101
|
Pentz R, Iulita MF, Ducatenzeiler A, Videla L, Benejam B, Carmona‐Iragui M, Blesa R, Lleó A, Fortea J, Cuello AC. Nerve growth factor (NGF) pathway biomarkers in Down syndrome prior to and after the onset of clinical Alzheimer's disease: A paired CSF and plasma study. Alzheimers Dement 2021; 17:605-617. [PMID: 33226181 PMCID: PMC8043977 DOI: 10.1002/alz.12229] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/28/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The discovery that nerve growth factor (NGF) metabolism is altered in Down syndrome (DS) and Alzheimer's disease (AD) brains offered a framework for the identification of novel biomarkers signalling NGF deregulation in AD pathology. METHODS We examined levels of NGF pathway proteins (proNGF, neuroserpin, tissue plasminogen activator [tPA], and metalloproteases [MMP]) in matched cerebrospinal fluid (CSF)/plasma samples from AD-symptomatic (DSAD) and AD-asymptomatic (aDS) individuals with DS, as well as controls (HC). RESULTS ProNGF and MMP-3 were elevated while tPA was decreased in plasma from individuals with DS. CSF from individuals with DS showed elevated proNGF, neuroserpin, MMP-3, and MMP-9. ProNGF and MMP-9 in CSF differentiated DSAD from aDS (area under the curve = 0.86, 0.87). NGF pathway markers associated with CSF amyloid beta and tau and differed by sex. DISCUSSION Brain NGF metabolism changes can be monitored in plasma and CSF, supporting relevance in AD pathology. These markers could assist staging, subtyping, or precision medicine for AD in DS.
Collapse
Affiliation(s)
- Rowan Pentz
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
| | - M. Florencia Iulita
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealCanada
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
| | | | - Laura Videla
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
- Barcelona Down Medical CenterFundación Catalana Síndrome de DownBarcelonaSpain
| | - Bessy Benejam
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
- Barcelona Down Medical CenterFundación Catalana Síndrome de DownBarcelonaSpain
| | - María Carmona‐Iragui
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
- Barcelona Down Medical CenterFundación Catalana Síndrome de DownBarcelonaSpain
| | - Rafael Blesa
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Alberto Lleó
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Juan Fortea
- Sant Pau Memory UnitDepartment of NeurologyHospital de la Santa Creu i Sant PauBiomedical Research Institute Sant PauUniversitat Autònoma de BarcelonaBarcelonaSpain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED)MadridSpain
- Barcelona Down Medical CenterFundación Catalana Síndrome de DownBarcelonaSpain
| | - A. Claudio Cuello
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
- Department of Pharmacology and TherapeuticsMcGill UniversityMontrealCanada
- Department of Anatomy and Cell BiologyMcGill UniversityMontrealCanada
- Department of PharmacologyOxford UniversityOxfordUK
| |
Collapse
|
102
|
The effect of nerve growth factor on supporting spatial memory depends upon hippocampal cholinergic innervation. Transl Psychiatry 2021; 11:162. [PMID: 33723225 PMCID: PMC7961060 DOI: 10.1038/s41398-021-01280-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/17/2020] [Accepted: 01/07/2021] [Indexed: 12/17/2022] Open
Abstract
Nerve growth factor (NGF) gene therapy has been used in clinical trials of Alzheimer's disease. Understanding the underlying mechanisms of how NGF influences memory may help develop new strategies for treatment. Both NGF and the cholinergic system play important roles in learning and memory. NGF is essential for maintaining cholinergic innervation of the hippocampus, but it is unclear whether the supportive effect of NGF on learning and memory is specifically dependent upon intact hippocampal cholinergic innervation. Here we characterize the behavior and hippocampal measurements of volume, neurogenesis, long-term potentiation, and cholinergic innervation, in brain-specific Ngf-deficient mice. Our results show that knockout mice exhibit increased anxiety, impaired spatial learning and memory, decreased adult hippocampal volume, neurogenesis, short-term potentiation, and cholinergic innervation. Overexpression of Ngf in the hippocampus of Ngf gene knockout mice rescued spatial memory and partially restored cholinergic innervations, but not anxiety. Selective depletion of hippocampal cholinergic innervation resulted in impaired spatial memory. However, Ngf overexpression in the hippocampus failed to rescue spatial memory in mice with hippocampal-selective cholinergic fiber depletion. In conclusion, we demonstrate the impact of Ngf deficiency in the brain and provide evidence that the effect of NGF on spatial memory is reliant on intact cholinergic innervations in the hippocampus. These results suggest that adequate cholinergic targeting may be a critical requirement for successful use of NGF gene therapy of Alzheimer's disease.
Collapse
|
103
|
Sarter M, Avila C, Kucinski A, Donovan E. Make a Left Turn: Cortico-Striatal Circuitry Mediating the Attentional Control of Complex Movements. Mov Disord 2021; 36:535-546. [PMID: 33615556 DOI: 10.1002/mds.28532] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND In movement disorders such as Parkinson's disease (PD), cholinergic signaling is disrupted by the loss of basal forebrain cholinergic neurons, as well as aberrant activity in striatal cholinergic interneurons (ChIs). Several lines of evidence suggest that gait imbalance, a key disabling symptom of PD, may be driven by alterations in high-level frontal cortical and cortico-striatal processing more typically associated with cognitive dysfunction. METHODS Here we describe the corticostriatal circuitry that mediates the cognitive-motor interactions underlying such complex movement control. The ability to navigate dynamic, obstacle-rich environments requires the continuous integration of information about the environment with movement selection and sequencing. The cortical-attentional processing of extero- and interoceptive cues requires modulation by cholinergic activity to guide striatal movement control. Cue-derived information is "transferred" to striatal circuitry primarily via fronto-striatal glutamatergic projections. RESULT Evidence from parkinsonian fallers and from a rodent model reproducing the dual cholinergic-dopaminergic losses observed in these patients supports the main hypotheses derived from this neuronal circuitry-guided conceptualization of parkinsonian falls. Furthermore, in the striatum, ChIs constitute a particularly critical node for the integration of cortical with midbrain dopaminergic afferents and thus for cues to control movements. CONCLUSION Procholinergic treatments that enhance or rescue cortical and striatal mechanisms may improve complex movement control in parkinsonian fallers and perhaps also in older persons suffering from gait disorders and a propensity for falls. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Martin Sarter
- Department of Psychology & Neuroscience Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Cassandra Avila
- Department of Psychology & Neuroscience Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Aaron Kucinski
- Department of Psychology & Neuroscience Program, University of Michigan, Ann Arbor, Michigan, USA
| | - Eryn Donovan
- Department of Psychology & Neuroscience Program, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
104
|
Abstract
This article presents an overview of imaging agents for PET that have been applied for research and diagnostic purposes in patients affected by dementia. Classified by the target which the agents visualize, seven groups of tracers can be distinguished, namely radiopharmaceuticals for: (1) Misfolded proteins (ß-amyloid, tau, α-synuclein), (2) Neuroinflammation (overexpression of translocator protein), (3) Elements of the cholinergic system, (4) Elements of monoamine neurotransmitter systems, (5) Synaptic density, (6) Cerebral energy metabolism (glucose transport/ hexokinase), and (7) Various other proteins. This last category contains proteins involved in mechanisms underlying neuroinflammation or cognitive impairment, which may also be potential therapeutic targets. Many receptors belong to this category: AMPA, cannabinoid, colony stimulating factor 1, metabotropic glutamate receptor 1 and 5 (mGluR1, mGluR5), opioid (kappa, mu), purinergic (P2X7, P2Y12), sigma-1, sigma-2, receptor for advanced glycation endproducts, and triggering receptor expressed on myeloid cells-1, besides several enzymes: cyclooxygenase-1 and 2 (COX-1, COX-2), phosphodiesterase-5 and 10 (PDE5, PDE10), and tropomyosin receptor kinase. Significant advances in neuroimaging have been made in the last 15 years. The use of 2-[18F]-fluoro-2-deoxy-D-glucose (FDG) for quantification of regional cerebral glucose metabolism is well-established. Three tracers for ß-amyloid plaques have been approved by the Food and Drug Administration and European Medicines Agency. Several tracers for tau neurofibrillary tangles are already applied in clinical research. Since many novel agents are in the preclinical or experimental stage of development, further advances in nuclear medicine imaging can be expected in the near future. PET studies with established tracers and tracers for novel targets may result in early diagnosis and better classification of neurodegenerative disorders and in accurate monitoring of therapy trials which involve these targets. PET data have prognostic value and may be used to assess the response of the human brain to interventions, or to select the appropriate treatment strategy for an individual patient.
Collapse
Affiliation(s)
- Aren van Waarde
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands.
| | - Sofia Marcolini
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands
| | - Peter Paul de Deyn
- University of Groningen, University Medical Center Groningen, Department of Neurology, Groningen, the Netherlands; University of Antwerp, Born-Bunge Institute, Neurochemistry and Behavior, Campus Drie Eiken, Wilrijk, Belgium
| | - Rudi A J O Dierckx
- University of Groningen, University Medical Center Groningen, Department of Nuclear Medicine and Molecular Imaging, Groningen, the Netherlands; Ghent University, Ghent, Belgium
| |
Collapse
|
105
|
Spatial topography of the basal forebrain cholinergic projections: Organization and vulnerability to degeneration. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:159-173. [PMID: 34225960 DOI: 10.1016/b978-0-12-819975-6.00008-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The basal forebrain (BF) cholinergic system constitutes a heterogeneous cluster of large projection neurons that innervate the entire cortical mantle and amygdala. Cholinergic neuromodulation plays a critical role in regulating cognition and behavior, as well as maintenance of cellular homeostasis. Decades of postmortem histology research have demonstrated that the BF cholinergic neurons are selectively vulnerable to aging and age-related neuropathology in degenerative diseases such as Alzheimer's and Parkinson's diseases. Emerging evidence from in vivo neuroimaging research, which permits longitudinal tracking of at-risk individuals, indicates that cholinergic neurodegeneration might play an earlier and more pivotal role in these diseases than was previously appreciated. Despite these advances, our understanding of the organization and functions of the BF cholinergic system mostly derives from nonhuman animal research. In this chapter, we begin with a review of the topographical organization of the BF cholinergic system in rodent and nonhuman primate models. We then discuss basic and clinical neuroscience research in humans, which has started to translate and extend the nonhuman animal research using novel noninvasive neuroimaging techniques. We focus on converging evidence indicating that the selective vulnerability of cholinergic neurons in Alzheimer's and Parkinson's diseases is expressed along a rostral-caudal topography in the BF. We close with a discussion of why this topography of vulnerability in the BF may occur and why it is relevant to the clinician.
Collapse
|
106
|
Modulation of arousal and sleep/wake architecture by M 1 PAM VU0453595 across young and aged rodents and nonhuman primates. Neuropsychopharmacology 2020; 45:2219-2228. [PMID: 32868847 PMCID: PMC7784923 DOI: 10.1038/s41386-020-00812-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/13/2020] [Indexed: 02/01/2023]
Abstract
Degeneration of basal forebrain cholinergic circuitry represents an early event in the development of Alzheimer's disease (AD). These alterations in central cholinergic function are associated with disruptions in arousal, sleep/wake architecture, and cognition. Changes in sleep/wake architecture are also present in normal aging and may represent a significant risk factor for AD. M1 muscarinic acetylcholine receptor (mAChR) positive allosteric modulators (PAMs) have been reported to enhance cognition across preclinical species and may also provide beneficial effects for age- and/or neurodegenerative disease-related changes in arousal and sleep. In the present study, electroencephalography was conducted in young animals (mice, rats and nonhuman primates [NHPs]) and in aged mice to examine the effects of the selective M1 PAM VU0453595 in comparison with the acetylcholinesterase inhibitor donepezil, M1/M4 agonist xanomeline (in NHPs), and M1 PAM BQCA (in rats) on sleep/wake architecture and arousal. In young wildtype mice, rats, and NHPs, but not in M1 mAChR KO mice, VU0453595 produced dose-related increases in high frequency gamma power, a correlate of arousal and cognition enhancement, without altering duration of time across all sleep/wake stages. Effects of VU0453595 in NHPs were observed within a dose range that did not induce cholinergic-mediated adverse effects. In contrast, donepezil and xanomeline increased time awake in rodents and engendered dose-limiting adverse effects in NHPs. Finally, VU0453595 attenuated age-related decreases in REM sleep duration in aged wildtype mice. Development of M1 PAMs represents a viable strategy for attenuating age-related and dementia-related pathological disturbances of sleep and arousal.
Collapse
|
107
|
Marzoughi S, Banerjee A, Jutzeler CR, Prado MAM, Rosner J, Cragg JJ, Cashman N. Tardive neurotoxicity of anticholinergic drugs: A review. J Neurochem 2020; 158:1334-1344. [PMID: 33222198 DOI: 10.1111/jnc.15244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
The cholinergic system is a complex neurotransmitter system with functional involvement at multiple levels of the nervous system including the cerebral cortex, spinal cord, autonomic nervous system, and neuromuscular junction. Anticholinergic medications are among the most prescribed medications, making up one-third to one-half of all medications prescribed for seniors. Recent evidence has linked long-term use of anticholinergic medications and dementia. Emerging evidence implicates the cholinergic system in the regulation of cerebral vasculature as well as neuroinflammation, suggesting that anticholinergic medications may contribute to absolute risk and progression of neurodegenerative diseases. In this review, we explore the involvement of the cholinergic system in various neurodegenerative diseases and the possible detrimental effects of anticholinergic medications on the onset and progression of these disorders. We identified references by searching the PubMed and Cochrane database between January 1990 and September 2019 for English-language animal and human studies including randomized clinical trials (RCTs), meta-analyses, systematic reviews, and observational studies. In addition, we conducted a manual search of reference lists from retrieved studies. Long-term anticholinergic medication exposure may have detrimental consequences beyond well-documented short-term cognitive effects, through a variety of mechanisms either directly impacting cholinergic neurotransmission or through receptors expressed on the vasculature or immune cells, providing a pathophysiological framework for complex interactions across the entire neuroaxis.
Collapse
Affiliation(s)
- Sina Marzoughi
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ankur Banerjee
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Catherine R Jutzeler
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Marco A M Prado
- Robarts Research Institute, The University of Western Ontario, London, ON, Canada
| | - Jan Rosner
- Collaboration for Outcomes Research and Evaluation (CORE), Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jacquelyn J Cragg
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Neil Cashman
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
108
|
Alterations of Brain Networks in Alzheimer's Disease and Mild Cognitive Impairment: A Resting State fMRI Study Based on a Population-specific Brain Template. Neuroscience 2020; 452:192-207. [PMID: 33197505 DOI: 10.1016/j.neuroscience.2020.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 10/18/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
This study aimed to investigate the alterations in brain networks in patients with Alzheimer's disease (AD) and mild cognitive impairment (MCI) based on a population-specific brain template. Previous studies on AD brain networks using graph theory rarely adopted brain templates specific for certain ethnicities. In this study, patients were divided into 3 groups: AD (n = 24), MCI (n = 27), and healthy controls (HCs, n = 33), and all of the subjects are Chinese. Functional brain networks were constructed for each group based on a Chinese brain template using resting-state functional magnetic resonance imaging (rs-fMRI) data; several graph metrics were calculated. Graph metrics with significant differences after false discovery rate (FDR) correction were analyzed with respect to correlations with four neuropsychological test scores: Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), Activities of Daily Living (ADL), and Clinical Dementia Rating (CDR), which assessed the subjects' cognitive functions and ability to engage in ADL. Graph metrics including assortativity coefficient, nodal degree centrality, nodal clustering coefficient, nodal efficiency, and nodal local efficiency of the frontal gyrus and cerebellum were significantly altered in AD and MCI compared with HC. Several graph metrics were significantly correlated with cognitive function and the ability to engage in daily activities. The findings suggest that altered graph metrics in the frontal gyrus may reflect brain plasticity, and that patients with MCI may have unique graph metric alterations in the cerebellum. Future graph analysis studies on functional brain networks in AD and MCI based on population-specific brain atlases for particular ethnicities may prove valuable.
Collapse
|
109
|
van der Zee S, Müller MLTM, Kanel P, van Laar T, Bohnen NI. Cholinergic Denervation Patterns Across Cognitive Domains in Parkinson's Disease. Mov Disord 2020; 36:642-650. [PMID: 33137238 DOI: 10.1002/mds.28360] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/17/2020] [Accepted: 10/05/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The cholinergic system plays a key role in cognitive impairment in Parkinson's disease (PD). Previous acetylcholinesterase positron emission tomography imaging studies found memory, attention, and executive function correlates of global cortical cholinergic losses. Vesicular acetylcholine transporter positron emission tomography allows for more accurate topographic assessment of not only cortical but also subcortical cholinergic changes. OBJECTIVE The objectiveof this study was to investigate the topographic relationship between cognitive functioning and regional cholinergic innervation in patients with PD. METHODS A total of 86 nondemented patients with PD (mean ± SD age 67.8 ± 7.6 years, motor disease duration 5.8 ± 4.6 years), and 12 healthy control participants (age 67.8 ± 7.8 years) underwent cholinergic [18 F]Fluoroethoxybenzovesamicol positron emission tomography imaging. Patients with PD underwent neuropsychological assessment. The z scores for each cognitive domain were determined using an age-matched, gender-matched, and educational level-matched control group. Correlations between domain-specific cognitive functioning and cholinergic innervation were examined, controlling for motor impairments and levodopa equivalent dose. Additional correlational analyses were performed using a mask limited to PD versus normal aging binding differences to assess for disease-specific versus normal aging effects. RESULTS Voxel-based whole-brain analysis demonstrated partial overlapping topography across cognitive domains, with most robust correlations in the domains of memory, attention, and executive functioning (P < 0.01, corrected for multiple comparisons). The shared pattern included the cingulate cortex, insula/operculum, and (visual) thalamus. CONCLUSION Our results confirm and expand on previous observations of cholinergic system involvement in cognitive functioning in PD. The topographic overlap across domains may reflect a partially shared cholinergic functionality underlying cognitive functioning, representing a combination of disease-specific and aging effects. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sygrid van der Zee
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurology and Department of Neuropsychology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn L T M Müller
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Prabesh Kanel
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Teus van Laar
- Department of Neurology and Department of Neuropsychology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nicolaas I Bohnen
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, Michigan, USA.,Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA.,Neurology Service and Geriatric Research Education and Clinical Center (GRECC), Veterans Administration Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
110
|
Cacabelos R. Pharmacogenetic considerations when prescribing cholinesterase inhibitors for the treatment of Alzheimer's disease. Expert Opin Drug Metab Toxicol 2020; 16:673-701. [PMID: 32520597 DOI: 10.1080/17425255.2020.1779700] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Cholinergic dysfunction, demonstrated in the late 1970s and early 1980s, led to the introduction of acetylcholinesterase inhibitors (AChEIs) in 1993 (Tacrine) to enhance cholinergic neurotransmission as the first line of treatment against Alzheimer's disease (AD). The new generation of AChEIs, represented by Donepezil (1996), Galantamine (2001) and Rivastigmine (2002), is the only treatment for AD to date, together with Memantine (2003). AChEIs are not devoid of side-effects and their cost-effectiveness is limited. An option to optimize the correct use of AChEIs is the implementation of pharmacogenetics (PGx) in the clinical practice. AREAS COVERED (i) The cholinergic system in AD, (ii) principles of AD PGx, (iii) PGx of Donepezil, Galantamine, Rivastigmine, Huperzine and other treatments, and (iv) practical recommendations. EXPERT OPINION The most relevant genes influencing AChEI efficacy and safety are APOE and CYPs. APOE-4 carriers are the worst responders to AChEIs. With the exception of Rivastigmine (UGT2B7, BCHE-K), the other AChEIs are primarily metabolized via CYP2D6, CYP3A4, and UGT enzymes, with involvement of ABC transporters and cholinergic genes (CHAT, ACHE, BCHE, SLC5A7, SLC18A3, CHRNA7) in most ethnic groups. Defective variants may affect the clinical response to AChEIs. PGx geno-phenotyping is highly recommended prior to treatment.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, EuroEspes Biomedical Research Center, International Center of Neuroscience and Genomic Medicine , Bergondo, Corunna, Spain
| |
Collapse
|
111
|
Fernández-Cabello S, Kronbichler M, Van Dijk KRA, Goodman JA, Spreng RN, Schmitz TW, on behalf of the Alzheimer’s Disease Neuroimaging Initiative. Basal forebrain volume reliably predicts the cortical spread of Alzheimer's degeneration. Brain 2020; 143:993-1009. [PMID: 32203580 PMCID: PMC7092749 DOI: 10.1093/brain/awaa012] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/21/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease neurodegeneration is thought to spread across anatomically and functionally connected brain regions. However, the precise sequence of spread remains ambiguous. The prevailing model used to guide in vivo human neuroimaging and non-human animal research assumes that Alzheimer's degeneration starts in the entorhinal cortices, before spreading to the temporoparietal cortex. Challenging this model, we previously provided evidence that in vivo markers of neurodegeneration within the nucleus basalis of Meynert (NbM), a subregion of the basal forebrain heavily populated by cortically projecting cholinergic neurons, precedes and predicts entorhinal degeneration. There have been few systematic attempts at directly comparing staging models using in vivo longitudinal biomarker data, and none to our knowledge testing if comparative evidence generalizes across independent samples. Here we addressed the sequence of pathological staging in Alzheimer's disease using two independent samples of the Alzheimer's Disease Neuroimaging Initiative (n1 = 284; n2 = 553) with harmonized CSF assays of amyloid-β and hyperphosphorylated tau (pTau), and longitudinal structural MRI data over 2 years. We derived measures of grey matter degeneration in a priori NbM and the entorhinal cortical regions of interest. To examine the spreading of degeneration, we used a predictive modelling strategy that tests whether baseline grey matter volume in a seed region accounts for longitudinal change in a target region. We demonstrated that predictive spread favoured the NbM→entorhinal over the entorhinal→NbM model. This evidence generalized across the independent samples. We also showed that CSF concentrations of pTau/amyloid-β moderated the observed predictive relationship, consistent with evidence in rodent models of an underlying trans-synaptic mechanism of pathophysiological spread. The moderating effect of CSF was robust to additional factors, including clinical diagnosis. We then applied our predictive modelling strategy to an exploratory whole-brain voxel-wise analysis to examine the spatial specificity of the NbM→entorhinal model. We found that smaller baseline NbM volumes predicted greater degeneration in localized regions of the entorhinal and perirhinal cortices. By contrast, smaller baseline entorhinal volumes predicted degeneration in the medial temporal cortex, recapitulating a prior influential staging model. Our findings suggest that degeneration of the basal forebrain cholinergic projection system is a robust and reliable upstream event of entorhinal and neocortical degeneration, calling into question a prevailing view of Alzheimer's disease pathogenesis.
Collapse
Affiliation(s)
- Sara Fernández-Cabello
- Department of Psychology, University of Salzburg, Salzburg, Austria
- Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| | - Martin Kronbichler
- Department of Psychology, University of Salzburg, Salzburg, Austria
- Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
- Neuroscience Institute, Christian-Doppler Medical Centre, Paracelsus Medical University, Salzburg, Austria
| | - Koene R A Van Dijk
- Clinical and Translational Imaging, Early Clinical Development, Pfizer Inc, Cambridge, MA, USA
| | - James A Goodman
- Clinical and Translational Imaging, Early Clinical Development, Pfizer Inc, Cambridge, MA, USA
| | - R Nathan Spreng
- Laboratory of Brain and Cognition, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Departments of Psychiatry and Psychology, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Verdun, QC, Canada
- McConnell Brain Imaging Centre, McGill University, Montreal, QC, Canada
| | - Taylor W Schmitz
- Brain and Mind Institute, Western University, London, ON, Canada
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | | |
Collapse
|
112
|
Liu H, Luo Z, Gu J, Su Y, Flores H, Parsons SM, Zhou Y, Perlmutter JS, Tu Z. The impact of dopamine D 2-like agonist/antagonist on [ 18F]VAT PET measurement of VAChT in the brain of nonhuman primates. Eur J Pharm Sci 2020; 143:105152. [PMID: 31740395 PMCID: PMC6980745 DOI: 10.1016/j.ejps.2019.105152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/15/2019] [Accepted: 11/14/2019] [Indexed: 01/11/2023]
Abstract
Vesicular acetylcholine transporter (VAChT) is a promising target for a PET measure of cholinergic deficits which contribute to cognitive impairments. Dopamine D2-like agonists and antagonists are frequently used in the elderly and could alter cholinergic function and VAChT level. Therefore, pretreatment with dopamine D2-like drugs may interfere with PET measures using [18F]VAT, a specific VAChT radioligand. Herein, we investigated the impact of dopaminergic D2-like antagonist/agonist on VAChT level in the brain of macaques using [18F]VAT PET. PET imaging studies were carried out on macaques at baseline or pretreatment conditions. For pretreatment, animals were injected using a VAChT inhibitor (-)-vesamicol, a D2-like antagonist (-)-eticlopride, and a D2-like agonist (-)-quinpirole, separately. (-)-Vesamicol was injected at escalating doses of 0.025, 0.05, 0.125, 0.25 and 0.35 mg/kg; (-)-eticlopride was injected at escalating doses of 0.01, 0.10 and 0.30 mg/kg; (-)-quinpirole was injected at escalating doses of 0.20, 0.30, and 0.50 mg/kg. PET data showed [18F]VAT uptake declined in a dose-dependent manner by (-)-vesamicol pretreatment, demonstrating [18F]VAT uptake is sensitive to reflect the availability of VAChT binding sites. Furthermore, (-)-eticlopride increased [18F]VAT striatal uptake in a dose-dependent manner, while (-)-quinpirole decreased its uptake, suggesting striatal VAChT levels can be regulated by D2-like drug administration. Our findings confirmed [18F]VAT offers a reliable tool to in vivo assess the availability of VAChT binding sites. More importantly, PET with [18F]VAT successfully quantified the impact of dopaminergic D2-like drugs on striatal VAChT level, suggesting [18F]VAT has great potential for investigating the interaction between dopaminergic and cholinergic systems in vivo.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zonghua Luo
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiwei Gu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Su
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hubert Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Yun Zhou
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
113
|
Cisneros-Franco JM, Voss P, Kang MS, Thomas ME, Côté J, Ross K, Gaudreau P, Rudko DA, Rosa-Neto P, de-Villers-Sidani É. PET Imaging of Perceptual Learning-Induced Changes in the Aged Rodent Cholinergic System. Front Neurosci 2020; 13:1438. [PMID: 32038142 PMCID: PMC6985428 DOI: 10.3389/fnins.2019.01438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
The cholinergic system enhances attention and gates plasticity, making it a major regulator of adult learning. With aging, however, progressive degeneration of the cholinergic system impairs both the acquisition of new skills and functional recovery following neurological injury. Although cognitive training and perceptual learning have been shown to enhance auditory cortical processing, their specific impact on the cholinergic system remains unknown. Here we used [18F]FEOBV, a positron emission tomography (PET) radioligand that selectively binds to the vesicular acetylcholine transporter (VAChT), as a proxy to assess whether training on a perceptual task results in increased cholinergic neurotransmission. We show for the first time that perceptual learning is associated with region-specific changes in cholinergic neurotransmission, as detected by [18F]FEOBV PET imaging and corroborated with immunohistochemistry.
Collapse
Affiliation(s)
- J Miguel Cisneros-Franco
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Patrice Voss
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Maryse E Thomas
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Jonathan Côté
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| | - Karen Ross
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Pierrette Gaudreau
- Réseau Québécois de Recherche sur le Vieillissement, Université de Montréal, Montreal, QC, Canada
| | - David A Rudko
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Research Centre for Studies in Aging, McGill University, Montreal, QC, Canada
| | - Étienne de-Villers-Sidani
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada.,Centre for Research on Brain, Language and Music, McGill University, Montreal, QC, Canada
| |
Collapse
|
114
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
115
|
Xhima K, Markham-Coultes K, Nedev H, Heinen S, Saragovi HU, Hynynen K, Aubert I. Focused ultrasound delivery of a selective TrkA agonist rescues cholinergic function in a mouse model of Alzheimer's disease. SCIENCE ADVANCES 2020; 6:eaax6646. [PMID: 32010781 PMCID: PMC6976301 DOI: 10.1126/sciadv.aax6646] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/18/2019] [Indexed: 05/24/2023]
Abstract
The degeneration of cholinergic neurons is a prominent feature of Alzheimer's disease (AD). In animal models of injury and aging, nerve growth factor (NGF) enhances cholinergic cell survival and function, contributing to improved memory. In the presence of AD pathology, however, NGF-related therapeutics have yet to fulfill their regenerative potential. We propose that stimulating the TrkA receptor, without p75NTR activation, is key for therapeutic efficacy. Supporting this hypothesis, the selective TrkA agonist D3 rescued neurotrophin signaling in TgCRND8 mice, whereas NGF, interacting with both TrkA and p75NTR, did not. D3, delivered intravenously and noninvasively to the basal forebrain using MRI-guided focused ultrasound (MRIgFUS)-mediated blood-brain barrier (BBB) permeability activated TrkA-related signaling cascades and enhanced cholinergic neurotransmission. Recent clinical trials support the safety and feasibility of MRIgFUS BBB modulation in AD patients. Neuroprotective agents targeting TrkA, combined with MRIgFUS BBB modulation, represent a promising strategy to counter neurodegeneration in AD.
Collapse
Affiliation(s)
- K. Xhima
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - K. Markham-Coultes
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - H. Nedev
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - S. Heinen
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - H. U. Saragovi
- Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - K. Hynynen
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - I. Aubert
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
116
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
117
|
Beaurain M, Salabert AS, Ribeiro MJ, Arlicot N, Damier P, Le Jeune F, Demonet JF, Payoux P. Innovative Molecular Imaging for Clinical Research, Therapeutic Stratification, and Nosography in Neuroscience. Front Med (Lausanne) 2019; 6:268. [PMID: 31828073 PMCID: PMC6890558 DOI: 10.3389/fmed.2019.00268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 11/01/2019] [Indexed: 01/06/2023] Open
Abstract
Over the past few decades, several radiotracers have been developed for neuroimaging applications, especially in PET. Because of their low steric hindrance, PET radionuclides can be used to label molecules that are small enough to cross the blood brain barrier, without modifying their biological properties. As the use of 11C is limited by its short physical half-life (20 min), there has been an increasing focus on developing tracers labeled with 18F for clinical use. The first such tracers allowed cerebral blood flow and glucose metabolism to be measured, and the development of molecular imaging has since enabled to focus more closely on specific targets such as receptors, neurotransmitter transporters, and other proteins. Hence, PET and SPECT biomarkers have become indispensable for innovative clinical research. Currently, the treatment options for a number of pathologies, notably neurodegenerative diseases, remain only supportive and symptomatic. Treatments that slow down or reverse disease progression are therefore the subject of numerous studies, in which molecular imaging is proving to be a powerful tool. PET and SPECT biomarkers already make it possible to diagnose several neurological diseases in vivo and at preclinical stages, yielding topographic, and quantitative data about the target. As a result, they can be used for assessing patients' eligibility for new treatments, or for treatment follow-up. The aim of the present review was to map major innovative radiotracers used in neuroscience, and explain their contribution to clinical research. We categorized them according to their target: dopaminergic, cholinergic or serotoninergic systems, β-amyloid plaques, tau protein, neuroinflammation, glutamate or GABA receptors, or α-synuclein. Most neurological disorders, and indeed mental disorders, involve the dysfunction of one or more of these targets. Combinations of molecular imaging biomarkers can afford us a better understanding of the mechanisms underlying disease development over time, and contribute to early detection/screening, diagnosis, therapy delivery/monitoring, and treatment follow-up in both research and clinical settings.
Collapse
Affiliation(s)
- Marie Beaurain
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Anne-Sophie Salabert
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| | - Maria Joao Ribeiro
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,Inserm CIC 1415, University Hospital, Tours, France.,CHRU Tours, Tours, France
| | - Philippe Damier
- Inserm U913, Neurology Department, University Hospital, Nantes, France
| | | | - Jean-François Demonet
- Leenards Memory Centre, Department of Clinical Neuroscience, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Pierre Payoux
- CHU de Toulouse, Toulouse, France.,ToNIC, Toulouse NeuroImaging Center, Inserm U1214, Toulouse, France
| |
Collapse
|
118
|
Richter N, Nellessen N, Dronse J, Dillen K, Jacobs HIL, Langen KJ, Dietlein M, Kracht L, Neumaier B, Fink GR, Kukolja J, Onur OA. Spatial distributions of cholinergic impairment and neuronal hypometabolism differ in MCI due to AD. NEUROIMAGE-CLINICAL 2019; 24:101978. [PMID: 31422337 PMCID: PMC6706587 DOI: 10.1016/j.nicl.2019.101978] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/24/2019] [Accepted: 08/08/2019] [Indexed: 12/31/2022]
Abstract
Elucidating the relationship between neuronal metabolism and the integrity of the cholinergic system is prerequisite for a profound understanding of cholinergic dysfunction in Alzheimer's disease. The cholinergic system can be investigated specifically using positron emission tomography (PET) with [11C]N-methyl-4-piperidyl-acetate (MP4A), while neuronal metabolism is often assessed with 2-deoxy-2-[18F]fluoro-d-glucose-(FDG) PET. We hypothesised a close correlation between MP4A-perfusion and FDG-uptake, permitting inferences about metabolism from MP4A-perfusion, and investigated the patterns of neuronal hypometabolism and cholinergic impairment in non-demented AD patients. MP4A-PET was performed in 18 cognitively normal adults and 19 patients with mild cognitive impairment (MCI) and positive AD biomarkers. In nine patients with additional FDG-PET, the sum images of every combination of consecutive early MP4A-frames were correlated with FDG-scans to determine the optimal time window for assessing MP4A-perfusion. Acetylcholinesterase (AChE) activity was estimated using a 3-compartmental model. Group comparisons of MP4A-perfusion and AChE-activity were performed using the entire sample. The highest correlation between MP4A-perfusion and FDG-uptake across the cerebral cortex was observed 60-450 s after injection (r = 0.867). The patterns of hypometabolism (FDG-PET) and hypoperfusion (MP4A-PET) in MCI covered areas known to be hypometabolic early in AD, while AChE activity was mainly reduced in the lateral temporal cortex and the occipital lobe, sparing posterior midline structures. Data indicate that patterns of cholinergic impairment and neuronal hypometabolism differ significantly at the stage of MCI in AD, implying distinct underlying pathologies, and suggesting potential predictors of the response to cholinergic pharmacotherapy.
Collapse
Affiliation(s)
- Nils Richter
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany.
| | - Nils Nellessen
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Julian Dronse
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Kim Dillen
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Heidi I L Jacobs
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States of America; The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States of America; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Karl-Josef Langen
- Medical Imaging Physics (INM-4), Institute of Neuroscience and Medicine (INM-4), Research Center Jülich, 52425 Jülich, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Lutz Kracht
- Max-Planck-Institute for Metabolism Research, 50937 Cologne, Germany; Department of Nuclear Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Bernd Neumaier
- Institute for Radiochemistry and Experimental Molecular Imaging, University Hospital Cologne, 50937 Cologne, Germany; Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Research Center Jülich, 52425 Jülich, Germany
| | - Gereon R Fink
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| | - Juraj Kukolja
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany; Department of Neurology and Neurophysiology, Helios University Hospital Wuppertal, 42283 Wuppertal, Germany
| | - Oezguer A Onur
- Department of Neurology, University Hospital Cologne, 50937 Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Jülich, 52425 Jülich, Germany
| |
Collapse
|
119
|
Tiepolt S, Patt M, Aghakhanyan G, Meyer PM, Hesse S, Barthel H, Sabri O. Current radiotracers to image neurodegenerative diseases. EJNMMI Radiopharm Chem 2019; 4:17. [PMID: 31659510 PMCID: PMC6660543 DOI: 10.1186/s41181-019-0070-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
The term of neurodegenerative diseases covers a heterogeneous group of disorders that are distinguished by progressive degeneration of the structure and function of the nervous system such as dementias, movement disorders, motor neuron disorders, as well as some prion disorders. In recent years, a paradigm shift started for the diagnosis of neurodegenerative diseases, for which successively clinical testing is supplemented by biomarker information. In research scenarios, it was even proposed recently to substitute the current syndromic by a biological definition of Alzheimer's diseases. PET examinations with various radiotracers play an important role in providing non-invasive biomarkers and co-morbidity information in neurodegeneration. Information on co-morbidity, e.g. Aβ plaques and Lewy-bodies or Aβ plaques in patients with aphasia or the absence of Aβ plaques in clinical AD patients are of interest to expand our knowledge about the pathogenesis of different phenotypically defined neurodegenerative diseases. Moreover, this information is also important in therapeutic trials targeting histopathological abnormalities.The aim of this review is to present an overview of the currently available radiotracers for imaging neurodegenerative diseases in research and in routine clinical settings. In this context, we also provide a short summary of the most frequent neurodegenerative diseases from a nuclear medicine point of view, their clinical and pathophysiological as well as nuclear imaging characteristics, and the resulting need for new radiotracers.
Collapse
Affiliation(s)
- Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Gayane Aghakhanyan
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Philipp M. Meyer
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| |
Collapse
|
120
|
Perani D, Iaccarino L, Lammertsma AA, Windhorst AD, Edison P, Boellaard R, Hansson O, Nordberg A, Jacobs AH. A new perspective for advanced positron emission tomography-based molecular imaging in neurodegenerative proteinopathies. Alzheimers Dement 2019; 15:1081-1103. [PMID: 31230910 DOI: 10.1016/j.jalz.2019.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/21/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Recent studies in neurodegenerative conditions have increasingly highlighted that the same neuropathology can trigger different clinical phenotypes or, vice-versa, that similar phenotypes can be triggered by different neuropathologies. This evidence has called for the adoption of a pathology spectrum-based approach to study neurodegenerative proteinopathies. These conditions share brain deposition of abnormal protein aggregates, leading to aberrant biochemical, metabolic, functional, and structural changes. Positron emission tomography (PET) is a well-recognized and unique tool for the in vivo assessment of brain neuropathology, and novel PET techniques are emerging for the study of specific protein species. Today, key applications of PET range from early research and clinical diagnostic tools to their use in clinical trials for both participants screening and outcome evaluation. This position article critically reviews the role of distinct PET molecular tracers for different neurodegenerative proteinopathies, highlighting their strengths, weaknesses, and opportunities, with special emphasis on methodological challenges and future applications.
Collapse
Affiliation(s)
- Daniela Perani
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Iaccarino
- Vita-Salute San Raffaele University, Nuclear Medicine Unit San Raffaele Hospital, Division of Neuroscience San Raffaele Scientific Institute, Milan, Italy
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul Edison
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK; Neurology Imaging Unit, Imperial College London, London, UK
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Center for Alzheimer Research, Stockholm, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany; Evangelische Kliniken Bonn gGmbH, Johanniter Krankenhaus, Bonn, Germany.
| | | |
Collapse
|
121
|
Gauthier S, Ng KP, Pascoal TA, Zhang H, Rosa-Neto P. Targeting Alzheimer's Disease at the Right Time and the Right Place: Validation of a Personalized Approach to Diagnosis and Treatment. J Alzheimers Dis 2019; 64:S23-S31. [PMID: 29504543 PMCID: PMC6004905 DOI: 10.3233/jad-179924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cautious optimism is appropriate for a near future (five years) time frame for a number of drugs acting on the different pathophysiological components of Alzheimer’s disease (amyloid deposition, tau hyperphosphorylation, neuroinflammation, vascular changes, to name the most important known so far). Since the relative weight of these components will be different between individuals and will even change over time for each individual, a ‘one drug fit for all’ approach is no longer defensible. Precision medicine using biomarkers in the diagnosis and treatment of Alzheimer’s disease is the new strategy.
Collapse
Affiliation(s)
- Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Tharick A Pascoal
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Hua Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Pedro Rosa-Neto
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| |
Collapse
|
122
|
Zee S, Vállez García D, Elsinga PH, Willemsen AT, Boersma HH, Gerritsen MJ, Spikman JM, Laar T. [
18
F]Fluoroethoxybenzovesamicol in Parkinson's disease patients: Quantification of a novel cholinergic positron emission tomography tracer. Mov Disord 2019; 34:924-926. [DOI: 10.1002/mds.27698] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/22/2019] [Accepted: 03/24/2019] [Indexed: 01/27/2023] Open
Affiliation(s)
- Sygrid Zee
- Department of NeurologyUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - David Vállez García
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - Antoon T.M. Willemsen
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - Hendrikus H. Boersma
- Department of Nuclear Medicine and Molecular ImagingUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
- Department of Clinical Pharmacy and PharmacologyUniversity Medical Center Groningen Groningen the Netherlands
| | - Marleen J.J. Gerritsen
- Department of NeurologyUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - Jacoba M. Spikman
- Department of NeurologyUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| | - Teus Laar
- Department of NeurologyUniversity of Groningen, University Medical Center Groningen Groningen the Netherlands
| |
Collapse
|
123
|
Bohnen NI, Kanel P, Zhou Z, Koeppe RA, Frey KA, Dauer WT, Albin RL, Müller MLTM. Cholinergic system changes of falls and freezing of gait in Parkinson's disease. Ann Neurol 2019; 85:538-549. [PMID: 30720884 PMCID: PMC6450746 DOI: 10.1002/ana.25430] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 02/03/2019] [Accepted: 02/03/2019] [Indexed: 01/16/2023]
Abstract
Objective Postural instability and gait difficulties (PIGDs) represent debilitating disturbances in Parkinson's disease (PD). Past acetylcholinesterase positron emission tomography (PET) imaging studies implicate cholinergic changes as significant contributors to PIGD features. These studies were limited in quantification of striatal cholinergic synapse integrity. Vesicular acetylcholine transporter (VAChT) PET ligands are better suited for evaluation of high binding areas. We examined associations between regional VAChT expression and freezing of gait (FoG) and falls. Methods Ninety‐four PD subjects underwent clinical assessment and VAChT ([18F]FEOBV) PET. Results Thirty‐five subjects (37.2%) reported a history of falls, and 15 (16%) had observed FoG. Univariate volume‐of‐interest analyses demonstrated significantly reduced thalamic (p = 0.0016) VAChT expression in fallers compared to nonfallers. VAChT expression was significantly reduced in the striatum (p = 0.0012) and limbic archicortex (p = 0.004) in freezers compared to nonfreezers. Whole‐brain voxel‐based analyses of FEOBV PET complemented these findings and showed more granular changes associated with falling history, including the right visual thalamus (especially the right lateral geniculate nucleus [LGN]), right caudate nucleus, and bilateral prefrontal regions. Freezers had prominent VAChT expression reductions in the bilateral striatum, temporal, and mesiofrontal limbic regions. Interpretation Our findings confirm and extend on previous PET findings of thalamic cholinergic deficits associated with falling history and now emphasize right visual thalamus complex changes, including the right LGN. FoG status is associated with reduced VAChT expression in striatal cholinergic interneurons and the limbic archicortex. These observations suggest different cholinergic systems changes underlying falls and FoG in PD. Ann Neurol 2019;85:538–549
Collapse
Affiliation(s)
- Nicolaas I Bohnen
- Radiology, University of Michigan, Ann Arbor, MI.,Neurology, University of Michigan, Ann Arbor, MI.,Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| | - Prabesh Kanel
- Radiology, University of Michigan, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| | - Zhi Zhou
- Radiology, University of Michigan, Ann Arbor, MI
| | - Robert A Koeppe
- Radiology, University of Michigan, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| | - Kirk A Frey
- Radiology, University of Michigan, Ann Arbor, MI.,Neurology, University of Michigan, Ann Arbor, MI
| | - William T Dauer
- Neurology, University of Michigan, Ann Arbor, MI.,Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| | - Roger L Albin
- Neurology, University of Michigan, Ann Arbor, MI.,Neurology Service and GRECC, Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| | - Martijn L T M Müller
- Radiology, University of Michigan, Ann Arbor, MI.,Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI
| |
Collapse
|
124
|
Quantification of brain cholinergic denervation in dementia with Lewy bodies using PET imaging with [ 18F]-FEOBV. Mol Psychiatry 2019; 24:322-327. [PMID: 30082840 PMCID: PMC6363916 DOI: 10.1038/s41380-018-0130-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 11/13/2022]
|
125
|
Prato FS, Pavlosky WF, Foster SC, Thiessen JD, Beaujot RP. Screening for Dementia Caused by Modifiable Lifestyle Choices Using Hybrid PET/MRI. J Alzheimers Dis Rep 2019; 3:31-45. [PMID: 30842996 PMCID: PMC6400112 DOI: 10.3233/adr-180098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2018] [Indexed: 12/19/2022] Open
Abstract
Significant advances in positron emission tomography (PET) and magnetic resonance imaging (MRI) brain imaging in the early detection of dementia indicate that hybrid PET/MRI would be an effective tool to screen for dementia in the population living with lifestyle risk factors. Here we investigate the associated costs and benefits along with the needed imaging infrastructure. A demographic analysis determined the prevalence of dementia and its incidence. The expected value of the screening program was calculated assuming a sensitivity and specificity of 0.9, a prevalence of 0.1, a QALY factor of 0.348, a willingness to pay $114,000 CAD and the cost per PET/MRI scan of $2,000 CAD. It was assumed that each head PET/MRI could screen 3,000 individuals per year. The prevalence of dementia is increasing by almost two-fold every 20 years due to the increased population at ages where dementia is more prevalent. It has been shown that a five-year delay in the incidence of dementia would decrease the prevalence by some 45%. In Canada, a five-year delay corresponds to a health care savings of $27,000 CAD per subject per year. The expected value for screening was estimated at $23,745 CAD. The number of subjects to be screened per year in Canada, USA, and China between 60 and 79 was 11,405,000. The corresponding number of head-only hybrid PET/MRI systems needed is 3,800. A brain PET/MRI screening program is financially justifiable with respect to health care costs and justifies the continuing development of MRI compatible brain PET technology.
Collapse
Affiliation(s)
- Frank S. Prato
- Department of Medical Biophysics, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | - William F. Pavlosky
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | | | - Jonathan D. Thiessen
- Department of Medical Biophysics, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | | |
Collapse
|
126
|
Bedard MA, Aghourian M, Legault-Denis C, Postuma RB, Soucy JP, Gagnon JF, Pelletier A, Montplaisir J. Brain cholinergic alterations in idiopathic REM sleep behaviour disorder: a PET imaging study with 18F-FEOBV. Sleep Med 2019; 58:35-41. [PMID: 31078078 DOI: 10.1016/j.sleep.2018.12.020] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/19/2018] [Accepted: 12/27/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND REM sleep behaviour disorder (RBD) occurs frequently in patients with synucleinopathies such as Parkinson's disease, dementia with Lewy body, or multiple system atrophy, but may also occur as a prodromal stage of those diseases; and is termed idiopathic RBD (iRBD) when not accompanied by other symptoms. Cholinergic degeneration of the mesopontine nuclei have been described in synucleinopathies with or without RBD, but this has not yet been explored in iRBD. We sought to assess cholinergic neuronal integrity in iRBD using PET neuroimaging with the 18F-fluoroethoxybenzovesamicol (FEOBV). METHODS The sample included 10 participants evenly divided between healthy subjects and patients with iRBD. Polysomnography and PET imaging with FEOBV were performed in all participants. Standardized uptake value ratios (SUVRs) were compared between the two groups using voxel wise t-tests. Non-parametric correlations were also computed in patients with iRBD between FEOBV uptake and muscle tonic and phasic activity during REM sleep. RESULTS Compared with healthy participants, significantly higher FEOBV uptakes were observed in patients with iRBD. The largest differences were observed in specific brainstem areas corresponding to the bulbar reticular formation, pontine coeruleus/subcoeruleus complex, tegmental periacqueductal grey, and mesopontine cholinergic nuclei. FEOBV uptake in iRBD was also higher than in controls in the ventromedial area of the thalamus, deep cerebellar nuclei, and some cortical territories (including the paracentral lobule, anterior cingulate, and orbitofrontal cortex). Significant correlation was found between muscle activity during REM sleep, and SUVR increases in both the mesopontine area and paracentral cortex. CONCLUSION We showed here for the first time the brain cholinergic alterations in patients with iRBD. As opposed to the cholinergic depletion described previously in RBD associated with clinical Parkinson's disease, increased cholinergic innervation was found in multiple areas in iRBD. The most significant changes were observed in brainstem areas containing structures involved in the promotion of REM sleep and muscle atonia. This suggests that iRBD might be a clinical condition in which compensatory cholinergic upregulation in those areas occurs in association with the initial phases of a neurodegenerative process leading to a clinically observable synucleinopathy.
Collapse
Affiliation(s)
- Marc-Andre Bedard
- NeuroQAM Centre, Université du Québec à Montréal (UQAM), Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, Canada
| | - Meghmik Aghourian
- NeuroQAM Centre, Université du Québec à Montréal (UQAM), Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, Canada
| | - Camille Legault-Denis
- NeuroQAM Centre, Université du Québec à Montréal (UQAM), Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, Canada
| | - Ronald B Postuma
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Canada; Department of Neurology and Neurosurgery, McGill University, Canada
| | - Jean-Paul Soucy
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Canada; Department of Radiology and Nuclear Medicine, Université de Montréal, Canada; PERFORM Centre, Concordia University, Canada
| | - Jean-François Gagnon
- NeuroQAM Centre, Université du Québec à Montréal (UQAM), Canada; Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Canada
| | - Amélie Pelletier
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Canada
| | - Jacques Montplaisir
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Coeur de Montréal, Canada; Department of Psychiatry, Université de Montréal, Canada.
| |
Collapse
|
127
|
Albin RL, Bohnen NI, Muller MLTM, Dauer WT, Sarter M, Frey KA, Koeppe RA. Regional vesicular acetylcholine transporter distribution in human brain: A [ 18 F]fluoroethoxybenzovesamicol positron emission tomography study. J Comp Neurol 2018; 526:2884-2897. [PMID: 30255936 DOI: 10.1002/cne.24541] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/07/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
Prior efforts to image cholinergic projections in human brain in vivo had significant technical limitations. We used the vesicular acetylcholine transporter (VAChT) ligand [18 F]fluoroethoxybenzovesamicol ([18 F]FEOBV) and positron emission tomography to determine the regional distribution of VAChT binding sites in normal human brain. We studied 29 subjects (mean age 47 [range 20-81] years; 18 men; 11 women). [18 F]FEOBV binding was highest in striatum, intermediate in the amygdala, hippocampal formation, thalamus, rostral brainstem, some cerebellar regions, and lower in other regions. Neocortical [18 F]FEOBV binding was inhomogeneous with relatively high binding in insula, BA24, BA25, BA27, BA28, BA34, BA35, pericentral cortex, and lowest in BA17-19. Thalamic [18 F]FEOBV binding was inhomogeneous with greatest binding in the lateral geniculate nuclei and relatively high binding in medial and posterior thalamus. Cerebellar cortical [18 F]FEOBV binding was high in vermis and flocculus, and lower in the lateral cortices. Brainstem [18 F]FEOBV binding was most prominent at the mesopontine junction, likely associated with the pedunculopontine-laterodorsal tegmental complex. Significant [18 F]FEOBV binding was present throughout the brainstem. Some regions, including the striatum, primary sensorimotor cortex, and anterior cingulate cortex exhibited age-related decreases in [18 F]FEOBV binding. These results are consistent with prior studies of cholinergic projections in other species and prior postmortem human studies. There is a distinctive pattern of human neocortical VChAT expression. The patterns of thalamic and cerebellar cortical cholinergic terminal distribution are likely unique to humans. Normal aging is associated with regionally specific reductions in [18 F]FEOBV binding in some cortical regions and the striatum.
Collapse
Affiliation(s)
- Roger L Albin
- Neurology Service & GRECC, VAAAHS, Ann Arbor, Michigan.,Department of Neurology, University of Michigan, Ann Arbor, Michigan.,University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Michigan Alzheimer Disease Center, Ann Arbor, Michigan
| | - Nicolaas I Bohnen
- Neurology Service & GRECC, VAAAHS, Ann Arbor, Michigan.,Department of Neurology, University of Michigan, Ann Arbor, Michigan.,University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Martijn L T M Muller
- University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - William T Dauer
- Neurology Service & GRECC, VAAAHS, Ann Arbor, Michigan.,Department of Neurology, University of Michigan, Ann Arbor, Michigan.,University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Martin Sarter
- University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Kirk A Frey
- Department of Neurology, University of Michigan, Ann Arbor, Michigan.,Department of Radiology, University of Michigan, Ann Arbor, Michigan
| | - Robert A Koeppe
- University of Michigan Morris K. Udall Center of Excellence for Research in Parkinson's Disease, Ann Arbor, Michigan.,Department of Radiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
128
|
Yue X, Luo Z, Liu H, Kaneshige K, Parsons SM, Perlmutter JS, Tu Z. Radiosynthesis and evaluation of a fluorine-18 labeled radioligand targeting vesicular acetylcholine transporter. Bioorg Med Chem Lett 2018; 28:3425-3430. [PMID: 30274694 DOI: 10.1016/j.bmcl.2018.09.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/21/2018] [Accepted: 09/22/2018] [Indexed: 12/28/2022]
Abstract
Vesicular acetylcholine transporter (VAChT) is a reliable biomarker for assessing the loss of cholinergic neurons in the brain that is associated with cognitive impairment of patients. 5-Hydrotetralin compound (±)-5-OH-VAT is potent (Ki = 4.64 ± 0.32 nM) and selective for VAChT (>1800-fold and 398-fold for σ1 and σ2 receptor, respectively) with favorable hydrophilicity (LogD = 1.78), while (-)-5-OH-VAT originally serves as the radiolabeling precursor of (-)-[18F]VAT, a promising VAChT radiotracer with a logD value of 2.56. To evaluate (-)-5-OH-[18F]VAT as a radiotracer for VAChT, we performed in vitro binding assay to determine the potency of the minus enantiomer (-)-5-OH-VAT and plus enantiomer (+)-5-OH-VAT, indicating that (-)-5-OH-VAT is a more potent VAChT enantiomer. Radiosynthesis of (-)-5-OH-[18F]VAT was explored using three strategies. (-)-5-OH-[18F]VAT was achieved with a good yield (24 ± 6%) and high molar activity (∼37 GBq/µmol, at the end of synthesis) using a microwave assisted two-step one-pot procedure that started with di-MOM protected nitro-containing precursor (-)-6. MicroPET studies in the brain of nonhuman primate (NHP) suggest that (-)-5-OH-[18F]VAT readily penetrated the blood brain barrier and specifically accumulated in the VAChT-enriched striatum with improved washout kinetics from striatum compared to [18F]VAT. Nevertheless, the lower target to non-target ratio may limit its use for in vivo measurement of the VAChT level in the brain.
Collapse
Affiliation(s)
- Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Zonghua Luo
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Kota Kaneshige
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, United States
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, United States
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, United States; Department of Neurology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St Louis, MO 63110, United States.
| |
Collapse
|
129
|
Molecular Imaging of the Cholinergic System in Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 141:211-250. [PMID: 30314597 DOI: 10.1016/bs.irn.2018.07.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
One of the first identified neurotransmitters in the brain, acetylcholine, is an important modulator that drives changes in neuronal and glial activity. For more than two decades, the main focus of molecular imaging of the cholinergic system in Parkinson's disease (PD) has been on cognitive changes. Imaging studies have confirmed that degeneration of the cholinergic system is a major determinant of dementia in PD. Within the last decade, the focus is expanding to studying cholinergic correlates of mobility impairments, dyskinesias, olfaction, sleep, visual hallucinations and risk taking behavior in this disorder. These studies increasingly recognize that the regional topography of cholinergic brain areas associates with specific functions. In parallel with this trend, more recent molecular cholinergic imaging approaches are investigating cholinergic modulatory functions and contributions to large-scale brain network functions. A novel area of research is imaging cholinergic innervation functions of peripheral autonomic organs that may have the potential of future prodromal diagnosis of PD. Finally, emerging evidence of hypercholinergic activity in prodromal and symptomatic leucine-rich repeat kinase 2 PD may reflect neuronal cholinergic compensation versus a response to neuro-inflammation. Molecular imaging of the cholinergic system has led to many new insights in the etiology of dopamine non-responsive symptoms of PD (more "malignant" hypocholinergic disease phenotype) and is poised to guide and evaluate future cholinergic drug development in this disorder.
Collapse
|
130
|
Hampel H, Cavedo E, Vergallo A. Reply: Optimal use of cholinergic drugs in Alzheimer's disease. Brain 2018; 141:e69. [PMID: 30084876 DOI: 10.1093/brain/awy205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Harald Hampel
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | - Enrica Cavedo
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | - Andrea Vergallo
- AXA Research Fund and Sorbonne University Chair, Paris, France.,Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.,Brain and Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, Paris, France.,Institute of Memory and Alzheimer's Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l'hôpital, Paris, France
| | | |
Collapse
|
131
|
Gauthier S, Herrmann N, Rosa-Neto P. Optimal use of cholinergic drugs in Alzheimer's disease. Brain 2018; 141:e68. [PMID: 30084978 DOI: 10.1093/brain/awy204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Serge Gauthier
- Alzheimer's Disease Research Unit, McGill University Research Center for Studies in Aging, Douglas Research Center and CIUSS de l'Ouest de l'Ile, Montreal, Canada
| | - Nathan Herrmann
- Division of Geriatric Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Pedro Rosa-Neto
- Alzheimer's Disease Research Unit, McGill University Research Center for Studies in Aging, Douglas Research Center and CIUSS de l'Ouest de l'Ile, Montreal, Canada
| |
Collapse
|
132
|
Longitudinal Alzheimer’s Degeneration Reflects the Spatial Topography of Cholinergic Basal Forebrain Projections. Cell Rep 2018; 24:38-46. [DOI: 10.1016/j.celrep.2018.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/09/2018] [Accepted: 05/30/2018] [Indexed: 10/28/2022] Open
|
133
|
Jin H, Yue X, Liu H, Han J, Flores H, Su Y, Parsons SM, Perlmutter JS, Tu Z. Kinetic modeling of [ 18 F]VAT, a novel radioligand for positron emission tomography imaging vesicular acetylcholine transporter in non-human primate brain. J Neurochem 2018; 144:791-804. [PMID: 29315563 DOI: 10.1111/jnc.14291] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 12/16/2017] [Accepted: 12/19/2017] [Indexed: 01/06/2023]
Abstract
Molecular imaging of vesicular acetylcholine transporter (VAChT) in the brain provides an important cholinergic biomarker for the pathophysiology and treatment of dementias including Alzheimer's disease. In this study, kinetics modeling methods were applied and compared for quantifying regional brain uptake of the VAChT-specific positron emission tomography radiotracer, ((-)-(1-(-8-(2-fluoroethoxy)-3-hydroxy-1,2,3,4-tetrahydronaphthalen-2-yl)piperidin-4-yl)(4-fluorophenyl)-methanone) ([18 F]VAT) in macaques. Total volume distribution (VT ) estimates were compared for one-tissue compartment model (1TCM), two-tissue compartment model (2TCM), Logan graphic analysis (LoganAIF) and multiple linear analysis (MA1) with arterial blood input function using data from three macaques. Using the cerebellum-hemispheres as the reference region with data from seven macaques, three additional models were compared: reference tissue model (RTM), simplified RTM (SRTM), and Logan graphic analysis (LoganREF). Model selection criterion indicated that a) 2TCM and SRTM were the most appropriate kinetics models for [18 F]VAT; and b) SRTM was strongly correlated with 2TCM (Pearson's coefficients r > 0.93, p < 0.05). Test-retest studies demonstrated that [18 F]VAT has good reproducibility and reliability (TRV < 10%, ICC > 0.72). These studies demonstrate [18 F]VAT is a promising VAChT positron emission tomography tracer for quantitative assessment of VAChT levels in the brain of living subjects.
Collapse
Affiliation(s)
- Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junbin Han
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hubert Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yi Su
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
134
|
Bohnen NI, Grothe MJ, Ray NJ, Müller ML, Teipel SJ. Recent advances in cholinergic imaging and cognitive decline-Revisiting the cholinergic hypothesis of dementia. CURRENT GERIATRICS REPORTS 2018; 7:1-11. [PMID: 29503795 PMCID: PMC5831510 DOI: 10.1007/s13670-018-0234-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Although the cholinergic hypothesis of dementia provided a successful paradigm for the development of new drugs for dementia, this hypothesis has waned in popularity. Cholinergic brain imaging may provide novel insights into the viability of this hypothesis. RECENT FINDINGS Cholinergic receptor and forebrain volumetric studies suggest an important role of the cholinergic system in maintaining brain network integrity that may deteriorate with cognitive decline in Alzheimer disease (AD) and Lewy body disorders (LBD). Bidirectional changes in regional receptor expression may suggest the presence of compensatory responses to neurodegenerative injury. Cholinergic system changes are more complex in LBD because of additional subcortical degenerations compared to AD. Cholinergic-dopaminergic interactions affect attentional, verbal learning and executive functions, and impairments in these two transmitter systems may jointly increase the risk of dementia in Parkinson disease. SUMMARY The cholinergic hypothesis is evolving from a primary focus on memory toward expanded cognitive functions modulated by regionally more complex and interactive brain networks. Cholinergic network adaptation may serve as a novel research target in neurodegeneration.
Collapse
Affiliation(s)
- Nicolaas I. Bohnen
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Veterans Administration Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI, United States
| | - Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Nicola J. Ray
- Department of Psychology, Manchester Metropolitan University, Manchester, United Kingdom
| | - Martijn L.T.M. Müller
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Morris K. Udall Center of Excellence for Parkinson's Disease Research, University of Michigan, Ann Arbor, MI, United States
| | - Stefan J. Teipel
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| |
Collapse
|