1
|
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
|
2
|
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
|
3
|
Olchanyi MD, Augustinack J, Haynes RL, Lewis LD, Cicero N, Li J, Destrieux C, Folkerth RD, Kinney HC, Fischl B, Brown EN, Iglesias JE, Edlow BL. Histology-guided MRI segmentation of brainstem nuclei critical to consciousness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.26.24314117. [PMID: 39399006 PMCID: PMC11469455 DOI: 10.1101/2024.09.26.24314117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
While substantial progress has been made in mapping the connectivity of cortical networks responsible for conscious awareness, neuroimaging analysis of subcortical arousal networks that modulate arousal (i.e., wakefulness) has been limited by a lack of a robust segmentation procedures for brainstem arousal nuclei. Automated segmentation of brainstem arousal nuclei is an essential step toward elucidating the physiology of arousal in human consciousness and the pathophysiology of disorders of consciousness. We created a probabilistic atlas of brainstem arousal nuclei built on diffusion MRI scans of five ex vivo human brain specimens scanned at 750 μm isotropic resolution. Labels of arousal nuclei used to generate the probabilistic atlas were manually annotated with reference to nucleus-specific immunostaining in two of the five brain specimens. We then developed a Bayesian segmentation algorithm that utilizes the probabilistic atlas as a generative model and automatically identifies brainstem arousal nuclei in a resolution- and contrast-agnostic manner. The segmentation method displayed high accuracy in both healthy and lesioned in vivo T1 MRI scans and high test-retest reliability across both T1 and T2 MRI contrasts. Finally, we show that the segmentation algorithm can detect volumetric changes and differences in magnetic susceptibility within brainstem arousal nuclei in Alzheimer's disease and traumatic coma, respectively. We release the probabilistic atlas and Bayesian segmentation tool in FreeSurfer to advance the study of human consciousness and its disorders.
Collapse
|
4
|
Skandalakis GP, Neudorfer C, Payne CA, Bond E, Tavakkoli AD, Barrios-Martinez J, Trutti AC, Koutsarnakis C, Coenen VA, Komaitis S, Hadjipanayis CG, Stranjalis G, Yeh FC, Banihashemi L, Hong J, Lozano AM, Kogan M, Horn A, Evans LT, Kalyvas A. Establishing connectivity through microdissections of midbrain stimulation-related neural circuits. Brain 2024; 147:3083-3098. [PMID: 38808482 PMCID: PMC11370807 DOI: 10.1093/brain/awae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/15/2024] [Accepted: 04/21/2024] [Indexed: 05/30/2024] Open
Abstract
Comprehensive understanding of the neural circuits involving the ventral tegmental area is essential for elucidating the anatomofunctional mechanisms governing human behaviour, in addition to the therapeutic and adverse effects of deep brain stimulation for neuropsychiatric diseases. Although the ventral tegmental area has been targeted successfully with deep brain stimulation for different neuropsychiatric diseases, the axonal connectivity of the region is not fully understood. Here, using fibre microdissections in human cadaveric hemispheres, population-based high-definition fibre tractography and previously reported deep brain stimulation hotspots, we find that the ventral tegmental area participates in an intricate network involving the serotonergic pontine nuclei, basal ganglia, limbic system, basal forebrain and prefrontal cortex, which is implicated in the treatment of obsessive-compulsive disorder, major depressive disorder, Alzheimer's disease, cluster headaches and aggressive behaviours.
Collapse
Affiliation(s)
- Georgios P Skandalakis
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Clemens Neudorfer
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Caitlin A Payne
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Evalina Bond
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Armin D Tavakkoli
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | | | - Anne C Trutti
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam 15926, The Netherlands
| | - Christos Koutsarnakis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Volker A Coenen
- Department of Stereotactic and Functional Neurosurgery, Medical Center of the University of Freiburg, Freiburg 79106, Germany
- Medical Faculty of the University of Freiburg, Freiburg 79110, Germany
- Center for Deep Brain Stimulation, Medical Center of the University of Freiburg, Freiburg 79106, Germany
| | - Spyridon Komaitis
- Queens Medical Center, Nottingham University Hospitals NHS Foundation Trust, Nottingham NG7 2UH, UK
| | | | - George Stranjalis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Layla Banihashemi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jennifer Hong
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Andres M Lozano
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Andreas Horn
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Linton T Evans
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Aristotelis Kalyvas
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
5
|
Watamura N, Kakiya N, Fujioka R, Kamano N, Takahashi M, Nilsson P, Saito T, Iwata N, Fujisawa S, Saido TC. The dopaminergic system promotes neprilysin-mediated degradation of amyloid-β in the brain. Sci Signal 2024; 17:eadk1822. [PMID: 39106321 DOI: 10.1126/scisignal.adk1822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 04/21/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Deposition of amyloid-β (Aβ) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aβ degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aβ-degrading enzyme neprilysin and reduced the amount of Aβ deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aβ deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aβ, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.
Collapse
Affiliation(s)
- Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Mika Takahashi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 171 64, Solna, Sweden
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi 464-8601, Japan
| | - Nobuhisa Iwata
- Department of Genome-based Drug Discovery & Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8521, Japan
| | - Shigeyoshi Fujisawa
- Laboratory for Systems Neurophysiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
6
|
Li S, Xiao Z. Recent Research Progress on the Use of Transcranial Magnetic Stimulation in the Treatment of Vascular Cognitive Impairment. Neuropsychiatr Dis Treat 2024; 20:1235-1246. [PMID: 38883416 PMCID: PMC11179638 DOI: 10.2147/ndt.s467357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/01/2024] [Indexed: 06/18/2024] Open
Abstract
Vascular Cognitive Impairment (VCI) is a condition where problems with brain blood vessels lead to a decline in cognitive abilities, commonly affecting the elderly and placing a significant burden on both patients and their families. Compared to medication and surgery, Transcranial Magnetic Stimulation (TMS) is a non-invasive treatment option with fewer risks and side effects, making it particularly suitable for elderly patients. TMS not only assesses the excitability and plasticity of the cerebral cortex, but its effectiveness in treating Vascular Cognitive Impairment (VCI) and its subtypes has also been validated in numerous clinical trials worldwide. However, there is still a lack of review on the physiological mechanisms of TMS treatment for VCI and its specific clinical application parameters. Therefore, this article initially provided a brief overview of the risk factors, pathological mechanisms, and classification of VCI. Next, the article explained the potential physiological mechanisms of TMS in treating VCI, particularly its role in promoting synaptic plasticity, regulating neurotransmitter balance, and improving the function of the default mode network. Additionally, The article also summarizes the application of rTMS in treating VCI and its subtypes, VCI-related sleep disorders, and the use of TMS in follow-up studies of VCI patients, providing empirical evidence for the clinical application of TMS and rTMS technologies.
Collapse
Affiliation(s)
- Sijing Li
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Clinical Research Center for Immune‑Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Zijian Xiao
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Clinical Research Center for Immune‑Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| |
Collapse
|
7
|
Wearn A, Tremblay SA, Tardif CL, Leppert IR, Gauthier CJ, Baracchini G, Hughes C, Hewan P, Tremblay-Mercier J, Rosa-Neto P, Poirier J, Villeneuve S, Schmitz TW, Turner GR, Spreng RN. Neuromodulatory subcortical nucleus integrity is associated with white matter microstructure, tauopathy and APOE status. Nat Commun 2024; 15:4706. [PMID: 38830849 PMCID: PMC11148077 DOI: 10.1038/s41467-024-48490-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: 11/29/2023] [Accepted: 05/01/2024] [Indexed: 06/05/2024] Open
Abstract
The neuromodulatory subcortical nuclei within the isodendritic core (IdC) are the earliest sites of tauopathy in Alzheimer's disease (AD). They project broadly throughout the brain's white matter. We investigated the relationship between IdC microstructure and whole-brain white matter microstructure to better understand early neuropathological changes in AD. Using multiparametric quantitative magnetic resonance imaging we observed two covariance patterns between IdC and white matter microstructure in 133 cognitively unimpaired older adults (age 67.9 ± 5.3 years) with familial risk for AD. IdC integrity related to 1) whole-brain neurite density, and 2) neurite orientation dispersion in white matter tracts known to be affected early in AD. Pattern 2 was associated with CSF concentration of phosphorylated-tau, indicating AD specificity. Apolipoprotein-E4 carriers expressed both patterns more strongly than non-carriers. IdC microstructure variation is reflected in white matter, particularly in AD-affected tracts, highlighting an early mechanism of pathological development.
Collapse
Affiliation(s)
- Alfie Wearn
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada.
| | - Stéfanie A Tremblay
- Department of Physics, Concordia University, Montreal, H4B 1R6, QC, Canada
- Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada
- School of Health, Concordia University, Montreal, H4B 1R6, QC, Canada
| | - Christine L Tardif
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Department of Biomedical Engineering, McGill University, McGill, H3A 2B4, QC, Canada
| | - Ilana R Leppert
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Claudine J Gauthier
- Department of Physics, Concordia University, Montreal, H4B 1R6, QC, Canada
- Montreal Heart Institute, Montreal, H1T 1C8, QC, Canada
- School of Health, Concordia University, Montreal, H4B 1R6, QC, Canada
| | - Giulia Baracchini
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Colleen Hughes
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
| | - Patrick Hewan
- Department of Psychology, York University, Toronto, M3J 1P3, ON, Canada
| | | | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada
| | - Taylor W Schmitz
- Department of Physiology & Pharmacology, Western Institute for Neuroscience, Western University, London, N6A 5C1, ON, Canada
| | - Gary R Turner
- Department of Psychology, York University, Toronto, M3J 1P3, ON, Canada
| | - R Nathan Spreng
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, H3A 2B4, QC, Canada.
- McConnell Brain Imaging Centre, McGill University, Montreal, H3A 2B4, QC, Canada.
- Douglas Mental Health University Institute-Research Center, Verdun, H4H 1R3, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, H3A 1A1, QC, Canada.
| |
Collapse
|
8
|
Armijo-Weingart L, San Martin L, Gallegos S, Araya A, Konar-Nie M, Fernandez-Pérez E, Aguayo LG. Loss of glycine receptors in the nucleus accumbens and ethanol reward in an Alzheimer´s Disease mouse model. Prog Neurobiol 2024; 237:102616. [PMID: 38723884 PMCID: PMC11163974 DOI: 10.1016/j.pneurobio.2024.102616] [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: 11/29/2023] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Alterations in cognitive and non-cognitive cerebral functions characterize Alzheimer's disease (AD). Cortical and hippocampal impairments related to extracellular accumulation of Aβ in AD animal models have been extensively investigated. However, recent reports have also implicated intracellular Aβ in limbic regions, such as the nucleus accumbens (nAc). Accumbal neurons express high levels of inhibitory glycine receptors (GlyRs) that are allosterically modulated by ethanol and have a role in controlling its intake. In the present study, we investigated how GlyRs in the 2xTg mice (AD model) affect nAc functions and ethanol intake behavior. Using transgenic and control aged-matched litter mates, we found that the GlyRα2 subunit was significantly decreased in AD mice (6-month-old). We also examined intracellular calcium dynamics using the fluorescent calcium protein reporter GCaMP in slice photometry. We also found that the calcium signal mediated by GlyRs, but not GABAAR, was also reduced in AD neurons. Additionally, ethanol potentiation was significantly decreased in accumbal neurons in the AD mice. Finally, we performed drinking in the dark (DID) experiments and found that 2xTg mice consumed less ethanol on the last day of DID, in agreement with a lower blood ethanol concentration. 2xTg mice also showed lower sucrose consumption, indicating that overall food reward was altered. In conclusion, the data support the role of GlyRs in nAc neuron excitability and a decreased glycinergic activity in the 2xTg mice that might lead to impairment in reward processing at an early stage of the disease.
Collapse
Affiliation(s)
- Lorena Armijo-Weingart
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Loreto San Martin
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Scarlet Gallegos
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Anibal Araya
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Macarena Konar-Nie
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile
| | - Eduardo Fernandez-Pérez
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile; Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Chile
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Chile.
| |
Collapse
|
9
|
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: 13] [Impact Index Per Article: 13.0] [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
|
10
|
Chaves T, Török B, Fazekas CL, Correia P, Sipos E, Várkonyi D, Tóth ZE, Dóra F, Dobolyi Á, Zelena D. The Dopaminergic Cells in the Median Raphe Region Regulate Social Behavior in Male Mice. Int J Mol Sci 2024; 25:4315. [PMID: 38673899 PMCID: PMC11050709 DOI: 10.3390/ijms25084315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
According to previous studies, the median raphe region (MRR) is known to contribute significantly to social behavior. Besides serotonin, there have also been reports of a small population of dopaminergic neurons in this region. Dopamine is linked to reward and locomotion, but very little is known about its role in the MRR. To address that, we first confirmed the presence of dopaminergic cells in the MRR of mice (immunohistochemistry, RT-PCR), and then also in humans (RT-PCR) using healthy donor samples to prove translational relevance. Next, we used chemogenetic technology in mice containing the Cre enzyme under the promoter of the dopamine transporter. With the help of an adeno-associated virus, designer receptors exclusively activated by designer drugs (DREADDs) were expressed in the dopaminergic cells of the MRR to manipulate their activity. Four weeks later, we performed an extensive behavioral characterization 30 min after the injection of the artificial ligand (Clozapine-N-Oxide). Stimulation of the dopaminergic cells in the MRR decreased social interest without influencing aggression and with an increase in social discrimination. Additionally, inhibition of the same cells increased the friendly social behavior during social interaction test. No behavioral changes were detected in anxiety, memory or locomotion. All in all, dopaminergic cells were present in both the mouse and human samples from the MRR, and the manipulation of the dopaminergic neurons in the MRR elicited a specific social response.
Collapse
Affiliation(s)
- Tiago Chaves
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Bibiána Török
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Csilla Lea Fazekas
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Pedro Correia
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
- János Szentágothai School of Neurosciences, Semmelweis University, H1085 Budapest, Hungary
| | - Eszter Sipos
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| | - Dorottya Várkonyi
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| | - Zsuzsanna E. Tóth
- Laboratory of Neuroendocrinology and in Situ Hybridization, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary;
| | - Fanni Dóra
- Human Brain Tissue Bank, Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, H1094 Budapest, Hungary;
| | - Árpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, H1117 Budapest, Hungary;
| | - Dóra Zelena
- Institute of Physiology, Medical School, Centre for Neuroscience, Szentágothai Research Centre, University of Pécs, H7624 Pécs, Hungary; (T.C.); (B.T.); (C.L.F.); (P.C.); (D.V.)
- Laboratory of Behavioral and Stress Studies, Institute of Experimental Medicine, H1083 Budapest, Hungary;
| |
Collapse
|
11
|
Hu M, Xu F, Liu S, Yao Y, Xia Q, Zhu C, Zhang X, Tang H, Qaiser Z, Liu S, Tang Y. Aging pattern of the brainstem based on volumetric measurement and optimized surface shape analysis. Brain Imaging Behav 2024; 18:396-411. [PMID: 38155336 DOI: 10.1007/s11682-023-00840-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] [Accepted: 12/11/2023] [Indexed: 12/30/2023]
Abstract
The brainstem, a small and crucial structure, is connected to the cerebrum, spinal cord, and cerebellum, playing a vital role in regulating autonomic functions, transmitting motor and sensory information, and modulating cognitive processes, emotions, and consciousness. While previous research has indicated that changes in brainstem anatomy can serve as a biomarker for aging and neurodegenerative diseases, the structural changes that occur in the brainstem during normal aging remain unclear. This study aimed to examine the age- and sex-related differences in the global and local structural measures of the brainstem in 187 healthy adults (ranging in age from 18 to 70 years) using structural magnetic resonance imaging. The findings showed a significant negative age effect on the volume of the two major components of the brainstem: the medulla oblongata and midbrain. The shape analysis revealed that atrophy primarily occurs in specific structures, such as the pyramid, cerebral peduncle, superior and inferior colliculi. Surface area and shape analysis showed a trend of flattening in the aging brainstem. There were no significant differences between the sexes or sex-by-age interactions in brainstem structural measures. These findings provide a systematic description of age associations with brainstem structures in healthy adults and may provide a reference for future research on brain aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Minqi Hu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Feifei Xu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shizhou Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuan Yao
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Qing Xia
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Caiting Zhu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Xinwen Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haiyan Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Zubair Qaiser
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Shuwei Liu
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China
| | - Yuchun Tang
- Department of Anatomy and Neurobiology, Research Center for Sectional and Imaging Anatomy, Shandong Provincial Key Laboratory of Mental Disorder, Shandong Key Laboratory of Digital Human and Clinical Anatomy, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, Shandong, China.
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
12
|
López-Ortiz S, Caruso G, Emanuele E, Menéndez H, Peñín-Grandes S, Guerrera CS, Caraci F, Nisticò R, Lucia A, Santos-Lozano A, Lista S. Digging into the intrinsic capacity concept: Can it be applied to Alzheimer's disease? Prog Neurobiol 2024; 234:102574. [PMID: 38266702 DOI: 10.1016/j.pneurobio.2024.102574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Historically, aging research has largely centered on disease pathology rather than promoting healthy aging. The World Health Organization's (WHO) policy framework (2015-2030) underscores the significance of fostering the contributions of older individuals to their families, communities, and economies. The WHO has introduced the concept of intrinsic capacity (IC) as a key metric for healthy aging, encompassing five primary domains: locomotion, vitality, sensory, cognitive, and psychological. Past AD research, constrained by methodological limitations, has focused on single outcome measures, sidelining the complexity of the disease. Our current scientific milieu, however, is primed to adopt the IC concept. This is due to three critical considerations: (I) the decline in IC is linked to neurocognitive disorders, including AD, (II) cognition, a key component of IC, is deeply affected in AD, and (III) the cognitive decline associated with AD involves multiple factors and pathophysiological pathways. Our study explores the application of the IC concept to AD patients, offering a comprehensive model that could revolutionize the disease's diagnosis and prognosis. There is a dearth of information on the biological characteristics of IC, which are a result of complex interactions within biological systems. Employing a systems biology approach, integrating omics technologies, could aid in unraveling these interactions and understanding IC from a holistic viewpoint. This comprehensive analysis of IC could be leveraged in clinical settings, equipping healthcare providers to assess AD patients' health status more effectively and devise personalized therapeutic interventions in accordance with the precision medicine paradigm. We aimed to determine whether the IC concept could be extended from older individuals to patients with AD, thereby presenting a model that could significantly enhance the diagnosis and prognosis of this disease.
Collapse
Affiliation(s)
- Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | | | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Saúl Peñín-Grandes
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain
| | - Claudia Savia Guerrera
- Department of Educational Sciences, University of Catania, 95125 Catania, Italy; Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", 00133 Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, 00143 Rome, Italy
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, 28670 Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), 28029 Madrid, Spain
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), 28041 Madrid, Spain
| | - Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), 47012 Valladolid, Spain.
| |
Collapse
|
13
|
Asad N, Deodato D, Asad N, Gore S, Dore TM. Multi-Photon-Sensitive Chromophore for the Photorelease of Biologically Active Phenols. ACS Chem Neurosci 2023; 14:4163-4175. [PMID: 37988406 DOI: 10.1021/acschemneuro.3c00552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
Phenols confer bioactivity to a plethora of organic compounds. Protecting the phenolic functionality with photoremovable protecting groups (PPGs) sensitive to two-photon excitation (2PE) can block the bioactivity and provide controlled release of these compounds in a spatially and temporally restricted manner by photoactivation with IR light. To develop an efficient 2PE-sensitive PPG for releasing phenols, the (8-cyano-7-hydroxyquinolin-2-yl)methyl (CyHQ) chromophore was functionalized at the C4 position with methyl, morpholine, methoxy, para-tolyl, and 3,4,5-trimethoxyphenyl groups to provide 4-methyl-CyHQ (Me-CyHQ), 4-morpholino-CyHQ (Mor-CyHQ), 4-methoxy-CyHQ (MeO-CyHQ), 4-(p-tolyl)-CyHQ (pTol-CyHQ), and 4-(3,4,5-trimethoxyphenyl)-CyHQ (TMP-CyHQ) PPGs. The probes possess attributes useful for biological use, including high quantum yield (Φu), hydrolytic stability, and good aqueous solubility in physiological conditions. The MeO-CyHQ PPG enhanced the two-photon uncaging action cross section (δu) of dopamine 3.5-fold (0.85 GM) compared to CyHQ (0.24 GM) at 740 nm and 1.49 GM at 720 nm. MeO-CyHQ was used to mediate photoactivation via 2PE of serotonin, rotigotine, N-vanillyl-nonanoylamide (VNA) (a capsaicin analogue), and eugenol. The constructs except rotigotine showed excellent efficiency in 2PE with δu ranging from 0.75 to 1.01 GM at 740 nm and from 1.31 to 1.36 GM at 720 nm high yielding release of the payloads. These probes also performed well by using conventional single photon excitation (1PE). The spatially and temporally controlled release of dopamine from CyHQ-DA and MeO-CyHQ-DA and serotonin (5-HT) from MeO-CyHQ-5HT was quantified in cell culture by using genetically encoded sensors for dopamine and serotonin, respectively. Calcium imaging was employed to quantify the release of VNA and eugenol (EG) from MeO-CyHQ-VNA and MeO-CyHQ-EG, respectively. These tools will enable experiments to understand the intricate mechanisms involved in neurological signaling and the roles played by neurotransmitters, such as dopamine and serotonin, in the activation of their respective receptors.
Collapse
Affiliation(s)
- Naeem Asad
- New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Davide Deodato
- New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Nadeem Asad
- New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Sangram Gore
- New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Timothy M Dore
- New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
14
|
La Barbera L, Spoleti E, D'Amelio M. Calcium handling: a strategy to fight neurodegeneration in Alzheimer's disease. Neural Regen Res 2023; 18:2685-2686. [PMID: 37449622 DOI: 10.4103/1673-5374.374004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Affiliation(s)
- Livia La Barbera
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio- Medico di Roma; Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Elena Spoleti
- Department of Sciences and Technologies for Humans and Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation; Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
15
|
Jeffs QL, Prather JF, Todd WD. Potential neural substrates underlying circadian and olfactory disruptions in preclinical Alzheimer's disease. Front Neurosci 2023; 17:1295998. [PMID: 38094003 PMCID: PMC10716239 DOI: 10.3389/fnins.2023.1295998] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/13/2023] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, with over 45 million patients worldwide, and poses significant economic and emotional burdens to both patients and caregivers, significantly raising the number of those affected. Unfortunately, much of the existing research on the disease only addresses a small subset of associated symptomologies and pathologies. In this review, we propose to target the earliest stages of the disease, when symptomology first arises. In these stages, before the onset of hallmark symptoms of AD such as cognitive impairments and memory loss, circadian and olfactory disruptions arise and are detectable. Functional similarities between circadian and olfactory systems provide a basis upon which to seek out common mechanisms in AD which may target them early on in the disease. Existing studies of interactions between these systems, while intriguing, leave open the question of the neural substrates underlying them. Potential substrates for such interactions are proposed in this review, such as indirect projections that may functionally connect the two systems and dopaminergic signaling. These substrates may have significant implications for mechanisms underlying disruptions to circadian and olfactory function in early stages of AD. In this review, we propose early detection of AD using a combination of circadian and olfactory deficits and subsequent early treatment of these deficits may provide profound benefits to both patients and caregivers. Additionally, we suggest that targeting research toward the intersection of these two systems in AD could uncover mechanisms underlying the broader set of symptoms and pathologies that currently elude researchers.
Collapse
Affiliation(s)
| | | | - William D. Todd
- Department of Zoology and Physiology, Program in Neuroscience, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
16
|
Motta C, Assogna M, Bonomi CG, Di Lorenzo F, Nuccetelli M, Mercuri NB, Koch G, Martorana A. Interplay between the catecholaminergic enzymatic axis and neurodegeneration/neuroinflammation processes in the Alzheimer's disease continuum. Eur J Neurol 2023; 30:839-848. [PMID: 36692274 DOI: 10.1111/ene.15691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE The locus coeruleus (LC) provides dopamine/noradrenaline (DA/NA) innervation throughout the brain and undergoes early degeneration in Alzheimer's disease (AD). We evaluated catecholaminergic enzyme levels in the cerebrospinal fluid (CSF) of a group of patients biologically defined as within the AD continuum (ADc) and explored their relationship with AD biomarkers and cytokine/growth factor levels to investigate their interplay with neurodegenerative and neuroinflammatory processes. METHODS The CSF concentration of DA transporter (DAT), tyrosine-hydroxylase (TH), DOPA-decarboxylase (DDC), and dopamine-β-hydroxylase (DβH), as well as cytokine/growth factor levels, were analyzed in 41 ADc patients stratified according to CSF beta-amyloid (Aβ)1-42 (A) and p-tau (T) in AD pathological changes (A+ T-) and AD (A+ T+) subgroups, as well as in 15 control subjects (A- T-). RESULTS The ADc group had lower CSF levels of DAT and TH but increased DβH levels to compensate for NA synthesis. DDC levels were higher in the A+ T+ subgroup but comparable with controls in the A+ T- subgroup, probably because the DA system is resilient to the degeneration of LC neurons in the absence of tau pathology. Adjusting for age, sex, APOE genotype, and cognitive status, a significant association was found between TH and Aβ1-42 (R2 = 0.25) and between DDC and p-tau (R2 = 0.33). Finally, TH correlated with interleukin (IL)-10 levels (p = 0.0008) and DβH with IL-1β (p = 0.03), IL-4 (p = 0.02), granulocyte colony-stimulating factor (p = 0.007), and IL-17 (p = 0.01). CONCLUSIONS Taken together, these findings suggest that catecholaminergic enzymes, functional markers of the catecholaminergic system, are closely linked to the neurodegenerative and neuroinflammatory processes in AD pathology.
Collapse
Affiliation(s)
- Caterina Motta
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Martina Assogna
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Chiara Giuseppina Bonomi
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Di Lorenzo
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giacomo Koch
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Alessandro Martorana
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
17
|
Cai W, Wakamatsu K, Zucca FA, Wang Q, Yang K, Mohamadzadehonarvar N, Srivastava P, Tanaka H, Holly G, Casella L, Ito S, Zecca L, Chen X. DOPA pheomelanin is increased in nigral neuromelanin of Parkinson's disease. Prog Neurobiol 2023; 223:102414. [PMID: 36746222 DOI: 10.1016/j.pneurobio.2023.102414] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Neuromelanin (NM) in dopaminergic neurons of human substantia nigra (SN) has a melanic component that consists of pheomelanin and eumelanin moieties and has been proposed as a key factor contributing to dopaminergic neuron vulnerability in Parkinson's disease (PD). While eumelanin is considered as an antioxidant, pheomelanin and related oxidative stress are associated with compromised drug and metal ion binding and melanoma risk. Using postmortem SN from patients with PD or Alzheimer's disease (AD) and unaffected controls, we identified increased L-3,4-dihydroxyphenylalanine (DOPA) pheomelanin and increased ratios of dopamine (DA) pheomelanin markers to DA in PD SN compared to controls. Eumelanins derived from both DOPA and DA were reduced in PD group. In addition, we report an increase in DOPA pheomelanin relative to DA pheomelanin in PD SN. In AD SN, we observed unaltered melanin markers despite reduced DOPA compared to controls. Furthermore, synthetic DOPA pheomelanin induced neuronal cell death in vitro while synthetic DOPA eumelanin showed no significant effect on cell viability. Our findings provide insights into the different roles of pheomelanin and eumelanin in PD pathophysiology. We anticipate our study will lead to further investigations on pheomelanin and eumelanin individually as biomarkers and possibly therapeutic targets for PD.
Collapse
Affiliation(s)
- Waijiao Cai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Institutes of Integrative Medicine, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Shanghai, China
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Qing Wang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Kai Yang
- Institutes of Integrative Medicine, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Shanghai, China
| | - Niyaz Mohamadzadehonarvar
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Pranay Srivastava
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Hitomi Tanaka
- Department of Medical Technology, School of Health Sciences, Gifu University of Medical Science, Seki, Japan
| | - Gabriel Holly
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA.
| |
Collapse
|
18
|
Nobili A, D'Amelio M, Viscomi MT. Nilotinib: from animal-based studies to clinical investigation in Alzheimer's disease patients. Neural Regen Res 2023; 18:803-804. [PMID: 36204843 PMCID: PMC9700081 DOI: 10.4103/1673-5374.350700] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Annalisa Nobili
- Department of Medicine and Surgery, University Campus Bio-Medico; Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Marcello D'Amelio
- Department of Medicine and Surgery, University Campus Bio-Medico; Department of Experimental Neuroscience, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Maria Teresa Viscomi
- Department of Life Science and Public Health Section of Histology and Embryology, Università Cattolica del Sacro Cuore; Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Rome, Italy
| |
Collapse
|
19
|
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: 25] [Impact Index Per Article: 12.5] [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
|
20
|
Puglisi-Allegra S, Lazzeri G, Busceti CL, Giorgi FS, Biagioni F, Fornai F. Lithium engages autophagy for neuroprotection and neuroplasticity: translational evidence for therapy. Neurosci Biobehav Rev 2023; 148:105148. [PMID: 36996994 DOI: 10.1016/j.neubiorev.2023.105148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
Here an overview is provided on therapeutic/neuroprotective effects of Lithium (Li+) in neurodegenerative and psychiatric disorders focusing on the conspicuous action of Li+ through autophagy. The effects on the autophagy machinery remain the key molecular mechanisms to explain the protective effects of Li+ for neurodegenerative diseases, offering potential therapeutic strategies for the treatment of neuropsychiatric disorders and emphasizes a crossroad linking autophagy, neurodegenerative disorders, and mood stabilization. Sensitization by psychostimulants points to several mechanisms involved in psychopathology, most also crucial in neurodegenerative disorders. Evidence shows the involvement of autophagy and metabotropic Glutamate receptors-5 (mGluR5) in neurodegeneration due to methamphetamine neurotoxicity as well as in neuroprotection, both in vitro and in vivo models. More recently, Li+ was shown to modulate autophagy through its action on mGluR5, thus pointing to an additional way of autophagy engagement by Li+ and to a substantial role of mGluR5 in neuroprotection related to neural e neuropsychiatry diseases. We propose Li+ engagement of autophagy through the canonical mechanisms of autophagy machinery and through the intermediary of mGluR5.
Collapse
|
21
|
Shaikh A, Ahmad F, Teoh SL, Kumar J, Yahaya MF. Honey and Alzheimer's Disease-Current Understanding and Future Prospects. Antioxidants (Basel) 2023; 12:427. [PMID: 36829985 PMCID: PMC9952506 DOI: 10.3390/antiox12020427] [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: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Alzheimer's disease (AD), a leading cause of dementia, has been a global concern. AD is associated with the involvement of the central nervous system that causes the characteristic impaired memory, cognitive deficits, and behavioral abnormalities. These abnormalities caused by AD is known to be attributed by extracellular aggregates of amyloid beta plaques and intracellular neurofibrillary tangles. Additionally, genetic factors such as abnormality in the expression of APOE, APP, BACE1, PSEN-1, and PSEN-2 play a role in the disease. As the current treatment aims to treat the symptoms and to slow the disease progression, there has been a continuous search for new nutraceutical agent or medicine to help prevent and cure AD pathology. In this quest, honey has emerged as a powerful nootropic agent. Numerous studies have demonstrated that the high flavonoids and phenolic acids content in honey exerts its antioxidant, anti-inflammatory, and neuroprotective properties. This review summarizes the effect of main flavonoid compounds found in honey on the physiological functioning of the central nervous system, and the effect of honey intake on memory and cognition in various animal model. This review provides a new insight on the potential of honey to prevent AD pathology, as well as to ameliorate the damage in the developed AD.
Collapse
Affiliation(s)
- Ammara Shaikh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Fairus Ahmad
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
22
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Primary prevention trials have shifted their focus to the earliest stages of Alzheimer's disease (AD). Autopsy data indicates that the neuromodulatory subcortical systems' (NSS) nuclei are specifically vulnerable to initial tau pathology, indicating that these nuclei hold great promise for early detection of AD in the context of the aging brain. The increasing availability of new imaging methods, ultra-high field scanners, new radioligands, and routine deep brain stimulation implants has led to a growing number of NSS neuroimaging studies on aging and neurodegeneration. Here, we review findings of current state-of-the-art imaging studies assessing the structure, function, and molecular changes of these nuclei during aging and AD. Furthermore, we identify the challenges associated with these imaging methods, important pathophysiologic gaps to fill for the AD NSS neuroimaging field, and provide future directions to improve our assessment, understanding, and clinical use of in vivo imaging of the NSS.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Elouise A Koops
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prokopis C Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tarun Singhal
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
23
|
Lansdell TA, Xu H, Galligan JJ, Dorrance AM. Effects of Striatal Amyloidosis on the Dopaminergic System and Behavior: A Comparative Study in Male and Female 5XFAD Mice. J Alzheimers Dis 2023; 94:1361-1375. [PMID: 37424461 DOI: 10.3233/jad-220905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
BACKGROUND Nearly two-thirds of patients diagnosed with Alzheimer's disease (AD) are female. In addition, female patients with AD have more significant cognitive impairment than males at the same disease stage. This disparity suggests there are sex differences in AD progression. While females appear to be more affected by AD, most published behavioral studies utilize male mice. In humans, there is an association between antecedent attention-deficit/hyperactivity disorder and increased risk of dementia. Functional connectivity studies indicate that dysfunctional cortico-striatal networks contribute to hyperactivity in attention deficit hyperactivity disorder. Higher plaque density in the striatum accurately predicts the presence of clinical AD pathology. In addition, there is a link between AD-related memory dysfunction and dysfunctional dopamine signaling. OBJECTIVE With the need to consider sex as a biological variable, we investigated the influence of sex on striatal plaque burden, dopaminergic signaling, and behavior in prodromal 5XFAD mice. METHODS Six-month-old male and female 5XFAD and C57BL/6J mice were evaluated for striatal amyloid plaque burden, locomotive behavior, and changes in dopaminergic machinery in the striatum. RESULTS 5XFAD female mice had a higher striatal amyloid plaque burden than male 5XFAD mice. 5XFAD females, but not males, were hyperactive. Hyperactivity in female 5XFAD mice was associated with increased striatal plaque burden and changes in dopamine signaling in the dorsal striatum. CONCLUSION Our results indicate that the progression of amyloidosis involves the striatum in females to a greater extent than in males. These studies have significant implications for using male-only cohorts in the study of AD progression.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Hui Xu
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
24
|
D’Amelio M, Viscomi M, Nobili A. Nilotinib: from animal-based studies to clinical investigation in Alzheimer’s disease patients. Neural Regen Res 2023. [DOI: 10.4103/5374.350700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
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
|
26
|
Accelerated atrophy in dopaminergic targets and medial temporo-parietal regions precedes the onset of delusions in patients with Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2023; 273:229-241. [PMID: 35554669 PMCID: PMC9958148 DOI: 10.1007/s00406-022-01417-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 04/11/2022] [Indexed: 12/21/2022]
Abstract
People with Alzheimer's disease (AD) and delusions have worse quality of life and prognosis. However, early markers of delusions have not been identified yet. The present study investigated whether there are any detectable differences in grey matter (GM) volume and cognitive changes in the year before symptom onset between patients with AD who did and did not develop delusions. Two matched samples of AD patients, 63 who did (PT-D) and 63 who did not develop delusions (PT-ND) over 1 year, were identified from the Alzheimer's Disease Neuroimaging Initiative database. The Neuropsychiatric Inventory (NPI) was used to assess the presence of delusions. Sixty-three additional matched healthy controls (HC) were selected. Repeated-measures ANCOVA models were used to investigate group-by-time effects on the volume of selected GM regions of interest and on cognitive performance. No neurocognitive differences were observed between patient groups prior to symptom onset. Greater episodic memory decline and GM loss in bilateral caudate nuclei, medio-temporal and midline cingulo-parietal regions were found in the PT-D compared with the PT-ND group. A pattern of faster GM loss in brain areas typically affected by AD and in cortical and subcortical targets of dopaminergic pathways, paralleled by worsening of episodic memory and behavioural symptoms, may explain the emergence of delusions in patients with AD.
Collapse
|
27
|
Broadbelt T, Mutlu-Smith M, Carnicero-Senabre D, Saido TC, Saito T, Wang SH. Impairment in novelty-promoted memory via behavioral tagging and capture before apparent memory loss in a knock-in model of Alzheimer's disease. Sci Rep 2022; 12:22298. [PMID: 36566248 PMCID: PMC9789965 DOI: 10.1038/s41598-022-26113-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is associated with cognitive impairments and age-dependent memory deficits which have been studied using genetic models of AD. Whether the processes for modulating memory persistence are more vulnerable to the influence of amyloid pathology than the encoding and consolidation of the memory remains unclear. Here, we investigated whether early amyloid pathology would affect peri-learning novelty in promoting memory, through a process called behavioral tagging and capture (BTC). AppNL-G-F/NL-G-F mice and wild-type littermates were trained in an appetitive delayed matching-to-place (ADMP) task which allows for the assessment of peri-learning novelty in facilitating memory. The results show that novelty enabled intermediate-term memory in wild-type mice, but not in AppNL-G-F/NL-G-F mice in adulthood. This effect preceded spatial memory impairment in the ADMP task seen in middle age. Other memory tests in the Barnes maze, Y-maze, novel object or location recognition tasks remained intact. Together, memory modulation through BTC is impaired before apparent deficits in learning and memory. Relevant biological mechanisms underlying BTC and the implication in AD are discussed.
Collapse
Affiliation(s)
- Tabitha Broadbelt
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Menekse Mutlu-Smith
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Daniel Carnicero-Senabre
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,grid.5515.40000000119578126Present Address: Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
| | - Takaomi C. Saido
- grid.474690.8Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, 351-0198 Japan
| | - Takashi Saito
- grid.260433.00000 0001 0728 1069Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, 467-8601 Japan
| | - Szu-Han Wang
- grid.4305.20000 0004 1936 7988Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| |
Collapse
|
28
|
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
|
29
|
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: 23] [Impact Index Per Article: 7.7] [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
|
30
|
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: 4.7] [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
|
31
|
Guzmán-Ramos K, Osorio-Gómez D, Bermúdez-Rattoni F. Cognitive impairment in alzheimer’s and metabolic diseases: A catecholaminergic hypothesis. Neuroscience 2022; 497:308-323. [DOI: 10.1016/j.neuroscience.2022.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022]
|
32
|
Cerebrospinal fluid catecholamines in Alzheimer's disease patients with and without biological disease. Transl Psychiatry 2022; 12:151. [PMID: 35397615 PMCID: PMC8994756 DOI: 10.1038/s41398-022-01901-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/08/2022] Open
Abstract
Noradrenergic and dopaminergic neurons are involved in cognitive functions, relate to behavioral and psychological symptoms in dementia and are affected in Alzheimer's disease (AD). Amyloid plaques (A), neurofibrillary tangles (T) and neurodegeneration (N) hallmarks the AD neuropathology. Today, the AT(N) pathophysiology can be assessed through biomarkers. Previous studies report cerebrospinal fluid (CSF) catecholamine concentrations in AD patients without biomarker refinement. We explored if CSF catecholamines relate to AD clinical presentation or neuropathology as reflected by CSF biomarkers. CSF catecholamines were analyzed in AD patients at the mild cognitive impairment (MCI; n = 54) or dementia stage (n = 240) and in cognitively unimpaired (n = 113). CSF biomarkers determined AT status and indicated synaptic damage (neurogranin). The AD patients (n = 294) had higher CSF noradrenaline and adrenaline concentrations, but lower dopamine concentrations compared to the cognitively unimpaired (n = 113). AD patients in the MCI and dementia stage of the disease had similar CSF catecholamine concentrations. In the CSF neurogranin positively associated with noradrenaline and adrenaline but not with dopamine. Adjusted regression analyses including AT status, CSF neurogranin, age, gender, and APOEε4 status verified the findings. In restricted analyses comparing A+T+ patients to A-T- cognitively unimpaired, the findings for CSF adrenaline remained significant (p < 0.001) but not for CSF noradrenaline (p = 0.07) and CSF dopamine (p = 0.33). There were no differences between A+T+ and A-T- cognitively unimpaired. Thus, we find alterations in CSF catecholamines in symptomatic AD and the CSF adrenergic transmitters to increase simultaneously with synaptic damage as indexed by CSF neurogranin.
Collapse
|
33
|
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: 3.0] [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
|
34
|
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: 16] [Impact Index Per Article: 4.0] [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
|
35
|
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: 6.3] [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
|
36
|
Fernandes C, Macedo I, Barbosa F, Marques-Teixeira J. Economic decision-making in the continuum between healthy aging and Alzheimer's Disease: A systematic review of 20 years of research. Neurosci Biobehav Rev 2021; 131:1243-1263. [PMID: 34715151 DOI: 10.1016/j.neubiorev.2021.10.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/23/2021] [Accepted: 10/23/2021] [Indexed: 01/21/2023]
Abstract
The effect of pathological aging on economic decision-making is a topic of major relevance as impairments in this domain place older adults at increased risk for financial abuse. This review aims to characterize decision-making across the continuum that goes from healthy aging to Alzheimer's Dementia. We included 42 studies comparing patients with Mild Cognitive Impairment (MCI) and healthy older adults, patients with Alzheimer's Disease (AD) and healthy older adults, and patients with MCI and patients with AD. Substantial evidence emerged suggesting that both MCI as AD affect economic decision-making. However, a non-negligible number of behavioural tasks failed to find significant differences between patients and controls, and no differences were reported between patients with MCI and AD. On the contrary, measures of financial capacity reached more robust findings, showing that healthy older adults had better performance than patients, while MCI patients showed better performance than AD patients. This review presents the main conclusions that may be drawn from significant findings, as well as the hypotheses and recommendations for future research.
Collapse
Affiliation(s)
- Carina Fernandes
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal.
| | - Inês Macedo
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| | - Fernando Barbosa
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| | - João Marques-Teixeira
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Portugal
| |
Collapse
|
37
|
Di Lazzaro V, Bella R, Benussi A, Bologna M, Borroni B, Capone F, Chen KHS, Chen R, Chistyakov AV, Classen J, Kiernan MC, Koch G, Lanza G, Lefaucheur JP, Matsumoto H, Nguyen JP, Orth M, Pascual-Leone A, Rektorova I, Simko P, Taylor JP, Tremblay S, Ugawa Y, Dubbioso R, Ranieri F. Diagnostic contribution and therapeutic perspectives of transcranial magnetic stimulation in dementia. Clin Neurophysiol 2021; 132:2568-2607. [PMID: 34482205 DOI: 10.1016/j.clinph.2021.05.035] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 04/22/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023]
Abstract
Transcranial magnetic stimulation (TMS) is a powerful tool to probe in vivo brain circuits, as it allows to assess several cortical properties such asexcitability, plasticity and connectivity in humans. In the last 20 years, TMS has been applied to patients with dementia, enabling the identification of potential markers of thepathophysiology and predictors of cognitive decline; moreover, applied repetitively, TMS holds promise as a potential therapeutic intervention. The objective of this paper is to present a comprehensive review of studies that have employed TMS in dementia and to discuss potential clinical applications, from the diagnosis to the treatment. To provide a technical and theoretical framework, we first present an overview of the basic physiological mechanisms of the application of TMS to assess cortical excitability, excitation and inhibition balance, mechanisms of plasticity and cortico-cortical connectivity in the human brain. We then review the insights gained by TMS techniques into the pathophysiology and predictors of progression and response to treatment in dementias, including Alzheimer's disease (AD)-related dementias and secondary dementias. We show that while a single TMS measure offers low specificity, the use of a panel of measures and/or neurophysiological index can support the clinical diagnosis and predict progression. In the last part of the article, we discuss the therapeutic uses of TMS. So far, only repetitive TMS (rTMS) over the left dorsolateral prefrontal cortex and multisite rTMS associated with cognitive training have been shown to be, respectively, possibly (Level C of evidence) and probably (Level B of evidence) effective to improve cognition, apathy, memory, and language in AD patients, especially at a mild/early stage of the disease. The clinical use of this type of treatment warrants the combination of brain imaging techniques and/or electrophysiological tools to elucidate neurobiological effects of neurostimulation and to optimally tailor rTMS treatment protocols in individual patients or specific patient subgroups with dementia or mild cognitive impairment.
Collapse
Affiliation(s)
- Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.
| | - Rita Bella
- Department of Medical and Surgical Sciences and Advanced Technologies, Section of Neurosciences, University of Catania, Catania, Italy
| | - Alberto Benussi
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Matteo Bologna
- Department of Human Neurosciences, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Kai-Hsiang S Chen
- Department of Neurology, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Robert Chen
- Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada; Division of Brain, Imaging& Behaviour, Krembil Brain Institute, Toronto, Canada
| | | | - Joseph Classen
- Department of Neurology, University Hospital Leipzig, Leipzig University Medical Center, Germany
| | - Matthew C Kiernan
- Department of Neurology, Royal Prince Alfred Hospital, Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy; Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Giuseppe Lanza
- Department of Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy; Department of Neurology IC, Oasi Research Institute-IRCCS, Troina, Italy
| | - Jean-Pascal Lefaucheur
- ENT Team, EA4391, Faculty of Medicine, Paris Est Créteil University, Créteil, France; Clinical Neurophysiology Unit, Department of Physiology, Henri Mondor Hospital, Assistance Publique - Hôpitaux de Paris, Créteil, France
| | | | - Jean-Paul Nguyen
- Pain Center, clinique Bretéché, groupe ELSAN, Multidisciplinary Pain, Palliative and Supportive care Center, UIC 22/CAT2 and Laboratoire de Thérapeutique (EA3826), University Hospital, Nantes, France
| | - Michael Orth
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland; Swiss Huntington's Disease Centre, Siloah, Bern, Switzerland
| | - Alvaro Pascual-Leone
- Hinda and Arthur Marcus Institute for Aging Research, Center for Memory Health, Hebrew SeniorLife, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; Guttmann Brain Health Institute, Universitat Autonoma Barcelona, Spain
| | - Irena Rektorova
- Applied Neuroscience Research Group, Central European Institute of Technology, Masaryk University (CEITEC MU), Brno, Czech Republic; Department of Neurology, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Patrik Simko
- Applied Neuroscience Research Group, Central European Institute of Technology, Masaryk University (CEITEC MU), Brno, Czech Republic; Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sara Tremblay
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, ON, Canada; Royal Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Yoshikazu Ugawa
- Department of Human Neurophysiology, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Raffaele Dubbioso
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Federico Ranieri
- Unit of Neurology, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
38
|
Babić Leko M, Nikolac Perković M, Nedić Erjavec G, Klepac N, Švob Štrac DK, Borovečki F, Pivac N, Hof PR, Šimić G. Association of the MAOB rs1799836 Single Nucleotide Polymorphism and APOE ɛ4 Allele in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:585-594. [PMID: 34533445 DOI: 10.2174/1567205018666210917162843] [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: 05/20/2020] [Revised: 07/01/2021] [Accepted: 08/22/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dopaminergic system is functionally compromised in Alzheimer's dis-ease (AD). The activity of monoamine oxidase B (MAOB), the enzyme involved in the degradation of dopamine, is increased during AD. Also, increased expression of MAOB occurs in the post-mortem hippocampus and neocortex of patients with AD. The MAOB rs1799836 polymorphism modulates MAOB transcription, consequently influencing protein translation and MAOB activity. We recently showed that cerebrospinal fluid levels of amyloid β1-42 are decreased in patients carry- ing the A allele in MAOB rs1799836 polymorphism. OBJECTIVE The present study compares MAOB rs1799836 polymorphism and APOE, the only con- firmed genetic risk factor for sporadic AD. METHOD We included 253 participants, 127 of whom had AD, 57 had mild cognitive impairment, 11 were healthy controls, and 58 suffered from other primary causes of dementia. MAOB and APOE polymorphisms were determined using TaqMan SNP Genotyping Assays. RESULTS We observed that the frequency of APOE ɛ4/ɛ4 homozygotes and APOE ɛ4 carriers is sig- nificantly increased among patients carrying the AA MAOB rs1799836 genotype. CONCLUSION These results indicate that the MAOB rs1799836 polymorphism is a potential genetic biomarker of AD and a potential target for the treatment of decreased dopaminergic transmission and cognitive deterioration in AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
39
|
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: 3.3] [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
|
40
|
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: 3.0] [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
|
41
|
Plini ERG, O’Hanlon E, Boyle R, Sibilia F, Rikhye G, Kenney J, Whelan R, Melnychuk MC, Robertson IH, Dockree PM. Examining the Role of the Noradrenergic Locus Coeruleus for Predicting Attention and Brain Maintenance in Healthy Old Age and Disease: An MRI Structural Study for the Alzheimer's Disease Neuroimaging Initiative. Cells 2021; 10:1829. [PMID: 34359997 PMCID: PMC8306442 DOI: 10.3390/cells10071829] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
The noradrenergic theory of Cognitive Reserve (Robertson, 2013-2014) postulates that the upregulation of the locus coeruleus-noradrenergic system (LC-NA) originating in the brainstem might facilitate cortical networks involved in attention, and protracted activation of this system throughout the lifespan may enhance cognitive stimulation contributing to reserve. To test the above-mentioned theory, a study was conducted on a sample of 686 participants (395 controls, 156 mild cognitive impairment, 135 Alzheimer's disease) investigating the relationship between LC volume, attentional performance and a biological index of brain maintenance (BrainPAD-an objective measure, which compares an individual's structural brain health, reflected by their voxel-wise grey matter density, to the state typically expected at that individual's age). Further analyses were carried out on reserve indices including education and occupational attainment. Volumetric variation across groups was also explored along with gender differences. Control analyses on the serotoninergic (5-HT), dopaminergic (DA) and cholinergic (Ach) systems were contrasted with the noradrenergic (NA) hypothesis. The antithetic relationships were also tested across the neuromodulatory subcortical systems. Results supported by Bayesian modelling showed that LC volume disproportionately predicted higher attentional performance as well as biological brain maintenance across the three groups. These findings lend support to the role of the noradrenergic system as a key mediator underpinning the neuropsychology of reserve, and they suggest that early prevention strategies focused on the noradrenergic system (e.g., cognitive-attentive training, physical exercise, pharmacological and dietary interventions) may yield important clinical benefits to mitigate cognitive impairment with age and disease.
Collapse
Affiliation(s)
- Emanuele R. G. Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Erik O’Hanlon
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychiatry, Royal College of Surgeons in Ireland, Hospital Rd, Beaumont, 9QRH+4F Dublin, Ireland
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Rory Boyle
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Francesca Sibilia
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Gaia Rikhye
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Joanne Kenney
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Robert Whelan
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Michael C. Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Ian H. Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Paul M. Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| |
Collapse
|
42
|
Gallo A, Pillet LE, Verpillot R. New frontiers in Alzheimer's disease diagnostic: Monoamines and their derivatives in biological fluids. Exp Gerontol 2021; 152:111452. [PMID: 34182050 DOI: 10.1016/j.exger.2021.111452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Current diagnosis of Alzheimer's disease (AD) relies on a combination of neuropsychological evaluations, biomarker measurements and brain imaging. Nevertheless, these approaches are either expensive, invasive or lack sensitivity to early AD stages. The main challenge of ongoing research is therefore to identify early non-invasive biomarkers to diagnose AD at preclinical stage. Accumulating evidence support the hypothesis that initial degeneration of profound monoaminergic nuclei may trigger a transneuronal spread of AD pathology towards hippocampus and cortex. These studies aroused great interest on monoamines, i.e. noradrenaline (NA), dopamine (D) ad serotonin (5-HT), as early hallmarks of AD pathology. The present work reviews current literature on the potential role of monoamines and related metabolites as biomarkers of AD. First, morphological changes in the monoaminergic systems during AD are briefly described. Second, we focus on concentration changes of these molecules and their derivatives in biological fluids, including cerebrospinal fluid, obtained by lumbar puncture, and blood or urine, sampled via less invasive procedures. Starting from initial observations, we then discuss recent insights on metabolomics-based analysis, highlighting the promising clinical utility of monoamines for the identification of a molecular AD signature, aimed at improving early diagnosis and discrimination from other dementia.
Collapse
|
43
|
Chen APF, Chen L, Kim TA, Xiong Q. Integrating the Roles of Midbrain Dopamine Circuits in Behavior and Neuropsychiatric Disease. Biomedicines 2021; 9:biomedicines9060647. [PMID: 34200134 PMCID: PMC8228225 DOI: 10.3390/biomedicines9060647] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 01/11/2023] Open
Abstract
Dopamine (DA) is a behaviorally and clinically diverse neuromodulator that controls CNS function. DA plays major roles in many behaviors including locomotion, learning, habit formation, perception, and memory processing. Reflecting this, DA dysregulation produces a wide variety of cognitive symptoms seen in neuropsychiatric diseases such as Parkinson’s, Schizophrenia, addiction, and Alzheimer’s disease. Here, we review recent advances in the DA systems neuroscience field and explore the advancing hypothesis that DA’s behavioral function is linked to disease deficits in a neural circuit-dependent manner. We survey different brain areas including the basal ganglia’s dorsomedial/dorsolateral striatum, the ventral striatum, the auditory striatum, and the hippocampus in rodent models. Each of these regions have different reported functions and, correspondingly, DA’s reflecting role in each of these regions also has support for being different. We then focus on DA dysregulation states in Parkinson’s disease, addiction, and Alzheimer’s Disease, emphasizing how these afflictions are linked to different DA pathways. We draw upon ideas such as selective vulnerability and region-dependent physiology. These bodies of work suggest that different channels of DA may be dysregulated in different sets of disease. While these are great advances, the fine and definitive segregation of such pathways in behavior and disease remains to be seen. Future studies will be required to define DA’s necessity and contribution to the functional plasticity of different striatal regions.
Collapse
Affiliation(s)
- Allen PF Chen
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Lu Chen
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
| | - Thomas A. Kim
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Medical Scientist Training Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology and Behavior, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA; (A.P.C.); (L.C.); (T.A.K.)
- Correspondence:
| |
Collapse
|
44
|
Iaccarino L, Sala A, Caminiti SP, Presotto L, Perani D. In vivo MRI Structural and PET Metabolic Connectivity Study of Dopamine Pathways in Alzheimer's Disease. J Alzheimers Dis 2021; 75:1003-1016. [PMID: 32390614 DOI: 10.3233/jad-190954] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by an involvement of brain dopamine (DA) circuitry, the presence of which has been associated with emergence of both neuropsychiatric symptoms and cognitive deficits. OBJECTIVE In order to investigate whether and how the DA pathways are involved in the pathophysiology of AD, we assessed by in vivo neuroimaging the structural and metabolic alterations of subcortical and cortical DA pathways and targets. METHODS We included 54 healthy control participants, 53 amyloid-positive subjects with mild cognitive impairment due to AD (MCI-AD), and 60 amyloid-positive patients with probable dementia due to AD (ADD), all with structural 3T MRI and 18F-FDG-PET scans. We assessed MRI-based gray matter reductions in the MCI-AD and ADD groups within an anatomical a priori-defined Nigrostriatal and Mesocorticolimbic DA pathways, followed by 18F-FDG-PET metabolic connectivity analyses to evaluate network-level metabolic connectivity changes. RESULTS We found significant tissue loss in the Mesocorticolimbic over the Nigrostriatal pathway. Atrophy was evident in the ventral striatum, orbitofrontal cortex, and medial temporal lobe structures, and already plateaued in the MCI-AD stage. Degree of atrophy in Mesocorticolimbic regions positively correlated with the severity of depression, anxiety, and apathy in MCI-AD and ADD subgroups. Additionally, we observed significant alterations of metabolic connectivity between the ventral striatum and fronto-cingulate regions in ADD, but not in MCI-AD. There were no metabolic connectivity changes within the Nigrostriatal pathway. CONCLUSION Our cross-sectional data support a clinically-meaningful, yet stage-dependent, involvement of the Mesocorticolimbic system in AD. Longitudinal and clinical correlation studies are needed to further establish the relevance of DA system involvement in AD.
Collapse
Affiliation(s)
- Leonardo Iaccarino
- Vita-Salute San Raffaele University, Milan, Italy.,In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Arianna Sala
- Vita-Salute San Raffaele University, Milan, Italy.,In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, Milan, Italy.,In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Presotto
- In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy.,In vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| | | |
Collapse
|
45
|
Carandini T, Cercignani M, Galimberti D, Scarpini E, Bozzali M. The distinct roles of monoamines in multiple sclerosis: A bridge between the immune and nervous systems? Brain Behav Immun 2021; 94:381-391. [PMID: 33662501 DOI: 10.1016/j.bbi.2021.02.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/20/2022] Open
Abstract
The monoaminergic neurotransmitters dopamine, noradrenaline, and serotonin are pivotal actors of the interplay between the nervous and the immune system due to their ability of binding to cell-receptors of both systems, crucially regulating their function within the central nervous system and the periphery. As monoamines are dysfunctional in many neurological and psychiatric diseases, they have been successfully used as pharmacological targets. Multiple sclerosis (MS) is one of the best examples of neurological disease caused by an altered interaction between the nervous and immune system and emerging evidence supports a dysregulation of monoaminergic systems in the pathogenesis of MS, secondary to both inflammation-induced reduction of monoamines' synthesis and structural damage to monoaminergic pathways within the brain. Here we review the evidence for monoamines being key mediators of neuroimmune interaction, affecting MS pathogenesis and course. Moreover, we discuss how the reduction/dysfunction of monoamines in MS may contribute to some clinical features typical of the disease, particularly fatigue and depression. Finally, we summarize different drugs targeting monoamines that are currently under evaluation for their potential efficacy to treat MS, as well as to alleviate fatigue and depression in MS.
Collapse
Affiliation(s)
- Tiziana Carandini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Mara Cercignani
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, UK; Neuroimaging Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; University of Milan, Dino Ferrari Center, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; University of Milan, Dino Ferrari Center, Milan, Italy
| | - Marco Bozzali
- Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, UK; Rita Levi Montalcini Department of Neuroscience, University of Torino, Turin, Italy
| |
Collapse
|
46
|
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: 54] [Impact Index Per Article: 13.5] [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
|
47
|
Autophagy status as a gateway for stress-induced catecholamine interplay in neurodegeneration. Neurosci Biobehav Rev 2021; 123:238-256. [PMID: 33497785 DOI: 10.1016/j.neubiorev.2021.01.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
The catecholamine-containing brainstem nuclei locus coeruleus (LC) and ventral tegmental area (VTA) are critically involved in stress responses. Alterations of catecholamine systems during chronic stress may contribute to neurodegeneration, including cognitive decline. Stress-related catecholamine alterations, while contributing to anxiety and depression, might accelerate neuronal degeneration by increasing the formation of toxic dopamine and norepinephrine by-products. These, in turn, may impair proteostasis within a variety of cortical and subcortical areas. In particular, the molecular events governing neurotransmission, neuroplasticity, and proteostasis within LC and VTA affect a variety of brain areas. Therefore, we focus on alterations of autophagy machinery in these nuclei as a relevant trigger in this chain of events. In fact, these catecholamine-containing areas are mostly prone to autophagy-dependent neurodegeneration. Thus, we propose a dynamic hypothesis according to which stress-induced autophagy alterations within the LC-VTA network foster a cascade towards early neurodegeneration within these nuclei.
Collapse
|
48
|
Reduced monoaminergic nuclei MRI signal detectable in pre-symptomatic older adults with future memory decline. Sci Rep 2020; 10:18707. [PMID: 33127923 PMCID: PMC7603335 DOI: 10.1038/s41598-020-71368-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
Evidence from murine models and human post-mortem studies indicates that monoaminergic nuclei undergo degeneration at the pre-symptomatic stage of Alzheimer’s disease (AD). Analysing 129 datasets from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) and relying on the Clinical Dementia Rating as group-defining instrument, we hypothesised that the MRI signal of monoaminergic nuclei would be a statistically significant predictor of memory decline in participants initially recruited in ADNI as healthy adults. As opposed to a group of cognitively stable participants, participants developing memory decline had reduced signal in the ventral tegmental area at baseline, before any evidence of functional decline emerged. These findings indicate that monoaminergic degeneration predates the onset of memory decline in an AD-centred initiative, with a crucial involvement of very-early changes of a dopaminergic region. This translates into potential informative avenues for pharmacological treatment of pre-symptomatic AD.
Collapse
|
49
|
D'Antonio F, De Bartolo MI, Ferrazzano G, Trebbastoni A, Amicarelli S, Campanelli A, de Lena C, Berardelli A, Conte A. Somatosensory Temporal Discrimination Threshold in Patients with Cognitive Disorders. J Alzheimers Dis 2020; 70:425-432. [PMID: 31177234 DOI: 10.3233/jad-190385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The temporal processing of sensory information can be evaluated by testing the somatosensory temporal discrimination threshold (STDT), which is defined as the shortest interstimulus interval needed to recognize two sequential sensory stimuli as separate in time. The STDT requires the functional integrity of the basal ganglia and of the somatosensory cortex (S1). Although there is evidence that time processing is impaired in patients with Alzheimer's disease (AD), no study has yet investigated STDT in patients with various degree of cognitive impairment. OBJECTIVE The aim of our study was to understand how cognition and attention deficits affect STDT values in patients with cognitive abnormalities. METHODS We enrolled 63 patients: 28 had mild-moderate AD, 16 had mild cognitive impairment (MCI), and the remaining 19 had subjective cognitive deficit (SCD). A group of 45 age-matched healthy subjects acted as controls. Paired tactile stimuli for STDT testing consisted of square-wave electrical pulses delivered with a constant current stimulator through surface electrodes over the distal phalanx of the index finger. RESULTS STDT values were higher in AD and MCI patients than in SCD subjects or healthy controls. Changes in the STDT in AD and MCI were similar in both conditions and did not correlate with disease severity. CONCLUSIONS STDT alterations in AD and MCI may reflect a dysfunction of the dopaminergic system, which signals salient events and includes the striatum and the mesocortical and mesolimbic circuits.
Collapse
Affiliation(s)
- Fabrizia D'Antonio
- Department of Human Neuroscience, Sapienza University of Rome, Rome Italy.,PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Sara Amicarelli
- Department of Human Neuroscience, Sapienza University of Rome, Rome Italy
| | | | - Carlo de Lena
- Department of Human Neuroscience, Sapienza University of Rome, Rome Italy
| | - Alfredo Berardelli
- Department of Human Neuroscience, Sapienza University of Rome, Rome Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | - Antonella Conte
- Department of Human Neuroscience, Sapienza University of Rome, Rome Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| |
Collapse
|
50
|
Todd WD. Potential Pathways for Circadian Dysfunction and Sundowning-Related Behavioral Aggression in Alzheimer's Disease and Related Dementias. Front Neurosci 2020; 14:910. [PMID: 33013301 PMCID: PMC7494756 DOI: 10.3389/fnins.2020.00910] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
Patients with Alzheimer's disease (AD) and related dementias are commonly reported to exhibit aggressive behavior and other emotional behavioral disturbances, which create a tremendous caretaker burden. There has been an abundance of work highlighting the importance of circadian function on mood and emotional behavioral regulation, and recent evidence demonstrates that a specific hypothalamic pathway links the circadian system to neurons that modulate aggressive behavior, regulating the propensity for aggression across the day. Such shared circuitry may have important ramifications for clarifying the complex interactions underlying "sundowning syndrome," a poorly understood (and even controversial) clinical phenomenon in AD and dementia patients that is characterized by agitation, aggression, and delirium during the late afternoon and early evening hours. The goal of this review is to highlight the potential output and input pathways of the circadian system that may underlie circadian dysfunction and behavioral aggression associated with sundowning syndrome, and to discuss possible ways these pathways might inform specific interventions for treatment. Moreover, the apparent bidirectional relationship between chronic disruptions of circadian and sleep-wake regulation and the pathology and symptoms of AD suggest that understanding the role of these circuits in such neurobehavioral pathologies could lead to better diagnostic or even preventive measures.
Collapse
Affiliation(s)
- William D Todd
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming, Laramie, WY, United States
| |
Collapse
|