotto A, Liguori C, Chiaravalloti A, Garibotto V, Frisoni GB, D'Amelio M, Paghera B, Schillaci O, Mercuri N, Padovani A, Perani D. In vivo human molecular neuroimaging of dopaminergic vulnerability along the Alzheimer's disease phases. Alzheimers Res Ther 2021; 13:187. [PMID: 34772450 PMCID: PMC8588696 DOI: 10.1186/s13195-021-00925-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 10/18/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Preclinical and pathology evidence suggests an involvement of brain dopamine (DA) circuitry in Alzheimer's disease (AD). We in vivo investigated if, when, and in which target regions [123I]FP-CIT-SPECT regional binding and molecular connectivity are damaged along the AD course. METHODS We retrospectively selected 16 amyloid-positive subjects with mild cognitive impairment due to AD (AD-MCI), 22 amyloid-positive patients with probable AD dementia (AD-D), and 74 healthy controls, all with available [123I]FP-CIT-SPECT imaging. We tested whether nigrostriatal vs. mesocorticolimbic dopaminergic targets present binding potential loss, via MANCOVA, and alterations in molecular connectivity, via partial correlation analysis. Results were deemed significant at p < 0.05, after Bonferroni correction for multiple comparisons. RESULTS We found significant reductions of [123I]FP-CIT binding in both AD-MCI and AD-D compared to controls. Binding reductions were prominent in the major targets of the ventrotegmental-mesocorticolimbic pathway, namely the ventral striatum and the hippocampus, in both clinical groups, and in the cingulate gyrus, in patients with dementia only. Within the nigrostriatal projections, only the dorsal caudate nucleus showed reduced [123I]FP-CIT binding, in both groups. Molecular connectivity assessment revealed a widespread loss of inter-connections among subcortical and cortical targets of the mesocorticolimbic network only (poor overlap with the control group as expressed by a Dice coefficient ≤ 0.25) and no alterations of the nigrostriatal network (high overlap with controls, Dice coefficient = 1). CONCLUSION Local- and system-level alterations of the mesocorticolimbic dopaminergic circuitry characterize AD, already in prodromal disease phases. These results might foster new therapeutic strategies for AD. The clinical correlates of these findings deserve to be carefully considered within the emergence of both neuropsychiatric symptoms and cognitive deficits.
Collapse
Affiliation(s)
- Arianna Sala
- Vita-Salute San Raffaele University, Via Olgettina 60, Milan, 20132, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Via Olgettina 60, Milan, 20132, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Luca Presotto
- Nuclear Medicine Unit, San Raffaele Hospital, 20132, Milan, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25121, Brescia, Italy
- Parkinson's Disease Rehabilitation Centre, FERB ONLUS - S. Isidoro Hospital, 24069, Trescore Balneario, Italy
| | - Claudio Liguori
- Division of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133, Rome, Italy
- IRCCS Neuromed, 86077, Pozzilli, Italy
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, University Hospitals of Geneva, and NIMTLab, Faculty of Medicine, Geneva University, 1205, Geneva, Switzerland
| | - Giovanni Battista Frisoni
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, University Hospitals of Geneva, and NIMTLab, Faculty of Medicine, Geneva University, 1205, Geneva, Switzerland
- Memory Clinic and LANVIE-Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, 1205, Geneva, Switzerland
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
- Unit of Molecular Neurosciences, Department of Medicine, University Campus-Biomedico, 00128, Rome, Italy
| | - Barbara Paghera
- Nuclear Medicine Unit, Spedali Civili Brescia, 25123, Brescia, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00133, Rome, Italy
- IRCCS Neuromed, 86077, Pozzilli, Italy
| | - Nicola Mercuri
- Division of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00179, Rome, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, 25121, Brescia, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Via Olgettina 60, Milan, 20132, Italy.
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy.
- Nuclear Medicine Unit, San Raffaele Hospital, 20132, Milan, Italy.
| |
Collapse
|
18
|
Caligiore D, Silvetti M, D'Amelio M, Puglisi-Allegra S, Baldassarre G. Computational Modeling of Catecholamines Dysfunction in Alzheimer's Disease at Pre-Plaque Stage. J Alzheimers Dis 2021; 77:275-290. [PMID: 32741822 PMCID: PMC7592658 DOI: 10.3233/jad-200276] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Alzheimer’s disease (AD) etiopathogenesis remains partially unexplained. The main conceptual framework used to study AD is the Amyloid Cascade Hypothesis, although the failure of recent clinical experimentation seems to reduce its potential in AD research. Objective: A possible explanation for the failure of clinical trials is that they are set too late in AD progression. Recent studies suggest that the ventral tegmental area (VTA) degeneration could be one of the first events occurring in AD progression (pre-plaque stage). Methods: Here we investigate this hypothesis through a computational model and computer simulations validated with behavioral and neural data from patients. Results: We show that VTA degeneration might lead to system-level adjustments of catecholamine release, triggering a sequence of events leading to relevant clinical and pathological signs of AD. These changes consist first in a midfrontal-driven compensatory hyperactivation of both VTA and locus coeruleus (norepinephrine) followed, with the progression of the VTA impairment, by a downregulation of catecholamine release. These processes could then trigger the neural degeneration at the cortical and hippocampal levels, due to the chronic loss of the neuroprotective role of norepinephrine. Conclusion: Our novel hypothesis might contribute to the formulation of a wider system-level view of AD which might help to devise early diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory (CTNLab), Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Massimo Silvetti
- Computational and Translational Neuroscience Laboratory (CTNLab), Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Marcello D'Amelio
- Unit of Molecular Neurosciences, Department of Medicine, University Campus-Biomedico, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | | | - Gianluca Baldassarre
- Laboratory of Computational Embodied Neuroscience (LOCEN), Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
19
|
Serra L, D'Amelio M, Esposito S, Di Domenico C, Koch G, Marra C, Mercuri NB, Caltagirone C, Artusi CA, Lopiano L, Cercignani M, Bozzali M. Ventral Tegmental Area Disconnection Contributes Two Years Early to Correctly Classify Patients Converted to Alzheimer's Disease: Implications for Treatment. J Alzheimers Dis 2021; 82:985-1000. [PMID: 34120905 DOI: 10.3233/jad-210171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent cross-sectional studies highlighted the loss of dopaminergic neurons in the ventral tegmental area (VTA) as an early pathophysiological event in Alzheimer's disease (AD). OBJECTIVE In this study, we longitudinally investigated by resting-state fMRI (rs-fMRI) a cohort of patients with mild cognitive impairment (MCI) due to AD to evaluate the impact of VTA disconnection in predicting the conversion to AD. METHODS A cohort of 35 patients with MCI due to AD were recruited and followed-up for 24 months. They underwent cognitive evaluation and rs-fMRI to assess VTA connectivity at baseline and at follow-up. RESULTS At 24-month follow-up, 16 out of 35 patients converted to AD. Although converters and non-converters to AD did not differ in demographic and behavioral characteristics at baseline, the first group showed a significant reduction of VTA-driven connectivity in the posterior cingulate and precentral cortex. This pattern of additional disconnection in MCI-Converters compared to non-converters remained substantially unchanged at 24-month follow-up. CONCLUSION This study reinforces the hypothesis of an early contribution of dopaminergic dysfunction to AD evolution by targeting the default-mode network. These results have potential implications for AD staging and prognosis and support new opportunities for therapeutic interventions to slow down disease progression.
Collapse
Affiliation(s)
- Laura Serra
- Neuroimaging Laboratory, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Marcello D'Amelio
- Laboratory Molecular Neurosciences, Fondazione Santa Lucia, IRCCS, Rome, Italy.,Unit of Molecular Neurosciences, Department of Medicine, University Campus-Biomedico, Rome, Italy
| | - Sharon Esposito
- Neuroimaging Laboratory, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | | | - Giacomo Koch
- Non Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Fondazione Santa Lucia, IRCCS, Rome, Italy.,Section of Human Physiology, University of Ferrara, Ferrara, Italy
| | - Camillo Marra
- Institute of Neurology, Catholic University, Rome, Italy
| | - Nicola Biagio Mercuri
- Laboratory of Experimental Neurology, Fondazione Santa Lucia, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavioural Neurology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Carlo Alberto Artusi
- 'Rita Levi Montalcini' Department of Neuroscience University of Torino, Turin, Italy
| | - Leonardo Lopiano
- 'Rita Levi Montalcini' Department of Neuroscience University of Torino, Turin, Italy
| | - Mara Cercignani
- Neuroimaging Laboratory, Fondazione Santa Lucia, IRCCS, Rome, Italy.,Cardiff University Brain Imaging Centre, School of Psychology, Cardiff University, Cardiff, Wales, United Kingdom
| | - Marco Bozzali
- 'Rita Levi Montalcini' Department of Neuroscience University of Torino, Turin, Italy.,Department of Neuroscience, Brighton & Sussex Medical School, University of Sussex, Brighton, East Sussex, United Kingdom
| |
Collapse
|
20
|
Patthy Á, Murai J, Hanics J, Pintér A, Zahola P, Hökfelt TGM, Harkany T, Alpár A. Neuropathology of the Brainstem to Mechanistically Understand and to Treat Alzheimer's Disease. J Clin Med 2021; 10:jcm10081555. [PMID: 33917176 PMCID: PMC8067882 DOI: 10.3390/jcm10081555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder as yet without effective therapy. Symptoms of this disorder typically reflect cortical malfunction with local neurohistopathology, which biased investigators to search for focal triggers and molecular mechanisms. Cortex, however, receives massive afferents from caudal brain structures, which do not only convey specific information but powerfully tune ensemble activity. Moreover, there is evidence that the start of AD is subcortical. The brainstem harbors monoamine systems, which establish a dense innervation in both allo- and neocortex. Monoaminergic synapses can co-release neuropeptides either by precisely terminating on cortical neurons or, when being “en passant”, can instigate local volume transmission. Especially due to its early damage, malfunction of the ascending monoaminergic system emerges as an early sign and possible trigger of AD. This review summarizes the involvement and cascaded impairment of brainstem monoaminergic neurons in AD and discusses cellular mechanisms that lead to their dysfunction. We highlight the significance and therapeutic challenges of transmitter co-release in ascending activating system, describe the role and changes of local connections and distant afferents of brainstem nuclei in AD, and summon the rapidly increasing diagnostic window during the last few years.
Collapse
Affiliation(s)
- Ágoston Patthy
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Murai
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - János Hanics
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
| | - Anna Pintér
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Péter Zahola
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
| | - Tomas G. M. Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
| | - Tibor Harkany
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, 17165 Stockholm, Sweden; (T.G.M.H.); (T.H.)
- Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, 1090 Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, H-1094 Budapest, Hungary; (Á.P.); (J.M.); (J.H.); (A.P.); (P.Z.)
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, H-1094 Budapest, Hungary
- Correspondence:
| |
Collapse
|
21
|
Carli G, Tondo G, Boccalini C, Perani D. Brain Molecular Connectivity in Neurodegenerative Conditions. Brain Sci 2021; 11:brainsci11040433. [PMID: 33800680 PMCID: PMC8067093 DOI: 10.3390/brainsci11040433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
Positron emission tomography (PET) allows for the in vivo assessment of early brain functional and molecular changes in neurodegenerative conditions, representing a unique tool in the diagnostic workup. The increased use of multivariate PET imaging analysis approaches has provided the chance to investigate regional molecular processes and long-distance brain circuit functional interactions in the last decade. PET metabolic and neurotransmission connectome can reveal brain region interactions. This review is an overview of concepts and methods for PET molecular and metabolic covariance assessment with evidence in neurodegenerative conditions, including Alzheimer’s disease and Lewy bodies disease spectrum. We highlight the effects of environmental and biological factors on brain network organization. All of the above might contribute to innovative diagnostic tools and potential disease-modifying interventions.
Collapse
Affiliation(s)
- Giulia Carli
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Giacomo Tondo
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Cecilia Boccalini
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Daniela Perani
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
- Nuclear Medicine Unit, San Raffaele Hospital, 20121 Milan, Italy
- Correspondence: ; Tel.: +39-02-26432224
| |
Collapse
|
22
|
La Barbera L, Vedele F, Nobili A, Krashia P, Spoleti E, Latagliata EC, Cutuli D, Cauzzi E, Marino R, Viscomi MT, Petrosini L, Puglisi-Allegra S, Melone M, Keller F, Mercuri NB, Conti F, D'Amelio M. Nilotinib restores memory function by preventing dopaminergic neuron degeneration in a mouse model of Alzheimer's Disease. Prog Neurobiol 2021; 202:102031. [PMID: 33684513 DOI: 10.1016/j.pneurobio.2021.102031] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/15/2021] [Accepted: 02/28/2021] [Indexed: 01/18/2023]
Abstract
What happens precociously to the brain destined to develop Alzheimer's Disease (AD) still remains to be elucidated and this is one reason why effective AD treatments are missing. Recent experimental and clinical studies indicate that the degeneration of the dopaminergic (DA) neurons in the Ventral Tegmental Area (VTA) could be one of the first events occurring in AD. However, the causes of the increased vulnerability of DA neurons in AD are missing. Here, we deeply investigate the physiology of DA neurons in the VTA before, at the onset, and after onset of VTA neurodegeneration. We use the Tg2576 mouse model of AD, overexpressing a mutated form of the human APP, to identify molecular targets that can be manipulated pharmacologically. We show that in Tg2576 mice, DA neurons of the VTA at the onset of degeneration undergo slight but functionally relevant changes in their electrophysiological properties and cell morphology. Importantly, these changes are associated with accumulation of autophagosomes, suggestive of a dysfunctional autophagy, and with enhanced activation of c-Abl, a tyrosine kinase previously implicated in the pathogenesis of neurodegenerative diseases. Chronic treatment of Tg2576 mice with Nilotinib, a validated c-Abl inhibitor, reduces c-Abl phosphorylation, improves autophagy, reduces Aβ levels and - more importantly - prevents degeneration as well as functional and morphological alterations in DA neurons of the VTA. Interestingly, the drug prevents the reduction of DA outflow to the hippocampus and ameliorates hippocampal-related cognitive functions. Our results strive to identify early pathological brain changes in AD, to provide a rational basis for new therapeutic interventions able to slow down the disease progression.
Collapse
Affiliation(s)
- Livia La Barbera
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Francescangelo Vedele
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Annalisa Nobili
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Paraskevi Krashia
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| | - Elena Spoleti
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | | | - Debora Cutuli
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Psychology, Sapienza University of Rome, 00185, Rome, Italy
| | - Emma Cauzzi
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Ramona Marino
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Life Science and Public Health Section of Histology and Embryology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Laura Petrosini
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | | | - Marcello Melone
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020, Ancona, Italy; Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020, Ancona, Italy
| | - Flavio Keller
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche (UNIVPM), 60020, Ancona, Italy; Center for Neurobiology of Aging, IRCCS Istituto Nazionale Ricovero e Cura Anziani (INRCA), 60020, Ancona, Italy; Foundation for Molecular Medicine, Università Politecnica delle Marche, 60020, Ancona, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, Department of Sciences and Technologies for Humans and Environment, University Campus Bio-Medico, 00128, Rome, Italy; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.
| |
Collapse
|
23
|
Solmi M, Bodini L, Cocozza S, Seeman MV, Vieta E, Dragioti E, Carvalho AF, Fusar-Poli P. Aripiprazole monotherapy as transdiagnostic intervention for the treatment of mental disorders: An umbrella review according to TRANSD criteria. Eur Neuropsychopharmacol 2020; 41:16-27. [PMID: 33077324 DOI: 10.1016/j.euroneuro.2020.09.635] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/12/2020] [Accepted: 09/21/2020] [Indexed: 12/26/2022]
Abstract
Aripiprazole is approved in different countries as treatment for several mental disorders, but its transdiagnostic potential has not yet been assessed according to established criteria such as the recently proposed TRANSD criteria. The present work aims to test whether aripiprazole monotherapy could be considered a transdiagnostic intervention. An umbrella review of meta-analyses of randomized controlled trials (RCTs) of aripiprazole monotherapy vs placebo was conducted for any disorder defined according to standard diagnostic criteria. Primary outcomes were levels of psychiatric disease-specific symptoms. TRANSD criteria were applied to assess transdiagnosticity, while the AMSTAR -2 tool was used to assess the quality of eligible meta-analyses. Four pairwise meta-analyses and three network meta-analyses were included, consisting of 25 RCTs of aripiprazole monotherapy vs. placebo (N=5469). Aripiprazole outperformed placebo on primary outcomes in Alzheimer's disease with behavioral disturbance (AD) (neuropsychiatric symptoms SMD 0.20, 95%CI 0.05-0.35, max 15 mg/day), autism spectrum disorder (ASD) (severity of symptoms SMD 0.39, 95%CI 0.30-0.48, max 15 mg/day), bipolar I disorder (BD) (severity of manic symptoms SMD 0.30, 95%CI 0.12-0.47, max 30 mg/day), and schizophrenia/schizoaffective disorder (response rate RR 0.84, 95%CI 0.78-0.92, max 30 mg/day). TRANSD criteria were met (pooled SMD 0.25, 95%CI 0.09-0.41) across these disorders Quality was high in all the included meta-analyses according to AMSTAR-2. According to TRANSD criteria, aripiprazole monotherapy is a (dose-specific) transdiagnostic intervention to treat psychiatric disease-specific symptoms across 5 diagnoses (AD, ASD, BD (mania), schizophrenia/schizoaffective disorder), across four DSM-5 diagnostic groups (neurocognitive disorders, neurodevelopmental disorders, bipolar and related disorders, schizophrenia spectrum and other psychotic disorders).
Collapse
Affiliation(s)
- Marco Solmi
- Neurosciences Department, University of Padua, Padua, Italy; Neuroscience Centre, University of Padua, Padua, Italy; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Luca Bodini
- Università degli Studi di Milano, Bicocca, Italy
| | - Susanna Cocozza
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Mary V Seeman
- Department of Psychiatry, University of Toronto, and the Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Eduard Vieta
- IDIBAPS, CIBERSAM, Clinical Institute of Neuroscience, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Elena Dragioti
- Pain and Rehabilitation Centre, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Andre F Carvalho
- Department of Psychiatry, University of Toronto, and the Centre for Addiction and Mental Health, Toronto, ON, Canada; IMPACT (Innovation in Mental and Physical Health and Clinical Treatment) Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, VIC, Australia
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London, UK; National Institute for Health Research, Maudsley Biomedical Research Centre, London, UK
| |
Collapse
|
24
|
Heredia RR, Blackburn AM, Vega LA. Moderation-Mediation Effects in Bilingualism and Cognitive Reserve. Front Psychol 2020; 11:572555. [PMID: 33101142 PMCID: PMC7554512 DOI: 10.3389/fpsyg.2020.572555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/09/2020] [Indexed: 01/01/2023] Open
Abstract
We first provide a critical review of the existing findings on bilingualism as a contributor to cognitive reserve from moderator-mediator warranting cause-effect research conclusions. We next address the question of direct or indirect effects between bilingualism and neurocognitive protective factors influencing the associated age-related mental deficits. The existing findings support bilingualism as a predictor and as a moderator. Third, we propose cognitive reserve models of bilingualism describing analytical approaches that allow testing of these models and hypotheses related to path strength and causal relationships between predictors, moderators, and mediators. Lastly and most importantly, we suggest using large datasets available via open repositories. This can aid in the testing of theoretical models, clarifying the roles of moderators and mediators, and assessing the research viability of multi-causal paths that can influence cognitive reserve. Creating collaborative datasets to test these models would greatly advance our field and identify critical variables in the study of the bilingual aging brain.
Collapse
Affiliation(s)
- Roberto R Heredia
- Department of Psychology and Communication, Texas A&M International University, Laredo, TX, United States
| | - Angélique M Blackburn
- Department of Psychology and Communication, Texas A&M International University, Laredo, TX, United States
| | - Luis A Vega
- Department of Psychology, California State University-Bakersfield, Bakersfield, CA, United States
| |
Collapse
|