1
|
Al-Jaf S, Soliman AY, El-Yazbi AF, Abd-Elrahman KS. Unveiling the Interplay: Neurovascular Coupling, Astrocytes and G Protein-Coupled Receptors in Alzheimer's Disease. ACS Pharmacol Transl Sci 2025; 8:271-285. [PMID: 39974631 PMCID: PMC11833731 DOI: 10.1021/acsptsci.4c00614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 02/21/2025]
Abstract
Astrocytes are a type of glial cell that are involved in actively modulating synaptic plasticity, neurotransmitter homeostasis, and neuroinflammatory responses. More importantly, they coordinate neuronal activity and cerebral blood flow (CBF) in what is known as neurovascular coupling (NVC). NVC is an essential mechanism that maintains the high energy demand the brain requires by supplying continuous and rapid supply of oxygen and nutrients through CBF. Impairment in NVC is one of the key events that triggers a spiral of occurrences that lead to the clinical advancement of Alzheimer's disease (AD). It is yet to be determined what the molecular manifestations of NVC impairment relate to; nonetheless, it is believed that alterations in G protein-coupled receptors (GPCRs) are responsible for exacerbating these effects. In this review, we summarize the current evidence supporting the involvement of GPCRs on astrocytes in NVC and the pathophysiology of AD. Additionally, we propose potential research directions to further elucidate the underlying mechanisms and evaluate the feasibility of targeting specific GPCRs as a therapeutic strategy to correct brain blood flow and memory impairments associated with AD.
Collapse
Affiliation(s)
- Sanarya Al-Jaf
- Department
of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian
Centre for Brain Health, The University
of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Alaa Y. Soliman
- Faculty
of Pharmacy and Research and Innovation Hub, Alamein International University, Alamein, Matrouh 51718, Egypt
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed F. El-Yazbi
- Faculty
of Pharmacy and Research and Innovation Hub, Alamein International University, Alamein, Matrouh 51718, Egypt
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Khaled S. Abd-Elrahman
- Department
of Anesthesiology, Pharmacology and Therapeutics, and Djavad Mowafaghian
Centre for Brain Health, The University
of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department
of Medical Sciences, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
2
|
Imbimbo C, Cotta Ramusino M, Leone S, Mazzacane F, De Franco V, Gatti A, Perini G, Costa A. Emerging Pharmacological Approaches for Psychosis and Agitation in Alzheimer's Disease. CNS Drugs 2025; 39:143-160. [PMID: 39623197 PMCID: PMC11769872 DOI: 10.1007/s40263-024-01133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 01/27/2025]
Abstract
Psychosis and agitation are among the most distressing neuropsychiatric symptoms (NPSs) of Alzheimer's disease (AD), linked to faster disease progression and earlier admission to nursing homes. While nonpharmacological treatments may alleviate mild behavioral symptoms, more severe syndromes often require pharmacological intervention. Brexpiprazole is the only medication approved for agitation in AD, although its limited clinical efficacy has raised criticism. No drugs have been approved for treating psychosis in AD, highlighting the critical need for new, effective, and safe treatments. Recent studies have elucidated part of the neurobiological basis of NPSs in the AD brain, offering insights for testing repurposed and novel drugs. We conducted a comprehensive nonsystematic literature review, aiming to provide a critical overview of both current treatments and emerging pharmacological interventions under clinical development for treating psychosis and agitation in AD. Additionally, we present strategies to optimize the clinical development of new drug candidates. We identify three promising compounds that are currently in phase 3 trials: xanomeline-trospium for AD psychosis, and dextromethorphan-bupropion and dexmedetomidine for agitation in AD. We propose that biomarkers linked to the neuropsychiatric traits of AD patients should be identified in dedicated studies and then included in phase 2 dose-range-finding studies with novel compounds to establish biological engagement. Furthermore, phase 3 placebo-controlled studies should be carried out in AD biomarker-confirmed subjects with narrower cognitive impairment ranges and precise NPS severity at screening. Alternative study designs, such as sequential phase approaches, may also be adopted.
Collapse
Affiliation(s)
- Camillo Imbimbo
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy.
| | - Silvia Leone
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Federico Mazzacane
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Valentino De Franco
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Alberto Gatti
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementias (CDCD), IRCCS Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
3
|
Koops EA, Dutta J, Hanseeuw BJ, Becker JA, Van Egroo M, Prokopiou PC, Price JC, Arnold SE, Sperling RA, Johnson KA, Jacobs HIL. Elevated locus coeruleus metabolism provides resilience against cognitive decline in preclinical Alzheimer's disease. Alzheimers Dement 2025; 21:e14385. [PMID: 39588792 PMCID: PMC11772725 DOI: 10.1002/alz.14385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Alterations in locus coeruleus' (LC) metabolic turnover are associated with Alzheimer's disease (AD)-pathology and cognitive impairment. However, the evolution of these changes across disease stages and their functional relevance remains unknown. METHODS We examined associations of [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) -derived LC metabolism with clinical diagnostic status, cerebrospinal fluid (CSF) -based AD biomarkers of AD pathology, and cognitive decline in Alzheimer's Disease Neuroimaging Initiative (ADNI) participants (n = 604). RESULTS FDG-PET-derived LC metabolism was elevated in the earliest preclinical stages and lower in later disease stages. Higher LC metabolism was associated with attenuated memory decline in preclinical stages, particularly in those with low CSF Aβ42, but not in AD patients with cognitive impairment. DISCUSSION Higher locus coeruleus [18F]-FDG-PET-derived signal in the early preclinical stages of AD can confer cognitive resilience and may reflect increased metabolic activity, whereas later stages are characterized by lower LC FDG-PET-derived signal, possibly due to neurodegeneration. HIGHLIGHTS LC FDG-PET signal is lower in Alzheimer's disease (AD) patients. LC FDG-PET signal is higher in the preclinical stage of AD. We observed less memory decline in those with higher LC FDG-PET signal. Higher LC FDG-PET signal conferred cognitive resilience in preclinical AD.
Collapse
Affiliation(s)
- Elouise A. Koops
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Joyita Dutta
- Department of Biomedical EngineeringUniversity of Massachusetts AmherstAmherstMassachusettsUSA
| | - Bernard J. Hanseeuw
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - J. Alex Becker
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Maxime Van Egroo
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Prokopis C. Prokopiou
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Julie C. Price
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Steven E. Arnold
- Alzheimer's Clinical and Translational Research Unit, Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Reisa A. Sperling
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Center for Alzheimer Research and Treatment, Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Keith A. Johnson
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Center for Alzheimer Research and Treatment, Department of NeurologyBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Heidi I. L. Jacobs
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Faculty of Health, Medicine and Life Sciences, Mental Health and Neuroscience Research Institute, Alzheimer Centre LimburgMaastricht UniversityMaastrichtMDThe Netherlands
| | | |
Collapse
|
4
|
Sighencea MG, Popescu RȘ, Trifu SC. From Fundamentals to Innovation in Alzheimer's Disease: Molecular Findings and Revolutionary Therapies. Int J Mol Sci 2024; 25:12311. [PMID: 39596378 PMCID: PMC11594972 DOI: 10.3390/ijms252212311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) is a global health concern and the leading cause of dementia in the elderly. The prevalence of this neurodegenerative condition is projected to increase concomitantly with increased life expectancy, resulting in a significant economic burden. With very few FDA-approved disease-modifying drugs available for AD, there is an urgent need to develop new compounds capable of impeding the progression of the disease. Given the unclear etiopathogenesis of AD, this review emphasizes the underlying mechanisms of this condition. It explores not only well-studied aspects, such as the accumulation of Aβ plaques and neurofibrillary tangles, but also novel areas, including glymphatic and lymphatic pathways, microbiota and the gut-brain axis, serotoninergic and autophagy alterations, vascular dysfunction, the metal hypothesis, the olfactory pathway, and oral health. Furthermore, the potential molecular targets arising from all these mechanisms have been reviewed, along with novel promising approaches such as nanoparticle-based therapy, neural stem cell transplantation, vaccines, and CRISPR-Cas9-mediated genome editing techniques. Taking into account the overlap of these various mechanisms, individual and combination therapies emerge as the future direction in the AD strategy.
Collapse
Affiliation(s)
| | - Ramona Ștefania Popescu
- Department of Infectious Diseases, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania;
| | - Simona Corina Trifu
- Department of Psychiatry, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
| |
Collapse
|
5
|
Li J, Liu Y, Yin C, Zeng Y, Mei Y. Structural and functional remodeling of neural networks in β-amyloid driven hippocampal hyperactivity. Ageing Res Rev 2024; 101:102468. [PMID: 39218080 DOI: 10.1016/j.arr.2024.102468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Early detection of Alzheimer's disease (AD) is essential for improving the patients outcomes and advancing our understanding of disease, allowing for timely intervention and treatment. However, accurate biomarkers are still lacking. Recent evidence indicates that hippocampal hyperexcitability precedes the diagnosis of AD decades ago, can predict cognitive decline. Thus, could hippocampal hyperactivity be a robust biomarker for early-AD, and what drives hippocampal hyperactivity in early-AD? these critical questions remain to be answered. Increasing clinical and experimental studies suggest that early hippocampal activation is closely associated with longitudinal β-amyloid (Aβ) accumulation, Aβ aggregates, in turn, enhances hippocampal activity. Therefore, in this narrative review, we discuss the role of Aβ-induced altered intrinsic neuronal properties as well as structural and functional remodeling of glutamatergic, GABAergic, cholinergic, noradrenergic, serotonergic circuits in hippocampal hyperactivity. In addition, we analyze the available therapies and trials that can potentially be used clinically to attenuate hippocampal hyperexcitability in AD. Overall, the present review sheds lights on the mechanism behind Aβ-induced hippocampal hyperactivity, and highlights that hippocampal hyperactivity could be a robust biomarker and therapeutic target in prodromal AD.
Collapse
Affiliation(s)
- Jinquan Li
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanjun Liu
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Chuhui Yin
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yan Zeng
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yufei Mei
- Hubei Clinical Research Center for Alzheimer's Disease, Brain Science and Advanced Technology Institute, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
6
|
Cummings JL, Brubaker M, Selzler KJ, Gonzalez ST, Patel M, Stahl SM. An overview of the pathophysiology of agitation in Alzheimer's dementia with a focus on neurotransmitters and circuits. CNS Spectr 2024:1-10. [PMID: 39438777 DOI: 10.1017/s1092852924000427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Alzheimer's dementia (AD) is a progressive, neurodegenerative disease often accompanied by neuropsychiatric symptoms that profoundly impact both patients and caregivers. Agitation is among the most prevalent and distressing of these symptoms and often requires treatment. Appropriate therapeutic interventions depend on understanding the biological basis of agitation and how it may be affected by treatment. This narrative review discusses a proposed pathophysiology of agitation in Alzheimer's dementia based on convergent evidence across research approaches. Available data indicate that agitation in Alzheimer's dementia is associated with an imbalance of activity between key prefrontal and subcortical brain regions. The monoamine neurotransmitter systems serve as key modulators of activity within these brain regions and circuits and are rendered abnormal in AD. Patients with AD who exhibited agitation symptoms during life have alterations in neurotransmitter nuclei and related systems when the brain is examined at autopsy. The authors present a model of agitation in Alzheimer's dementia in which noradrenergic hyperactivity along with serotonergic deficits and dysregulated striatal dopamine release contribute to agitated and aggressive behaviors.
Collapse
Affiliation(s)
- Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Nevada, USA
| | - Malaak Brubaker
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, New Jersey, USA
| | | | | | - Mehul Patel
- Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, New Jersey, USA
| | - Stephen M Stahl
- Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, California; Department of Psychiatry and Neurology, University of California, Riverside School of Medicine, Riverside, California, USA
| |
Collapse
|
7
|
Amada N, Sato S, Ishikawa D, Nakamura M, Suzuki M, Futamura T, Maeda K. Brexpiprazole: A new option in treating agitation in Alzheimer's dementia-Insights from transgenic mouse models. Neuropsychopharmacol Rep 2024; 44:557-568. [PMID: 38924384 PMCID: PMC11544443 DOI: 10.1002/npr2.12461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
AIM Brexpiprazole is the first FDA-approved treatment for agitation associated with dementia due to Alzheimer's disease. Agitation in Alzheimer's dementia (AAD) occurs in high prevalence and is a great burden for patients and caregivers. Efficacy, safety, and tolerability of brexpiprazole were demonstrated in the AAD clinical trials. To demonstrate the agitation-ameliorating effect of brexpiprazole in animals, we evaluated brexpiprazole in two AAD mouse models. METHODS The resident-intruder test was conducted in 5- to 6-month-old Tg2576 mice, given vehicle or brexpiprazole (0.01 or 0.03 mg/kg) orally 1 h before the test. Locomotor activity was measured in 6-month-old APPSL-Tg mice given vehicle or brexpiprazole (0.01 or 0.03 mg/kg) orally the evening before the start of locomotor measurement for 3 days. RESULTS In the resident-intruder test, Tg2576 mice showed significantly higher attack number and shorter latency to first attack compared to non-Tg mice. In the Tg mice, brexpiprazole treatment (0.03 mg/kg) significantly delayed the latency to first attack and showed a trend toward a decrease in attack number. APPSL-Tg mice (≧6 months old) showed significantly higher locomotion during dark period Phase II (Zeitgeber time [ZT] 16-20) and Phase III (ZT20-24) compared to non-Tg mice, correlating with the clinical observations of late afternoon agitation in Alzheimer's disease. Brexpiprazole treatment (0.01 and 0.03 mg/kg) significantly decreased hyperlocomotion during the Phase III in APPSL-Tg mice. CONCLUSION The suppression of attack behavior and the reduction of nocturnal hyperlocomotion in these Tg mice may be indicative of the therapeutic effect of brexpiprazole on AAD, as demonstrated in the clinical trials.
Collapse
Affiliation(s)
- Naoki Amada
- Otsuka Pharmaceutical Co., Ltd.TokushimaJapan
| | | | | | | | | | | | | |
Collapse
|
8
|
Plini ERG, Melnychuk MC, Andrews R, Boyle R, Whelan R, Spence JS, Chapman SB, Robertson IH, Dockree PM. Greater physical fitness ( VO 2 max ) in healthy older adults associated with increased integrity of the locus coeruleus-noradrenergic system. Acta Physiol (Oxf) 2024; 240:e14191. [PMID: 38895950 PMCID: PMC11250687 DOI: 10.1111/apha.14191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
AIM Physical activity (PA) is a key component for brain health and Reserve, and it is among the main dementia protective factors. However, the neurobiological mechanisms underpinning Reserve are not fully understood. In this regard, a noradrenergic (NA) theory of cognitive reserve (Robertson, 2013) has proposed that the upregulation of NA system might be a key factor for building reserve and resilience to neurodegeneration because of the neuroprotective role of NA across the brain. PA elicits an enhanced catecholamine response, in particular for NA. By increasing physical commitment, a greater amount of NA is synthetised in response to higher oxygen demand. More physically trained individuals show greater capabilities to carry oxygen resulting in greaterVo 2 max - a measure of oxygen uptake and physical fitness (PF). METHODS We hypothesized that greaterVo 2 max would be related to greater Locus Coeruleus (LC) MRI signal intensity. In a sample of 41 healthy subjects, we performed Voxel-Based Morphometry analyses, then repeated for the other neuromodulators as a control procedure (Serotonin, Dopamine and Acetylcholine). RESULTS As hypothesized, greaterVo 2 max related to greater LC signal intensity, and weaker associations emerged for the other neuromodulators. CONCLUSION This newly established link betweenVo 2 max and LC-NA system offers further understanding of the neurobiology underpinning Reserve in relationship to PA. While this study supports Robertson's theory proposing the upregulation of the NA system as a possible key factor building Reserve, it also provides ground for increasing LC-NA system resilience to neurodegeneration viaVo 2 max enhancement.
Collapse
Affiliation(s)
- Emanuele R G Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Michael C Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Ralph Andrews
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Robert Whelan
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Jeffrey S Spence
- Center for BrainHealth, The University of Texas at Dallas, Dallas, Texas, USA
| | - Sandra B Chapman
- Center for BrainHealth, The University of Texas at Dallas, Dallas, Texas, USA
| | - Ian H Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Paul M Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Engels-Domínguez N, Koops EA, Hsieh S, Wiklund EE, Schultz AP, Riphagen JM, Prokopiou PC, Hanseeuw BJ, Rentz DM, Sperling RA, Johnson KA, Jacobs HIL. Lower in vivo locus coeruleus integrity is associated with lower cortical thickness in older individuals with elevated Alzheimer's pathology: a cohort study. Alzheimers Res Ther 2024; 16:129. [PMID: 38886798 PMCID: PMC11181564 DOI: 10.1186/s13195-024-01500-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Autopsy work indicates that the widely-projecting noradrenergic pontine locus coeruleus (LC) is among the earliest regions to accumulate hyperphosphorylated tau, a neuropathological Alzheimer's disease (AD) hallmark. This early tau deposition is accompanied by a reduced density of LC projections and a reduction of norepinephrine's neuroprotective effects, potentially compromising the neuronal integrity of LC's cortical targets. Previous studies suggest that lower magnetic resonance imaging (MRI)-derived LC integrity may signal cortical tissue degeneration in cognitively healthy, older individuals. However, whether these observations are driven by underlying AD pathology remains unknown. To that end, we examined potential effect modifications by cortical beta-amyloid and tau pathology on the association between in vivo LC integrity, as quantified by LC MRI signal intensity, and cortical neurodegeneration, as indexed by cortical thickness. METHODS A total of 165 older individuals (74.24 ± 9.72 years, ~ 60% female, 10% cognitively impaired) underwent whole-brain and dedicated LC 3T-MRI, Pittsburgh Compound-B (PiB, beta-amyloid) and Flortaucipir (FTP, tau) positron emission tomography. Linear regression analyses with bootstrapped standard errors (n = 2000) assessed associations between bilateral cortical thickness and i) LC MRI signal intensity and, ii) LC MRI signal intensity interacted with cortical FTP or PiB (i.e., EC FTP, IT FTP, neocortical PiB) in the entire sample and a low beta-amyloid subsample. RESULTS Across the entire sample, we found a direct effect, where lower LC MRI signal intensity was associated with lower mediolateral temporal cortical thickness. Evaluation of potential effect modifications by FTP or PiB revealed that lower LC MRI signal intensity was related to lower cortical thickness, particularly in individuals with elevated (EC, IT) FTP or (neocortical) PiB. The latter result was present starting from subthreshold PiB values. In low PiB individuals, lower LC MRI signal intensity was related to lower EC cortical thickness in the context of elevated EC FTP. CONCLUSIONS Our findings suggest that LC-related cortical neurodegeneration patterns in older individuals correspond to regions representing early Braak stages and may reflect a combination of LC projection density loss and emergence of cortical AD pathology. This provides a novel understanding that LC-related cortical neurodegeneration may signal downstream consequences of AD-related pathology, rather than being exclusively a result of aging.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Elouise A Koops
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Stephanie Hsieh
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Emma E Wiklund
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Aaron P Schultz
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Prokopis C Prokopiou
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA
| | - Bernard J Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- The Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02129, USA.
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Maturana-Quijada P, Chavarría-Elizondo P, Del Cerro I, Martínez-Zalacaín I, Juaneda-Seguí A, Guinea-Izquierdo A, Gascón-Bayarri J, Reñé R, Urretavizcaya M, Menchón JM, Ferrer I, Soria V, Soriano-Mas C. Effective connectivity of the locus coeruleus in patients with late-life Major Depressive Disorder or mild cognitive impairment. SPANISH JOURNAL OF PSYCHIATRY AND MENTAL HEALTH 2024:S2950-2853(24)00015-2. [PMID: 38453029 DOI: 10.1016/j.sjpmh.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION We compared effective connectivity from the locus coeruleus (LC) during the resting-state in patients with late-life Major Depressive Disorder (MDD), individuals with amnestic Mild Cognitive Impairment (aMCI), and Healthy Controls (HCs). PARTICIPANTS 23 patients with late-life MDD, 22 patients with aMCI, and 28 HCs. MATERIAL AND METHODS Participants were assessed in two time-points, 2 years apart. They underwent a resting-state functional magnetic resonance imaging and a high-resolution anatomical acquisition, as well as clinical assessments. Functional imaging data were analyzed with dynamic causal modeling, and parametric empirical Bayes model was used to map effective connectivity between 7 distinct nodes: 4 from the locus coeruleus and 3 regions displaying gray matter decreases during the two-year follow-up period. RESULTS Longitudinal analysis of structural data identified three clusters of larger over-time gray matter volume reduction in patients (MDD+aMCI vs. HCs): the right precuneus, and the visual association and parahippocampal cortices. aMCI patients showed decreased effective connectivity from the left rostral to caudal portions of the LC, while connectivity from the left rostral LC to the parahippocampal cortex increased. In MDD, there was a decline in effective connectivity across LC caudal seeds, and increased connectivity from the left rostral to the left caudal LC seed over time. Connectivity alterations with cortical regions involved cross-hemisphere increases and same-hemisphere decreases. CONCLUSIONS Our discoveries provide insight into the dynamic changes in effective connectivity in individuals with late-life MDD and aMCI, also shedding light on the mechanisms potentially contributing to the onset of neurodegenerative disorders.
Collapse
Affiliation(s)
- Pablo Maturana-Quijada
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | - Pamela Chavarría-Elizondo
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain; Network Center for Biomedical Research on Mental Health (CIBERSAM), Carlos III Health Institute (ISCIII), Barcelona, Spain
| | - Inés Del Cerro
- Department of Psychology, Medical School, Catholic University of Murcia, Murcia, Spain
| | - Ignacio Martínez-Zalacaín
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Radiology Department, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Asier Juaneda-Seguí
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Andrés Guinea-Izquierdo
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Jordi Gascón-Bayarri
- Dementia Diagnostic and Treatment Unit, Department of Neurology, Bellvitge University Hospital, Barcelona, Spain
| | - Ramón Reñé
- Dementia Diagnostic and Treatment Unit, Department of Neurology, Bellvitge University Hospital, Barcelona, Spain
| | - Mikel Urretavizcaya
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain; Network Center for Biomedical Research on Mental Health (CIBERSAM), Carlos III Health Institute (ISCIII), Barcelona, Spain
| | - José M Menchón
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain; Network Center for Biomedical Research on Mental Health (CIBERSAM), Carlos III Health Institute (ISCIII), Barcelona, Spain
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Bellvitge Biomedical Research Institute-IDIBELL, Department of Pathologic Anatomy, Bellvitge University Hospital, Barcelona, Spain; Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Virginia Soria
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Network Center for Biomedical Research on Mental Health (CIBERSAM), Carlos III Health Institute (ISCIII), Barcelona, Spain; Department of Mental Health, Parc Taulí Hospital Universitari, Sabadell, Barcelona, Spain
| | - Carles Soriano-Mas
- Psychiatry and Mental Health Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain; Network Center for Biomedical Research on Mental Health (CIBERSAM), Carlos III Health Institute (ISCIII), Barcelona, Spain; Department of Social Psychology and Quantitative Psychology, Institute of Neurosciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
11
|
Lin CP, Frigerio I, Bol JGJM, Bouwman MMA, Wesseling AJ, Dahl MJ, Rozemuller AJM, van der Werf YD, Pouwels PJW, van de Berg WDJ, Jonkman LE. Microstructural integrity of the locus coeruleus and its tracts reflect noradrenergic degeneration in Alzheimer's disease and Parkinson's disease. Transl Neurodegener 2024; 13:9. [PMID: 38336865 PMCID: PMC10854137 DOI: 10.1186/s40035-024-00400-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Degeneration of the locus coeruleus (LC) noradrenergic system contributes to clinical symptoms in Alzheimer's disease (AD) and Parkinson's disease (PD). Diffusion magnetic resonance imaging (MRI) has the potential to evaluate the integrity of the LC noradrenergic system. The aim of the current study was to determine whether the diffusion MRI-measured integrity of the LC and its tracts are sensitive to noradrenergic degeneration in AD and PD. METHODS Post-mortem in situ T1-weighted and multi-shell diffusion MRI was performed for 9 AD, 14 PD, and 8 control brain donors. Fractional anisotropy (FA) and mean diffusivity were derived from the LC, and from tracts between the LC and the anterior cingulate cortex, the dorsolateral prefrontal cortex (DLPFC), the primary motor cortex (M1) or the hippocampus. Brain tissue sections of the LC and cortical regions were obtained and immunostained for dopamine-beta hydroxylase (DBH) to quantify noradrenergic cell density and fiber load. Group comparisons and correlations between outcome measures were performed using linear regression and partial correlations. RESULTS The AD and PD cases showed loss of LC noradrenergic cells and fibers. In the cortex, the AD cases showed increased DBH + immunoreactivity in the DLPFC compared to PD cases and controls, while PD cases showed reduced DBH + immunoreactivity in the M1 compared to controls. Higher FA within the LC was found for AD, which was correlated with loss of noradrenergic cells and fibers in the LC. Increased FA of the LC-DLPFC tract was correlated with LC noradrenergic fiber loss in the combined AD and control group, whereas the increased FA of the LC-M1 tract was correlated with LC noradrenergic neuronal loss in the combined PD and control group. The tract alterations were not correlated with cortical DBH + immunoreactivity. CONCLUSIONS In AD and PD, the diffusion MRI-detected alterations within the LC and its tracts to the DLPFC and the M1 were associated with local noradrenergic neuronal loss within the LC, rather than noradrenergic changes in the cortex.
Collapse
Affiliation(s)
- Chen-Pei Lin
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands.
| | - Irene Frigerio
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - John G J M Bol
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Maud M A Bouwman
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - Alex J Wesseling
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
| | - Martin J Dahl
- Center for Lifespan Psychology, Max Planck Institute for Human Development, 14195, Berlin, Germany
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Annemieke J M Rozemuller
- Amsterdam UMC, Department of Pathology, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Ysbrand D van der Werf
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Compulsivity, Impulsivity and Attention Program, Amsterdam, The Netherlands
| | - Petra J W Pouwels
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Radiology and Nuclear Medicine, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Laura E Jonkman
- Amsterdam UMC, Department of Anatomy and Neurosciences, Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Brain imaging, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Quattrini G, Pini L, Boscolo Galazzo I, Jelescu IO, Jovicich J, Manenti R, Frisoni GB, Marizzoni M, Pizzini FB, Pievani M. Microstructural alterations in the locus coeruleus-entorhinal cortex pathway in Alzheimer's disease and frontotemporal dementia. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12513. [PMID: 38213948 PMCID: PMC10781651 DOI: 10.1002/dad2.12513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/04/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024]
Abstract
INTRODUCTION We investigated in vivo the microstructural integrity of the pathway connecting the locus coeruleus to the transentorhinal cortex (LC-TEC) in patients with Alzheimer's disease (AD) and frontotemporal dementia (FTD). METHODS Diffusion-weighted MRI scans were collected for 21 AD, 20 behavioral variants of FTD (bvFTD), and 20 controls. Fractional anisotropy (FA), mean, axial, and radial diffusivities (MD, AxD, RD) were computed in the LC-TEC pathway using a normative atlas. Atrophy was assessed using cortical thickness and correlated with microstructural measures. RESULTS We found (i) higher RD in AD than controls; (ii) higher MD, RD, and AxD, and lower FA in bvFTD than controls and AD; and (iii) a negative association between LC-TEC MD, RD, and AxD, and entorhinal cortex (EC) thickness in bvFTD (all p < 0.050). DISCUSSION LC-TEC microstructural alterations are more pronounced in bvFTD than AD, possibly reflecting neurodegeneration secondary to EC atrophy. Highlights Microstructural integrity of LC-TEC pathway is understudied in AD and bvFTD.LC-TEC microstructural alterations are present in both AD and bvFTD.Greater LC-TEC microstructural alterations in bvFTD than AD.LC-TEC microstructural alterations in bvFTD are associated to EC neurodegeneration.
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
| | - Lorenzo Pini
- Padova Neuroscience CenterUniversity of PadovaPadovaItaly
| | | | - Ileana O. Jelescu
- Department of RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Jorge Jovicich
- Center of Mind/Brain SciencesUniversity of TrentoRoveretoItaly
| | - Rosa Manenti
- Neuropsychology UnitIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Giovanni B. Frisoni
- Memory Center and LANVIE ‐ Laboratory of Neuroimaging of AgingUniversity Hospitals and University of GenevaGenevaSwitzerland
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Laboratory of Biological PsychiatryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Francesca B. Pizzini
- Department of Engineering for Innovation MedicineUniversity of VeronaVeronaItaly
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE)IRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| |
Collapse
|
13
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
14
|
Jin M, Wei Z, Ramalingam N, Xiao M, Xu A, Yu X, Song Q, Liu W, Zhao J, Zhang D, Selkoe DJ, Li S. Activation of β 2-adrenergic receptors prevents AD-type synaptotoxicity via epigenetic mechanisms. Mol Psychiatry 2023; 28:4877-4888. [PMID: 37365243 DOI: 10.1038/s41380-023-02145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
We previously reported that prolonged exposure to an enriched environment (EE) enhances hippocampal synaptic plasticity, with one of the significant mechanistic pathways being activation of β2-adrenergic receptor (β2-AR) signaling, thereby mitigating the synaptotoxic effects of soluble oligomers of amyloid β-protein (oAβ). However, the detailed mechanism remained elusive. In this work, we recorded field excitatory postsynaptic potentials (fEPSP) in the CA1 region of mouse hippocampal slices treated with or without toxic Aβ-species. We found that pharmacological activation of β2-AR, but not β1-AR, selectively mimicked the effects of EE in enhancing LTP and preventing oAβ-induced synaptic dysfunction. Mechanistic analyses showed that certain histone deacetylase (HDAC) inhibitors mimicked the benefits of EE, but this was not seen in β2-AR knockout mice, suggesting that activating β2-AR prevents oAβ-mediated synaptic dysfunction via changes in histone acetylation. EE or activation of β-ARs each decreased HDAC2, whereas Aβ oligomers increased HDAC2 levels in the hippocampus. Further, oAβ-induced inflammatory effects and neurite degeneration were prevented by either β2-AR agonists or certain specific HDAC inhibitors. These preclinical results suggest that activation of β2-AR is a novel potential therapeutic strategy to mitigate oAβ-mediated features of AD.
Collapse
Affiliation(s)
- Ming Jin
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhiyun Wei
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Nagendran Ramalingam
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Meng Xiao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, 518172, China
| | - Anqi Xu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Xiaohan Yu
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Qingyang Song
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
| | - Wen Liu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jianhua Zhao
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Henan Key Laboratory of Neurorestoratology, Xinxiang, Henan, 453100, China
| | - Dainan Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, 453100, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
15
|
Prinzi C, Kostenko A, de Leo G, Gulino R, Leanza G, Caccamo A. Selective Noradrenaline Depletion in the Neocortex and Hippocampus Induces Working Memory Deficits and Regional Occurrence of Pathological Proteins. BIOLOGY 2023; 12:1264. [PMID: 37759663 PMCID: PMC10526041 DOI: 10.3390/biology12091264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023]
Abstract
Noradrenaline (NA) depletion occurs in Alzheimer's disease (AD); however, its relationship with the pathological expression of Tau and transactive response DNA-binding protein 43 (TDP-43), two major hallmarks of AD, remains elusive. Here, increasing doses of a selective noradrenergic immunotoxin were injected into developing rats to generate a model of mild or severe NA loss. At about 12 weeks post-lesion, dose-dependent working memory deficits were detected in these animals, associated with a marked increase in cortical and hippocampal levels of TDP-43 phosphorylated at Ser 409/410 and Tau phosphorylated at Thr 217. Notably, the total levels of both proteins were largely unaffected, suggesting a direct relationship between neocortical/hippocampal NA depletion and the phosphorylation of pathological Tau and TDP-43 proteins. As pTD43 is present in 23% of AD cases and pTau Thr217 has been detected in patients with mild cognitive impairment that eventually would develop into AD, improvement of noradrenergic function in AD might represent a viable therapeutic approach with disease-modifying potential.
Collapse
Affiliation(s)
- Chiara Prinzi
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
| | - Anna Kostenko
- B.R.A.I.N. (Basic Research and Integrative Neuroscience) Laboratory for Neurogenesis and Repair, Department of Life Sciences, University of Trieste, 34100 Trieste, Italy;
| | - Gioacchino de Leo
- SISSA, Scuola Internazionale Superiore di Studi Avanzati, 34136 Triste, Italy;
| | - Rosario Gulino
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95125 Catania, Italy;
| | - Giampiero Leanza
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
- Molecular Preclinical and Translational Imaging Research Centre-IMPRonTE, University of Catania, 95125 Catania, Italy
| | - Antonella Caccamo
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy;
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
16
|
Plini ERG, Melnychuk MC, Andrews R, Boyle R, Whelan R, Spence JS, Chapman SB, Robertson IH, Dockree PM. Greater physical fitness (Vo2Max) in healthy older adults associated with increased integrity of the Locus Coeruleus-Noradrenergic system. RESEARCH SQUARE 2023:rs.3.rs-2556690. [PMID: 36798156 PMCID: PMC9934752 DOI: 10.21203/rs.3.rs-2556690/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Physical activity (PA) is a key component for brain health and Reserve, and it is among the main dementia protective factors. However, the neurobiological mechanisms underpinning Reserve are not fully understood. In this regard, a noradrenergic (NA) theory of cognitive reserve (Robertson, 2013) has proposed that the upregulation of NA system might be a key factor for building reserve and resilience to neurodegeneration because of the neuroprotective role of NA across the brain. PA elicits an enhanced catecholamine response, in particular for NA. By increasing physical commitment, a greater amount of NA is synthetised in response to higher oxygen demand. More physically trained individuals show greater capabilities to carry oxygen resulting in greater Vo2max - a measure of oxygen uptake and physical fitness (PF). In the current study, we hypothesised that greater Vo2 max would be related to greater Locus Coeruleus (LC) MRI signal intensity. As hypothesised, greater Vo2max related to greater LC signal intensity across 41 healthy adults (age range 60-72). As a control procedure, in which these analyses were repeated for the other neuromodulators' seeds (for Serotonin, Dopamine and Acetylcholine), weaker associations emerged. This newly established link between Vo2max and LC-NA system offers further understanding of the neurobiology underpinning Reserve in relationship to PA. While this study supports Robertson's theory proposing the upregulation of the noradrenergic system as a possible key factor building Reserve, it also provide grounds for increasing LC-NA system resilience to neurodegeneration via Vo2max enhancement.
Collapse
Affiliation(s)
- Emanuele RG Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Michael C Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Ralph Andrews
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown MA, USA
| | - Robert Whelan
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Jeffrey S. Spence
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Sandra B. Chapman
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Ian H Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Building 149, Charlestown MA, USA
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| | - Paul M Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland
| |
Collapse
|
17
|
Zhou H, Wang K, Xu Z, Liu D, Wang Y, Guo M. Chronic unpredictable stress induces depression/anxiety-related behaviors and alterations of hippocampal monoamine receptor mRNA expression in female mice at different ages. Heliyon 2023; 9:e18369. [PMID: 37539192 PMCID: PMC10393760 DOI: 10.1016/j.heliyon.2023.e18369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 08/05/2023] Open
Abstract
Depression and anxiety are the most common mental health disorders. Though they affect people at any age and occur more often in females, the pathophysiological changes under these conditions are less investigated. In the present study, we examined the effects of age and stress on depression- and anxiety-related behaviors in female mice. Saccharin preference and the open field test were carried out before and after chronic unpredictable stress in 4-, 14- and 25-month-old female mice. After behavioral tests, mRNA levels of monoamine receptors in the hippocampus were measured by real-time RT-PCR. Chronic unpredictable stress decreased saccharin preference in 4-, 14- and 25-month-old mice and the time spent in the center in the open field test in 25-month-old mice. For monoamine receptors, analysis of variance revealed significant effects of age on mRNA levels of Htr1a, Htr2a, Htr6, Adra1a, Adrb2, and Adrb3, significant effects of stress on mRNA levels of Htr4, Adra2c, Adrb1, and Adrb2, and interactions of age × stress on mRNA levels of Htr1a, Htr5b, Adra1d, Adra2a, Adra2c, and Adrb1. Chronic unpredictable stress decreased mRNA levels of Htr4, Htr5b, Adra2c, and Adrb1 in 4-month-old female mice. Correlations were observed between saccharin preference and mRNA levels of Htr4, Htr5b, Htr6, Adra1d, Adra2a, and Adra2c in 4-month-old mice and between the time spent in the center in the open field test and mRNA levels of Htr1b in 4-month-old mice, Htr3a, Htr7, and Adrb2 in 14-month-old mice, and Drd2 in 4- and 14-month-old mice. Our findings support that stress induces depression- and anxiety-related behaviors and the expression of hippocampal monoamine receptors in an age-dependent manner in female mice.
Collapse
Affiliation(s)
- Han Zhou
- Department of Psychology, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Kaixin Wang
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Zhicheng Xu
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Dunjiang Liu
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Yameng Wang
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| | - Ming Guo
- Department of Psychology, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
- Medical Research Center, Binzhou Medical University Hospital, The First School of Clinical Medicine of Binzhou Medical University, Binzhou, Shandong, 256603, China
| |
Collapse
|
18
|
Papay RS, Stauffer SR, Perez DM. A PAM of the α 1A-Adrenergic receptor rescues biomarker, long-term potentiation, and cognitive deficits in Alzheimer's disease mouse models without effects on blood pressure. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100160. [PMID: 37448695 PMCID: PMC10336260 DOI: 10.1016/j.crphar.2023.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/30/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
α1-Adrenergic Receptors (ARs) regulate the sympathetic nervous system by the binding of norepinephrine (NE) and epinephrine (Epi) through different subtypes (α1A, α1B, α1D). α1A-AR activation is hypothesized to be memory forming and cognitive enhancing but drug development has been stagnant due to unwanted side effects on blood pressure. We recently reported the pharmacological characterization of the first positive allosteric modulator (PAM) for the α1A-AR with predictive pro-cognitive and memory properties. In this report, we now demonstrate the in vivo characteristics of Compound 3 (Cmpd-3) in two genetically-different Alzheimer's Disease (AD) mouse models. Drug metabolism and pharmacokinetic studies indicate sufficient brain penetrance and rapid uptake into the brain with low to moderate clearance, and a favorable inhibition profile against the major cytochrome p450 enzymes. Oral administration of Cmpd-3 (3-9 mg/kg QD) can fully rescue long-term potentiation defects and AD biomarker profile (amyloid β-40, 42) within 3 months of dosing to levels that were non-significant from WT controls and which outperformed donepezil (1 mg/kg QD). There were also significant effects on paired pulse facilitation and cognitive behavior. Long-term and high-dose in vivo studies with Cmpd-3 revealed no effects on blood pressure. Our results suggest that Cmpd-3 can maintain lasting therapeutic levels and efficacy with disease modifying effects with a once per day dosing regimen in AD mouse models with no observed side effects.
Collapse
Affiliation(s)
- Robert S. Papay
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| | - Shaun R. Stauffer
- Center of Therapeutics Discovery, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, Ohio, 44195, USA
| | - Dianne M. Perez
- The Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave, Cleveland, OH, 44195, USA
| |
Collapse
|
19
|
Orlando IF, Shine JM, Robbins TW, Rowe JB, O'Callaghan C. Noradrenergic and cholinergic systems take centre stage in neuropsychiatric diseases of ageing. Neurosci Biobehav Rev 2023; 149:105167. [PMID: 37054802 DOI: 10.1016/j.neubiorev.2023.105167] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/28/2023] [Accepted: 03/28/2023] [Indexed: 04/15/2023]
Abstract
Noradrenergic and cholinergic systems are among the most vulnerable brain systems in neuropsychiatric diseases of ageing, including Alzheimer's disease, Parkinson's disease, Lewy body dementia, and progressive supranuclear palsy. As these systems fail, they contribute directly to many of the characteristic cognitive and psychiatric symptoms. However, their contribution to symptoms is not sufficiently understood, and pharmacological interventions targeting noradrenergic and cholinergic systems have met with mixed success. Part of the challenge is the complex neurobiology of these systems, operating across multiple timescales, and with non-linear changes across the adult lifespan and disease course. We address these challenges in a detailed review of the noradrenergic and cholinergic systems, outlining their roles in cognition and behaviour, and how they influence neuropsychiatric symptoms in disease. By bridging across levels of analysis, we highlight opportunities for improving drug therapies and for pursuing personalised medicine strategies.
Collapse
Affiliation(s)
- Isabella F Orlando
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia
| | - James M Shine
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia
| | - Trevor W Robbins
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, CB2 3EB, United Kingdom
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, CB2 0SZ, United Kingdom
| | - Claire O'Callaghan
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Australia.
| |
Collapse
|
20
|
Kelberman MA, Rorabaugh JM, Anderson CR, Marriott A, DePuy SD, Rasmussen K, McCann KE, Weiss JM, Weinshenker D. Age-dependent dysregulation of locus coeruleus firing in a transgenic rat model of Alzheimer's disease. Neurobiol Aging 2023; 125:98-108. [PMID: 36889122 PMCID: PMC10038926 DOI: 10.1016/j.neurobiolaging.2023.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Hyperphosphorylated tau in the locus coeruleus (LC) is ubiquitous in prodromal Alzheimer's disease (AD), and LC neurons degenerate as AD progresses. Hyperphosphorylated tau alters firing rates in other brain regions, but its effects on LC neurons are unknown. We assessed single unit LC activity in anesthetized wild-type (WT) and TgF344-AD rats at 6 months, which represents a prodromal stage when LC neurons are the only cells containing hyperphosphorylated tau in TgF344-AD animals, and at 15 months when amyloid-β (Aβ) and tau pathology are both abundant in the forebrain. At baseline, LC neurons from TgF344-AD rats were hypoactive at both ages compared to WT littermates but showed elevated spontaneous bursting properties. Differences in footshock-evoked LC firing depended on age, with 6-month TgF344-AD rats demonstrating aspects of hyperactivity, and 15-month transgenic rats showing hypoactivity. Early LC hyperactivity is consistent with appearance of prodromal neuropsychiatric symptoms and is followed by LC hypoactivity which contributes to cognitive impairment. These results support further investigation into disease stage-dependent noradrenergic interventions for AD.
Collapse
Affiliation(s)
| | | | | | - Alexia Marriott
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | | | | | | | - Jay M Weiss
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
21
|
Motta C, Assogna M, Bonomi CG, Di Lorenzo F, Nuccetelli M, Mercuri NB, Koch G, Martorana A. Interplay between the catecholaminergic enzymatic axis and neurodegeneration/neuroinflammation processes in the Alzheimer's disease continuum. Eur J Neurol 2023; 30:839-848. [PMID: 36692274 DOI: 10.1111/ene.15691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE The locus coeruleus (LC) provides dopamine/noradrenaline (DA/NA) innervation throughout the brain and undergoes early degeneration in Alzheimer's disease (AD). We evaluated catecholaminergic enzyme levels in the cerebrospinal fluid (CSF) of a group of patients biologically defined as within the AD continuum (ADc) and explored their relationship with AD biomarkers and cytokine/growth factor levels to investigate their interplay with neurodegenerative and neuroinflammatory processes. METHODS The CSF concentration of DA transporter (DAT), tyrosine-hydroxylase (TH), DOPA-decarboxylase (DDC), and dopamine-β-hydroxylase (DβH), as well as cytokine/growth factor levels, were analyzed in 41 ADc patients stratified according to CSF beta-amyloid (Aβ)1-42 (A) and p-tau (T) in AD pathological changes (A+ T-) and AD (A+ T+) subgroups, as well as in 15 control subjects (A- T-). RESULTS The ADc group had lower CSF levels of DAT and TH but increased DβH levels to compensate for NA synthesis. DDC levels were higher in the A+ T+ subgroup but comparable with controls in the A+ T- subgroup, probably because the DA system is resilient to the degeneration of LC neurons in the absence of tau pathology. Adjusting for age, sex, APOE genotype, and cognitive status, a significant association was found between TH and Aβ1-42 (R2 = 0.25) and between DDC and p-tau (R2 = 0.33). Finally, TH correlated with interleukin (IL)-10 levels (p = 0.0008) and DβH with IL-1β (p = 0.03), IL-4 (p = 0.02), granulocyte colony-stimulating factor (p = 0.007), and IL-17 (p = 0.01). CONCLUSIONS Taken together, these findings suggest that catecholaminergic enzymes, functional markers of the catecholaminergic system, are closely linked to the neurodegenerative and neuroinflammatory processes in AD pathology.
Collapse
Affiliation(s)
- Caterina Motta
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Martina Assogna
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Chiara Giuseppina Bonomi
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Di Lorenzo
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giacomo Koch
- Non-Invasive Brain Stimulation Unit/Department of Behavioral and Clinical Neurology, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Alessandro Martorana
- UOSD Centro Demenze, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
22
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Primary prevention trials have shifted their focus to the earliest stages of Alzheimer's disease (AD). Autopsy data indicates that the neuromodulatory subcortical systems' (NSS) nuclei are specifically vulnerable to initial tau pathology, indicating that these nuclei hold great promise for early detection of AD in the context of the aging brain. The increasing availability of new imaging methods, ultra-high field scanners, new radioligands, and routine deep brain stimulation implants has led to a growing number of NSS neuroimaging studies on aging and neurodegeneration. Here, we review findings of current state-of-the-art imaging studies assessing the structure, function, and molecular changes of these nuclei during aging and AD. Furthermore, we identify the challenges associated with these imaging methods, important pathophysiologic gaps to fill for the AD NSS neuroimaging field, and provide future directions to improve our assessment, understanding, and clinical use of in vivo imaging of the NSS.
Collapse
Affiliation(s)
- Nina Engels-Domínguez
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Elouise A Koops
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prokopis C Prokopiou
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Maxime Van Egroo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands
| | - Christoph Schneider
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joost M Riphagen
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tarun Singhal
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi I L Jacobs
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
23
|
Portela Moreira I, Henriques T, Vieira-Coelho MA, Guimarães J. Dysfunction of norepinephrine and its metabolites in Alzheimer's dementia - A review with meta-analysis. Ageing Res Rev 2023; 83:101784. [PMID: 36368648 DOI: 10.1016/j.arr.2022.101784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Some studies point locus coeruleus cell loss, the central nervous system main source of norepinephrine, to be one of the earliest neuropathological events of Alzheimer's disease (AD). However, there are conflicting reports regarding the level of norepinephrine and its metabolites (3-Methoxy-4-hydroxyphenylglycol (MHPG), 3,5-dihydroxyphenylglycine (DHPG) and 3,4 -dihydroxyphenylglycolaldehyde (DOPEGAL)) in AD patients. Uncover these alterations may be a key factor for understanding cognitive deficits and AD pathology. We review the literature that compare norepinephrine and its metabolites between AD patients and non-demented controls. A meta-analysis did not reveal significant statistical differences, but there was a trend towards a lower level of norepinephrine of AD, with almost no difference in MHPG in the cerebrospinal fluid. Regarding MHPG in plasma, DHPG and DOPEGAL we only performed a qualitative analyse due to the small or absent number of studies. These findings point to a decrease in norepinephrine, what is in line with locus coeluleus cell loss in AD. The absence of statistical difference and an equal level of MHGP could indicate a compensatory mechanism.
Collapse
Affiliation(s)
- Isabel Portela Moreira
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Hospital Privado de Gaia do Grupo Trofa Saúde, Vila Nova de Gaia, Portugal.
| | - Teresa Henriques
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine of Porto University, Porto, Portugal; Centre for Health Technology Services Research (CINTESIS), Faculty of Medicine of Porto University, Porto, Portugal
| | - Maria Augusta Vieira-Coelho
- Department of Biomedicine, Faculty of Medicine, Faculty of Medicine of Porto University, Porto, Portugal; Department of Psychiatry and Mental Health, University Hospital Center of São João, Porto, Portugal
| | - Joana Guimarães
- Clinical Neurosciences and Mental Health Department, Faculty of Medicine of Porto University, Porto, Portugal; Neurology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
24
|
Iannitelli AF, Kelberman MA, Lustberg DJ, Korukonda A, McCann KE, Mulvey B, Segal A, Liles LC, Sloan SA, Dougherty JD, Weinshenker D. The Neurotoxin DSP-4 Dysregulates the Locus Coeruleus-Norepinephrine System and Recapitulates Molecular and Behavioral Aspects of Prodromal Neurodegenerative Disease. eNeuro 2023; 10:ENEURO.0483-22.2022. [PMID: 36635251 PMCID: PMC9829100 DOI: 10.1523/eneuro.0483-22.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
The noradrenergic locus coeruleus (LC) is among the earliest sites of tau and α-synuclein pathology in Alzheimer's disease (AD) and Parkinson's disease (PD), respectively. The onset of these pathologies coincides with loss of noradrenergic fibers in LC target regions and the emergence of prodromal symptoms including sleep disturbances and anxiety. Paradoxically, these prodromal symptoms are indicative of a noradrenergic hyperactivity phenotype, rather than the predicted loss of norepinephrine (NE) transmission following LC damage, suggesting the engagement of complex compensatory mechanisms. Because current therapeutic efforts are targeting early disease, interest in the LC has grown, and it is critical to identify the links between pathology and dysfunction. We employed the LC-specific neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4), which preferentially damages LC axons, to model early changes in the LC-NE system pertinent to AD and PD in male and female mice. DSP-4 (two doses of 50 mg/kg, one week apart) induced LC axon degeneration, triggered neuroinflammation and oxidative stress, and reduced tissue NE levels. There was no LC cell death or changes to LC firing, but transcriptomics revealed reduced expression of genes that define noradrenergic identity and other changes relevant to neurodegenerative disease. Despite the dramatic loss of LC fibers, NE turnover and signaling were elevated in terminal regions and were associated with anxiogenic phenotypes in multiple behavioral tests. These results represent a comprehensive analysis of how the LC-NE system responds to axon/terminal damage reminiscent of early AD and PD at the molecular, cellular, systems, and behavioral levels, and provides potential mechanisms underlying prodromal neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Alexa F Iannitelli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Michael A Kelberman
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Daniel J Lustberg
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Anu Korukonda
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Katharine E McCann
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Bernard Mulvey
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Arielle Segal
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - L Cameron Liles
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
25
|
Phillips JR, Matar E, Ehgoetz Martens KA, Moustafa AA, Halliday GM, Lewis SJG. Exploring the Sensitivity of Prodromal Dementia with Lewy Bodies Research Criteria. Brain Sci 2022; 12:1594. [PMID: 36552054 PMCID: PMC9775171 DOI: 10.3390/brainsci12121594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is an insidious neurodegenerative disease characterised by a precipitous decline in cognition, sleep disturbances, motor impairment and psychiatric features. Recently, criteria for prodromal DLB (pDLB) including clinical features and biomarkers have been put forward to aid the classification and research of this ambiguous cohort of patients. Researchers can use these criteria to classify patients with mild cognitive impairment (MCI) with Lewy bodies (MCI-LB) as either possible (either one core clinical feature or one biomarker are present) or probable pDLB (at least two core clinical features, or one core clinical feature and at least one biomarker present). However, as isolated REM sleep behaviour disorder (iRBD) confirmed with polysomnography (PSG) can be included as both a clinical and a biomarker feature, potentially reducing the specificity of these diagnostic criteria. To address this issue, the current study classified a cohort of 47 PSG-confirmed iRBD patients as probable prodromal DLB only in the presence of an additional core feature or if there was an additional non-PSG biomarker. Thirteen iRBD patients demonstrated MCI (iRBD-MCI). In the iRBD-MCI group, one presented with parkinsonism and was thus classified as probable pDLB, whilst the remaining 12 were classified as only possible pDLB. All patients performed three tasks designed to measure attentional deficits, visual hallucinations and visuospatial impairment. Patients also attended clinical follow-ups to monitor for transition to DLB or another synucleinopathy. Findings indicated that the only patient categorised by virtue of having two core clinical features as probable pDLB transitioned over 28 months to a diagnosis of DLB. The performance of this probable pDLB patient was also ranked second-highest for their hallucinatory behaviours and had comparatively lower visuospatial accuracy. These findings highlight the need for more stringent diagnostic research criteria for pDLB, given that only one of the 13 patients who would have satisfied the current guidelines for probable pDLB transitioned to DLB after two years and was indeed the patient with two orthogonal core clinical features.
Collapse
Affiliation(s)
- Joseph R. Phillips
- Faculty of Medicine and Health, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
- School of Psychology & Marcs Institute for Brain and Behaviour, Western Sydney University, Sydney, NSW 2145, Australia
| | - Elie Matar
- Faculty of Medicine and Health, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
| | - Kaylena A. Ehgoetz Martens
- Faculty of Medicine and Health, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
- Department of Kinesiology, Faculty of Health, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Ahmed A. Moustafa
- Department of Human Anatomy and Physiology, The Faculty of Health Sciences, University of Johannesburg, Johannesburg 2092, South Africa
- School of Psychology, Faculty of Society and Design, Bond University, Gold Coast, QLD 4217, Australia
| | - Glenda M. Halliday
- Faculty of Medicine and Health, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
- Dementia and Movement Disorders Laboratory, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Simon J. G. Lewis
- Faculty of Medicine and Health, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
| |
Collapse
|
26
|
Shade RD, Ross JA, Van Bockstaele EJ. Targeting the cannabinoid system to counteract the deleterious effects of stress in Alzheimer’s disease. Front Aging Neurosci 2022; 14:949361. [PMID: 36268196 PMCID: PMC9577232 DOI: 10.3389/fnagi.2022.949361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder characterized histologically in postmortem human brains by the presence of dense protein accumulations known as amyloid plaques and tau tangles. Plaques and tangles develop over decades of aberrant protein processing, post-translational modification, and misfolding throughout an individual’s lifetime. We present a foundation of evidence from the literature that suggests chronic stress is associated with increased disease severity in Alzheimer’s patient populations. Taken together with preclinical evidence that chronic stress signaling can precipitate cellular distress, we argue that chronic psychological stress renders select circuits more vulnerable to amyloid- and tau- related abnormalities. We discuss the ongoing investigation of systemic and cellular processes that maintain the integrity of protein homeostasis in health and in degenerative conditions such as Alzheimer’s disease that have revealed multiple potential therapeutic avenues. For example, the endogenous cannabinoid system traverses the central and peripheral neural systems while simultaneously exerting anti-inflammatory influence over the immune response in the brain and throughout the body. Moreover, the cannabinoid system converges on several stress-integrative neuronal circuits and critical regions of the hypothalamic-pituitary-adrenal axis, with the capacity to dampen responses to psychological and cellular stress. Targeting the cannabinoid system by influencing endogenous processes or exogenously stimulating cannabinoid receptors with natural or synthetic cannabis compounds has been identified as a promising route for Alzheimer’s Disease intervention. We build on our foundational framework focusing on the significance of chronic psychological and cellular stress on the development of Alzheimer’s neuropathology by integrating literature on cannabinoid function and dysfunction within Alzheimer’s Disease and conclude with remarks on optimal strategies for treatment potential.
Collapse
Affiliation(s)
- Ronnie D. Shade
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Jennifer A. Ross
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
- *Correspondence: Jennifer A. Ross,
| | - Elisabeth J. Van Bockstaele
- Department of Pharmacology and Physiology, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
27
|
Eide PK, Pripp AH, Berge B, Hrubos-Strøm H, Ringstad G, Valnes LM. Altered glymphatic enhancement of cerebrospinal fluid tracer in individuals with chronic poor sleep quality. J Cereb Blood Flow Metab 2022; 42:1676-1692. [PMID: 35350917 PMCID: PMC9441729 DOI: 10.1177/0271678x221090747] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic sleep disturbance is a risk factor for dementia disease, possibly due to impaired sleep-dependent clearance of toxic metabolic by-products. We compared enrichment of a cerebrospinal fluid (CSF) tracer within brain of patients reporting good or poor sleep quality, assessed by the Pittsburgh Sleep Quality Index (PSQI) questionnaire. Tracer enrichment in a selection of brain regions was assessed using multiphase magnetic resonance imaging up to 48 hours after intrathecal administration of the contrast agent gadobutrol (0.5 ml of 1 mmol/ml) serving as tracer. Tracer enrichment differed between patients with good (PSQI ≤5) and poor (PSQI >5) sleep quality in a cohort of non-dementia individuals (n = 44; age 42.3 ± 14.5 years), and in patients with the dementia subtype idiopathic normal pressure hydrocephalus (n = 24; age 71.0 ± 4.9 years). Sleep impairment was associated with increased CSF tracer enrichment in several brain regions. Cortical brain volume as well as entorhinal cortex thickness was reduced in the oldest cohort and was correlated with the severity of sleep disturbance and the degree of cortical tracer enrichment. We suggest chronic sleep disturbance is accompanied by altered glymphatic function along enlarged perivascular spaces.
Collapse
Affiliation(s)
- Per Kristian Eide
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway.,Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | | | - Harald Hrubos-Strøm
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Otorhinolaryngology, Surgical Division, Akershus University Hospital, Nordbyhagen, Norway
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Lars Magnus Valnes
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| |
Collapse
|
28
|
Sola P, Krishnamurthy PT, Kumari M, Byran G, Gangadharappa HV, Garikapati KK. Neuroprotective approaches to halt Parkinson's disease progression. Neurochem Int 2022; 158:105380. [PMID: 35718278 DOI: 10.1016/j.neuint.2022.105380] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
One of the most significant threats in Parkinson's disease (PD) is neurodegeneration. Neurodegeneration at both nigral as well as non-nigral regions of the brain is considered responsible for disease progression in PD. The key factors that initiate neurodegeneration are oxidative stress, neuroinflammation, mitochondrial complex-1 inhibition, and abnormal α-synuclein (SNCA) protein aggregations. Nigral neurodegeneration results in motor symptoms (tremor, bradykinesia, rigidity, shuffling gait, and postural instability) whereas; non-nigral neurodegeneration is responsible for non-motor symptoms (depression, cognitive dysfunctions, sleep disorders, hallucination, and psychosis). The available therapies for PD aim at increasing dopamine levels. The medications such as Monoamine oxidase B (MAO-B) inhibitors, catechol o-methyltransferase (COMT) inhibitors, Dopamine precursor (Levodopa), dopamine agonists, and dopamine reuptake inhibitors drastically improve the motor symptoms and quality of life only in the early stages of the disease. However, dopa resistant motor symptoms (abnormality in posture, speech impediment, gait, and balance problems), dopa resistant non-motor signs (sleep problems, autonomic dysfunction, mood, and cognitive impairment, pain), and drug-related side effects (motor fluctuations, psychosis, and dyskinesias) are considered responsible for the failure of these therapies. Further, none of the treatments, alone or in combination, are capable of halting the disease progression in the long run. Therefore, there is a need to develop safe and efficient neuroprotective agents, which can slow or stop the disease progression for the better management of PD. In this review, an effort has been made to discuss the various mechanisms responsible for progressive neurodegeneration (disease progression) in PD and also multiple strategies available for halting disease progression.
Collapse
Affiliation(s)
- Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India.
| | - Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | - Gowramma Byran
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| | | | - Kusuma Kumari Garikapati
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, 643001, India
| |
Collapse
|
29
|
Gutiérrez IL, Dello Russo C, Novellino F, Caso JR, García-Bueno B, Leza JC, Madrigal JLM. Noradrenaline in Alzheimer's Disease: A New Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms23116143. [PMID: 35682822 PMCID: PMC9181823 DOI: 10.3390/ijms23116143] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/13/2022] Open
Abstract
A growing body of evidence demonstrates the important role of the noradrenergic system in the pathogenesis of many neurodegenerative processes, especially Alzheimer’s disease, due to its ability to control glial activation and chemokine production resulting in anti-inflammatory and neuroprotective effects. Noradrenaline involvement in this disease was first proposed after finding deficits of noradrenergic neurons in the locus coeruleus from Alzheimer’s disease patients. Based on this, it has been hypothesized that the early loss of noradrenergic projections and the subsequent reduction of noradrenaline brain levels contribute to cognitive dysfunctions and the progression of neurodegeneration. Several studies have focused on analyzing the role of noradrenaline in the development and progression of Alzheimer’s disease. In this review we summarize some of the most relevant data describing the alterations of the noradrenergic system normally occurring in Alzheimer’s disease as well as experimental studies in which noradrenaline concentration was modified in order to further analyze how these alterations affect the behavior and viability of different nervous cells. The combination of the different studies here presented suggests that the maintenance of adequate noradrenaline levels in the central nervous system constitutes a key factor of the endogenous defense systems that help prevent or delay the development of Alzheimer’s disease. For this reason, the use of noradrenaline modulating drugs is proposed as an interesting alternative therapeutic option for Alzheimer’s disease.
Collapse
Affiliation(s)
- Irene L. Gutiérrez
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Cinzia Dello Russo
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, Liverpool L69 3GL, UK
| | - Fabiana Novellino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council, 88100 Catanzaro, Italy
| | - Javier R. Caso
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - Juan C. Leza
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
| | - José L. M. Madrigal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid (UCM), Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Instituto de Investigación Neuroquímica (IUINQ-UCM), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Avda. Complutense s/n, 28040 Madrid, Spain; (I.L.G.); (F.N.); (J.R.C.); (B.G.-B.); (J.C.L.)
- Correspondence: ; Tel.: +34-91-394-1463
| |
Collapse
|
30
|
Guzmán-Ramos K, Osorio-Gómez D, Bermúdez-Rattoni F. Cognitive impairment in alzheimer’s and metabolic diseases: A catecholaminergic hypothesis. Neuroscience 2022; 497:308-323. [DOI: 10.1016/j.neuroscience.2022.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 12/16/2022]
|
31
|
Cerebrospinal fluid catecholamines in Alzheimer's disease patients with and without biological disease. Transl Psychiatry 2022; 12:151. [PMID: 35397615 PMCID: PMC8994756 DOI: 10.1038/s41398-022-01901-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/08/2022] Open
Abstract
Noradrenergic and dopaminergic neurons are involved in cognitive functions, relate to behavioral and psychological symptoms in dementia and are affected in Alzheimer's disease (AD). Amyloid plaques (A), neurofibrillary tangles (T) and neurodegeneration (N) hallmarks the AD neuropathology. Today, the AT(N) pathophysiology can be assessed through biomarkers. Previous studies report cerebrospinal fluid (CSF) catecholamine concentrations in AD patients without biomarker refinement. We explored if CSF catecholamines relate to AD clinical presentation or neuropathology as reflected by CSF biomarkers. CSF catecholamines were analyzed in AD patients at the mild cognitive impairment (MCI; n = 54) or dementia stage (n = 240) and in cognitively unimpaired (n = 113). CSF biomarkers determined AT status and indicated synaptic damage (neurogranin). The AD patients (n = 294) had higher CSF noradrenaline and adrenaline concentrations, but lower dopamine concentrations compared to the cognitively unimpaired (n = 113). AD patients in the MCI and dementia stage of the disease had similar CSF catecholamine concentrations. In the CSF neurogranin positively associated with noradrenaline and adrenaline but not with dopamine. Adjusted regression analyses including AT status, CSF neurogranin, age, gender, and APOEε4 status verified the findings. In restricted analyses comparing A+T+ patients to A-T- cognitively unimpaired, the findings for CSF adrenaline remained significant (p < 0.001) but not for CSF noradrenaline (p = 0.07) and CSF dopamine (p = 0.33). There were no differences between A+T+ and A-T- cognitively unimpaired. Thus, we find alterations in CSF catecholamines in symptomatic AD and the CSF adrenergic transmitters to increase simultaneously with synaptic damage as indexed by CSF neurogranin.
Collapse
|
32
|
Cassidy CM, Therriault J, Pascoal TA, Cheung V, Savard M, Tuominen L, Chamoun M, McCall A, Celebi S, Lussier F, Massarweh G, Soucy JP, Weinshenker D, Tardif C, Ismail Z, Gauthier S, Rosa-Neto P. Association of locus coeruleus integrity with Braak stage and neuropsychiatric symptom severity in Alzheimer's disease. Neuropsychopharmacology 2022; 47:1128-1136. [PMID: 35177805 PMCID: PMC8938499 DOI: 10.1038/s41386-022-01293-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/07/2022] [Accepted: 02/02/2022] [Indexed: 12/16/2022]
Abstract
The clinical and pathophysiological correlates of locus coeruleus (LC) degeneration in Alzheimer's disease (AD) could be clarified using a method to index LC integrity in vivo, neuromelanin-sensitive MRI (NM-MRI). We examined whether integrity of the LC-norepinephrine system, assessed with NM-MRI, is associated with stage of AD and with neuropsychiatric symptoms (NPS), independent of cortical pathophysiology (amyloid-β and tau burden). Cognitively normal older adults (n = 118), and individuals with mild cognitive impairment (MCI, n = 44), and AD (n = 28) underwent MR imaging and tau and amyloid-β positron emission tomography (with [18F]MK6240 and [18F]AZD4694, respectively). Integrity of the LC-norepinephrine system was assessed based on contrast-to-noise ratio of the LC on NM-MRI images. Braak stage of AD was derived from regional binding of [18F]MK6240. NPS were assessed with the Mild Behavioral Impairment Checklist (MBI-C). LC signal contrast was decreased in tau-positive participants (t186 = -4.00, p = 0.0001) and negatively correlated to Braak stage (Spearman ρ = -0.31, p = 0.00006). In tau-positive participants (n = 51), higher LC signal predicted NPS severity (ρ = 0.35, p = 0.019) independently of tau burden, amyloid-β burden, and cortical gray matter volume. This relationship appeared to be driven by the impulse dyscontrol domain of NPS, which was highly correlated to LC signal (ρ = 0.44, p = 0.0027). NM-MRI reveals loss of LC integrity that correlates to severity of AD. However, LC preservation in AD may also have negative consequences by conferring risk for impulse control symptoms. NM-MRI shows promise as a practical biomarker that could have utility in predicting the risk of NPS or guiding their treatment in AD.
Collapse
Affiliation(s)
- Clifford M. Cassidy
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada ,grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada
| | - Joseph Therriault
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Tharick A. Pascoal
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Victoria Cheung
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Melissa Savard
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Lauri Tuominen
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Mira Chamoun
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Adelina McCall
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Seyda Celebi
- grid.28046.380000 0001 2182 2255Institute of Mental Health Research, University of Ottawa, Ottawa, ON Canada
| | - Firoza Lussier
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada
| | - Gassan Massarweh
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Jean-Paul Soucy
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - David Weinshenker
- grid.189967.80000 0001 0941 6502Department of Human Genetics, Emory University School of Medicine, Atlanta, GA USA
| | - Christine Tardif
- grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| | - Zahinoor Ismail
- grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Serge Gauthier
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Alzheimer’s Disease Research Unit, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC Canada
| | - Pedro Rosa-Neto
- grid.14709.3b0000 0004 1936 8649Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Douglas Research Institute, Le Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Neurology and Neurosurgery, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Department of Psychiatry, McGill University, Montreal, QC Canada ,grid.14709.3b0000 0004 1936 8649Montreal Neurological Institute, McGill University, Montreal, QC Canada
| |
Collapse
|
33
|
Abstract
Cognitive impairment affects up to 80% of patients with Parkinson's disease (PD) and is associated with poor quality of life. PD cognitive dysfunction includes poor working memory, impairments in executive function and difficulty in set-shifting. The pathophysiology underlying cognitive impairment in PD is still poorly understood, but there is evidence to support involvements of the cholinergic, dopaminergic, and noradrenergic systems. Only rivastigmine, an acetyl- and butyrylcholinesterase inhibitor, is efficacious for the treatment of PD dementia, which limits management of cognitive impairment in PD. Whereas the role of the serotonergic system in PD cognition is less understood, through its interactions with other neurotransmitters systems, namely, the cholinergic system, it may be implicated in cognitive processes. In this chapter, we provide an overview of the pharmacological, clinical and pathological evidence that implicates the serotonergic system in mediating cognition in PD.
Collapse
|
34
|
Yu ZY, Yi X, Wang YR, Zeng GH, Tan CR, Cheng Y, Sun PY, Liu ZH, Wang YJ, Liu YH. Inhibiting α1-adrenergic receptor signaling pathway ameliorates AD-type pathologies and behavioral deficits in APPswe/PS1 mouse model. J Neurochem 2022; 161:293-307. [PMID: 35244207 DOI: 10.1111/jnc.15603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 01/25/2022] [Accepted: 02/23/2022] [Indexed: 12/01/2022]
Affiliation(s)
- Zhong-Yuan Yu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xu Yi
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ye-Ran Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Cheng-Rong Tan
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Pu-Yang Sun
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Hao Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Hui Liu
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Bastos P, Barbosa R. Motor reserve: How to build neuronal resilience against ageing and neurodegeneration? Rev Neurol (Paris) 2022; 178:845-854. [DOI: 10.1016/j.neurol.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/14/2021] [Accepted: 12/17/2021] [Indexed: 10/19/2022]
|
36
|
Azhar L, Kusumo RW, Marotta G, Lanctôt KL, Herrmann N. Pharmacological Management of Apathy in Dementia. CNS Drugs 2022; 36:143-165. [PMID: 35006557 DOI: 10.1007/s40263-021-00883-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 12/11/2022]
Abstract
Apathy is a highly prevalent symptom of dementia. Despite its association with faster cognitive and functional decline, decreased quality of life and increased mortality, no therapies are currently approved to treat apathy. The objective of this review was to summarize the drugs that have been studied for apathy treatment in patients with dementia (specifically Alzheimer's disease [AD], Huntington's disease [HD] and Parkinson's disease [PD] dementia; dementia with Lewy bodies [DLB]; vascular dementia [VaD]; and frontotemporal dementia [FTD]) based on their putative mechanisms of action. A search for relevant studies was performed using ClinicalTrials.gov and PubMed. Eligible studies were randomized controlled trials that were available in English and included at least one drug intervention and an apathy measure scale. A total of 52 studies that included patients with AD (n = 33 studies), PD (n = 5), HD (n = 1), DLB (n = 1), FTD (n = 3), VaD (n = 1), VaD and AD (n = 4), VaD and mixed dementia (n = 1), and AD, VaD and mixed dementia (n = 3) were eligible for inclusion. These studies showed that methylphenidate, olanzapine, cholinesterase inhibitors, choline alphoscerate, citalopram, memantine, and mibampator are the only beneficial drugs in AD-related apathy. For PD-related apathy, only methylphenidate, rotigotine and rivastigmine showed benefits. Regarding FTD- and DLB-related apathy, initial studies with agomelatine and rivastigmine showed benefits, respectively. As for HD- and only-VaD-related apathy, no drugs demonstrated benefits. With regards to mixed populations, memantine, galantamine and gingko biloba showed effects on apathy in the AD plus VaD populations and nimodipine in the VaD plus mixed dementia populations. Of the drugs with positive results, some are already prescribed to patients with dementia to target other symptoms, some have characteristics-such as medical contraindications (e.g., cardiovascular) and adverse effects (e.g., gastrointestinal disturbances)-that limit their clinical use and some require further study. Future studies should investigate apathy as a primary outcome, making use of appropriate sample sizes and study durations to ensure durability of results. There should also be a consensus on using scales with high test/retest and interrater reliabilities to limit the inconsistencies between clinical trials. In conclusion, there are currently no US FDA-approved drugs that target apathy in dementia, so there is an ongoing need for the development of such drugs.
Collapse
Affiliation(s)
- Laiba Azhar
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Raphael W Kusumo
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Giovanni Marotta
- Geriatric Medicine Division, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Krista L Lanctôt
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Kelberman MA, Anderson CR, Chlan E, Rorabaugh JM, McCann KE, Weinshenker D. Consequences of Hyperphosphorylated Tau in the Locus Coeruleus on Behavior and Cognition in a Rat Model of Alzheimer's Disease. J Alzheimers Dis 2022; 86:1037-1059. [PMID: 35147547 PMCID: PMC9007891 DOI: 10.3233/jad-215546] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The locus coeruleus (LC) is one of the earliest brain regions to accumulate hyperphosphorylated tau, but a lack of animal models that recapitulate this pathology has hampered our understanding of its contributions to Alzheimer's disease (AD) pathophysiology. OBJECTIVE We previously reported that TgF344-AD rats, which overexpress mutant human amyloid precursor protein and presenilin-1, accumulate early endogenous hyperphosphorylated tau in the LC. Here, we used TgF344-AD rats and a wild-type (WT) human tau virus to interrogate the effects of endogenous hyperphosphorylated rat tau and human tau in the LC on AD-related neuropathology and behavior. METHODS Two-month-old TgF344-AD and WT rats received bilateral LC infusions of full-length WT human tau or mCherry control virus driven by the noradrenergic-specific PRSx8 promoter. Rats were subsequently assessed at 6 and 12 months for arousal (sleep latency), anxiety-like behavior (open field, elevated plus maze, novelty-suppressed feeding), passive coping (forced swim task), and learning and memory (Morris water maze and fear conditioning). Hippocampal microglia, astrocyte, and AD pathology were evaluated using immunohistochemistry. RESULTS In general, the effects of age were more pronounced than genotype or treatment; older rats displayed greater hippocampal pathology, took longer to fall asleep, had reduced locomotor activity, floated more, and had impaired cognition compared to younger animals. TgF344-AD rats showed increased anxiety-like behavior and impaired learning and memory. The tau virus had negligible influence on most measures. CONCLUSION Effects of hyperphosphorylated tau on AD-like neuropathology and behavioral symptoms were subtle. Further investigation of different forms of tau is warranted.
Collapse
Affiliation(s)
- Michael A. Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA 30322
- Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA 30322
| | | | - Eli Chlan
- Neuroscience Program, Laney Graduate School, Emory University, Atlanta, GA 30322
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | | | | | | |
Collapse
|
38
|
Gan J, Chen Z, Han J, Ma L, Liu S, Wang XD, Ji Y. Orexin-A in Patients With Lewy Body Disease: A Systematic Review and Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:765701. [PMID: 34867809 PMCID: PMC8635768 DOI: 10.3389/fendo.2021.765701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Background Abnormal orexin-A levels in cerebrospinal fluid (CSF) have been identified in Alzheimer's disease (AD) and other neurodegenerative diseases. However, few studies have focused on Lewy body disease (LBD) and often with debatable outcomes. Thus, we performed this systematic review and meta-analysis to investigate orexin-A levels in LBD by incorporating data from different studies. Methods We gathered studies comparing orexin-A levels in patients with LBD and controls (including healthy controls and other dementia subtypes). In the initial search, 117 relevant articles were identified. After a selection process, seven studies, conducted in Japan, USA, Spain, Switzerland, France, Italy, and Netherlands, were chosen. Results In total, 179 patients with LBD and 253 controls were included. Patients with LBD had significantly lower mean orexin-A CSF levels when compared with patients with AD [standard mean difference (SMD): -0.35, 95% confidence interval (CI): -0.70 to -0.00, Z = 1.96, P = 0.05], whereas mean orexin-A levels were significantly higher when compared with patients with frontotemporal lobar degeneration (FTLD) (SMD: 0.61, 95% CI: 0.23-0.99, Z = 3.12, P = 0.002). Orexin-A CSF levels in LBD patients were approximately equal to levels in healthy elderly individuals, whereas they were significantly decreased in LBD patients with excessive daytime sleepiness (EDS) (SMD: -0.15, 95% CI: -0.59 to 0.29, Z = 0.67, P = 0.50). Conclusions We showed that orexin-A levels in patients with LBD were not very different from those in normal elderly individuals, whereas they were lower than those in AD patients and higher than those in FTLD patients. The influence of hypersomnia on orexin-A levels should be carefully interpreted. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42021265900.
Collapse
Affiliation(s)
- Jinghuan Gan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhichao Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiuyan Han
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lingyun Ma
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Xiao-Dan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
39
|
Ludwig M, Wienke C, Betts MJ, Zaehle T, Hämmerer D. Current challenges in reliably targeting the noradrenergic locus coeruleus using transcutaneous auricular vagus nerve stimulation (taVNS). Auton Neurosci 2021; 236:102900. [PMID: 34781120 DOI: 10.1016/j.autneu.2021.102900] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/03/2021] [Accepted: 10/15/2021] [Indexed: 12/11/2022]
Abstract
Transcutaneous auricular vagus nerve stimulation (taVNS), as a non-invasive brain stimulation technique may influence the locus coeruleus-norepinephrine system (LC-NE system) via modulation of the Vagus Nerve (VN) which projects to the LC. Few human studies exist examining the effects of taVNS on the LC-NE system and studies to date assessing the ability of taVNS to target the LC yield heterogeneous results. The aim of this review is to present an overview of the current challenges in assessing effects of taVNS on LC function and how translational approaches spanning animal and human research can help in this regard. A particular emphasis of the review discusses how the effects of taVNS may be influenced by changes in structure and function of the LC-NE system across the human lifespan and in disease.
Collapse
Affiliation(s)
- Mareike Ludwig
- Institute for Cognitive Neurology and Dementia Research, Faculty of Medicine, University Hospital Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, Magdeburg, Germany.
| | - Christian Wienke
- Department of Neurology, Section of Neuropsychology, Otto-v.-Guericke University, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Matthew J Betts
- Institute for Cognitive Neurology and Dementia Research, Faculty of Medicine, University Hospital Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Tino Zaehle
- Department of Neurology, Section of Neuropsychology, Otto-v.-Guericke University, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Dorothea Hämmerer
- Institute for Cognitive Neurology and Dementia Research, Faculty of Medicine, University Hospital Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London, UK; Department of Psychology, University of Innsbruck; CBBS Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
40
|
Henjum K, Godang K, Quist-Paulsen E, Idland AV, Neerland BE, Sandvig H, Brugård A, Raeder J, Frihagen F, Wyller TB, Hassel B, Bollerslev J, Watne LO. Cerebrospinal fluid catecholamines in delirium and dementia. Brain Commun 2021; 3:fcab121. [PMID: 34423298 PMCID: PMC8374970 DOI: 10.1093/braincomms/fcab121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 11/28/2022] Open
Abstract
Dopamine and noradrenaline are functionally connected to delirium and have been targets for pharmacological interventions but the biochemical evidence to support this notion is limited. To study the CSF levels of dopamine, noradrenaline and the third catecholamine adrenaline in delirium and dementia, these were quantified in three patient cohorts: (i) cognitively normal elderly patients (n = 122); (ii) hip fracture patients with or without delirium and dementia (n = 118); and (iii) patients with delirium precipitated by another medical condition (medical delirium, n = 26). Delirium was assessed by the Confusion Assessment Method. The hip fracture cohort had higher CSF levels of noradrenaline and adrenaline than the two other cohorts (both P < 0.001). Within the hip fracture cohort those with delirium (n = 65) had lower CSF adrenaline and dopamine levels than those without delirium (n = 52, P = 0.03, P = 0.002). Similarly, the medical delirium patients had lower CSF dopamine levels than the cognitively normal elderly (P < 0.001). Age did not correlate with the CSF catecholamine levels. These findings with lower CSF dopamine levels in hip fracture- and medical delirium patients challenge the theory of dopamine excess in delirium and question use of antipsychotics in delirium. The use of alpha-2 agonists with the potential to reduce noradrenaline release needs further examination.
Collapse
Affiliation(s)
- Kristi Henjum
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway.,Department of Geriatric Medicine, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Kristin Godang
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Ane-Victoria Idland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Bjørn Erik Neerland
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Heidi Sandvig
- Medical Department, Kristiansund Hospital, Møre og Romsdal Hospital Trust, 6508 Kristiansund, Norway
| | - Anniken Brugård
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Johan Raeder
- Department of Anesthesiology, Oslo University Hospital, 0424 Oslo, Norway
| | - Frede Frihagen
- Division of Orthopedic Surgery, Oslo University Hospital, 0424 Oslo, Norway
| | - Torgeir Bruun Wyller
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway.,Department of Geriatric Medicine, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway
| | - Bjørnar Hassel
- Department of Neurohabilitation, Oslo University Hospital, 0424 Oslo, Norway
| | - Jens Bollerslev
- Department of Geriatric Medicine, Institute of Clinical Medicine, University of Oslo, 0424 Oslo, Norway.,Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, 0424 Oslo, Norway
| | - Leiv Otto Watne
- Oslo Delirium Research Group, Department of Geriatric Medicine, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
41
|
Eide PK, Vinje V, Pripp AH, Mardal KA, Ringstad G. Sleep deprivation impairs molecular clearance from the human brain. Brain 2021; 144:863-874. [PMID: 33829232 DOI: 10.1093/brain/awaa443] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/24/2020] [Accepted: 10/08/2020] [Indexed: 01/02/2023] Open
Abstract
It remains an enigma why human beings spend one-third of their life asleep. Experimental data suggest that sleep is required for clearance of waste products from brain metabolism. This has, however, never been verified in humans. The primary aim of the present study was to examine in vivo whether one night of total sleep deprivation affects molecular clearance from the human brain. Secondarily, we examined whether clearance was affected by subsequent sleep. Multiphase MRI with standardized T1 sequences was performed up to 48 h after intrathecal administration of the contrast agent gadobutrol (0.5 ml of 1 mmol/ml), which served as a tracer molecule. Using FreeSurfer software, we quantified tracer enrichment within 85 brain regions as percentage change from baseline of normalized T1 signals. The cerebral tracer enrichment was compared between two cohorts of individuals; one cohort (n = 7) underwent total sleep deprivation from Day 1 to Day 2 (sleep deprivation group) while an age and gender-matched control group (n = 17; sleep group) was allowed free sleep from Day 1 to Day 2. From Day 2 to 3 all individuals were allowed free sleep. The tracer enriched the brains of the two groups similarly. Sleep deprivation was the sole intervention. One night of sleep deprivation impaired clearance of the tracer substance from most brain regions, including the cerebral cortex, white matter and limbic structures, as demonstrated on the morning of Day 2 after intervention (sleep deprivation/sleep). Moreover, the impaired cerebral clearance in the sleep deprivation group was not compensated by subsequent sleep from Day 2 to 3. The present results provide in vivo evidence that one night of total sleep deprivation impairs molecular clearance from the human brain, and that humans do not catch up on lost sleep.
Collapse
Affiliation(s)
- Per Kristian Eide
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Vegard Vinje
- Center for Biomedical Computing, Simula Research Laboratory, Lysaker, Norway
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Kent-Andre Mardal
- Center for Biomedical Computing, Simula Research Laboratory, Lysaker, Norway.,Department of Mathematics, University of Oslo, Oslo, Norway
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| |
Collapse
|
42
|
Mather M. Noradrenaline in the aging brain: Promoting cognitive reserve or accelerating Alzheimer's disease? Semin Cell Dev Biol 2021; 116:108-124. [PMID: 34099360 PMCID: PMC8292227 DOI: 10.1016/j.semcdb.2021.05.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022]
Abstract
Many believe that engaging in novel and mentally challenging activities promotes brain health and prevents Alzheimer's disease in later life. However, mental stimulation may also have risks as well as benefits. As neurons release neurotransmitters, they often also release amyloid peptides and tau proteins into the extracellular space. These by-products of neural activity can aggregate into the tau tangle and amyloid plaque signatures of Alzheimer's disease. Over time, more active brain regions accumulate more pathology. Thus, increasing brain activity can have a cost. But the neuromodulator noradrenaline, released during novel and mentally stimulating events, may have some protective effects-as well as some negative effects. Via its inhibitory and excitatory effects on neurons and microglia, noradrenaline sometimes prevents and sometimes accelerates the production and accumulation of amyloid-β and tau in various brain regions. Both α2A- and β-adrenergic receptors influence amyloid-β production and tau hyperphosphorylation. Adrenergic activity also influences clearance of amyloid-β and tau. Furthermore, some findings suggest that Alzheimer's disease increases noradrenergic activity, at least in its early phases. Because older brains clear the by-products of synaptic activity less effectively, increased synaptic activity in the older brain risks accelerating the accumulation of Alzheimer's pathology more than it does in the younger brain.
Collapse
Affiliation(s)
- Mara Mather
- Leonard Davis School of Gerontology, Department of Psychology, & Department of Biomedical Engineering, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089, United States.
| |
Collapse
|
43
|
Tilley BS, Patel SR, Goldfinger MH, Pearce RKB, Gentleman SM. Locus Coeruleus Pathology Indicates a Continuum of Lewy Body Dementia. JOURNAL OF PARKINSONS DISEASE 2021; 11:1641-1650. [PMID: 34334423 DOI: 10.3233/jpd-212748] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Lewy body dementia (LBD) has two main phenotypes of LBD, Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB), separated by the 'one-year-rule'. They also show different symptom profiles: core DLB features include fluctuating cognition, REM-sleep behaviur disorder, and visual hallucinations. These symptoms are sometimes present in PDD, representing an intermediate 'PDD-DLB' phenotype. OBJECTIVE DLB-like features may reflect deficits in the functions of the noradrenergic nucleus locus coeruleus (LC). Therefore, we compared the LC in the LBD phenotypes, PD, and controls. METHODS 38 PD, 56 PDD, 22 DLB, and 11 age-matched control cases from the Parkinson's UK tissue bank were included. LC tissue sections were immunostained for tyrosine-hydroxylase (TH), α-synuclein, tau, and amyloid-β. TH-neurons were quantified and pathologic burden calculated by %-coverage method. RESULTS The LC shows a stepwise reduction in neuron count from controls, PD, PDD, to DLB. PDD-DLB cases showed an intermediate clinical phenotype that was reflected pathologically. Cell counts were significantly reduced in DLB compared to PDD after correction for demographic factors. LC degeneration contributed significantly to the onset of all DLB symptoms. While α-synuclein was not significantly different between PDD and DLB cases, DLB exhibited significantly less tau pathology. CONCLUSION DLB and DLB-like symptoms represent noradrenergic deficits resulting from neuronal loss in the LC. PDD and DLB are likely to represent a clinical continuum based on the presence or absence of DLB-like symptoms mirrored by a pathological continuum in the LC.
Collapse
Affiliation(s)
- Bension S Tilley
- Neuropathology Unit, Department of Brain Sciences, Imperial College London, London, UK
| | - Shivani R Patel
- Neuropathology Unit, Department of Brain Sciences, Imperial College London, London, UK
| | - Marc H Goldfinger
- Neuropathology Unit, Department of Brain Sciences, Imperial College London, London, UK
| | - Ronald K B Pearce
- Neuropathology Unit, Department of Brain Sciences, Imperial College London, London, UK
| | - Steve M Gentleman
- Neuropathology Unit, Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
44
|
Plini ERG, O’Hanlon E, Boyle R, Sibilia F, Rikhye G, Kenney J, Whelan R, Melnychuk MC, Robertson IH, Dockree PM. Examining the Role of the Noradrenergic Locus Coeruleus for Predicting Attention and Brain Maintenance in Healthy Old Age and Disease: An MRI Structural Study for the Alzheimer's Disease Neuroimaging Initiative. Cells 2021; 10:1829. [PMID: 34359997 PMCID: PMC8306442 DOI: 10.3390/cells10071829] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
The noradrenergic theory of Cognitive Reserve (Robertson, 2013-2014) postulates that the upregulation of the locus coeruleus-noradrenergic system (LC-NA) originating in the brainstem might facilitate cortical networks involved in attention, and protracted activation of this system throughout the lifespan may enhance cognitive stimulation contributing to reserve. To test the above-mentioned theory, a study was conducted on a sample of 686 participants (395 controls, 156 mild cognitive impairment, 135 Alzheimer's disease) investigating the relationship between LC volume, attentional performance and a biological index of brain maintenance (BrainPAD-an objective measure, which compares an individual's structural brain health, reflected by their voxel-wise grey matter density, to the state typically expected at that individual's age). Further analyses were carried out on reserve indices including education and occupational attainment. Volumetric variation across groups was also explored along with gender differences. Control analyses on the serotoninergic (5-HT), dopaminergic (DA) and cholinergic (Ach) systems were contrasted with the noradrenergic (NA) hypothesis. The antithetic relationships were also tested across the neuromodulatory subcortical systems. Results supported by Bayesian modelling showed that LC volume disproportionately predicted higher attentional performance as well as biological brain maintenance across the three groups. These findings lend support to the role of the noradrenergic system as a key mediator underpinning the neuropsychology of reserve, and they suggest that early prevention strategies focused on the noradrenergic system (e.g., cognitive-attentive training, physical exercise, pharmacological and dietary interventions) may yield important clinical benefits to mitigate cognitive impairment with age and disease.
Collapse
Affiliation(s)
- Emanuele R. G. Plini
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Erik O’Hanlon
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychiatry, Royal College of Surgeons in Ireland, Hospital Rd, Beaumont, 9QRH+4F Dublin, Ireland
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Rory Boyle
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Francesca Sibilia
- Department of Psychiatry, School of Medicine Dublin, Trinity College Dublin, 152-160 Pearse St, 8QV3+99 Dublin, Ireland;
| | - Gaia Rikhye
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Joanne Kenney
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Robert Whelan
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Michael C. Melnychuk
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| | - Ian H. Robertson
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
- Department of Psychology, Global Brain Health Institute, Trinity College Dublin, Lloyd Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland;
| | - Paul M. Dockree
- Department of Psychology, Trinity College Institute of Neuroscience, Trinity College Dublin, Llyod Building, 42A Pearse St, 8PVX+GJ Dublin, Ireland; (E.O.); (R.B.); (G.R.); (J.K.); (M.C.M.); (I.H.R.); (P.M.D.)
| |
Collapse
|
45
|
Gallo A, Pillet LE, Verpillot R. New frontiers in Alzheimer's disease diagnostic: Monoamines and their derivatives in biological fluids. Exp Gerontol 2021; 152:111452. [PMID: 34182050 DOI: 10.1016/j.exger.2021.111452] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 04/29/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Current diagnosis of Alzheimer's disease (AD) relies on a combination of neuropsychological evaluations, biomarker measurements and brain imaging. Nevertheless, these approaches are either expensive, invasive or lack sensitivity to early AD stages. The main challenge of ongoing research is therefore to identify early non-invasive biomarkers to diagnose AD at preclinical stage. Accumulating evidence support the hypothesis that initial degeneration of profound monoaminergic nuclei may trigger a transneuronal spread of AD pathology towards hippocampus and cortex. These studies aroused great interest on monoamines, i.e. noradrenaline (NA), dopamine (D) ad serotonin (5-HT), as early hallmarks of AD pathology. The present work reviews current literature on the potential role of monoamines and related metabolites as biomarkers of AD. First, morphological changes in the monoaminergic systems during AD are briefly described. Second, we focus on concentration changes of these molecules and their derivatives in biological fluids, including cerebrospinal fluid, obtained by lumbar puncture, and blood or urine, sampled via less invasive procedures. Starting from initial observations, we then discuss recent insights on metabolomics-based analysis, highlighting the promising clinical utility of monoamines for the identification of a molecular AD signature, aimed at improving early diagnosis and discrimination from other dementia.
Collapse
|
46
|
Carrarini C, Russo M, Dono F, Barbone F, Rispoli MG, Ferri L, Di Pietro M, Digiovanni A, Ajdinaj P, Speranza R, Granzotto A, Frazzini V, Thomas A, Pilotto A, Padovani A, Onofrj M, Sensi SL, Bonanni L. Agitation and Dementia: Prevention and Treatment Strategies in Acute and Chronic Conditions. Front Neurol 2021; 12:644317. [PMID: 33935943 PMCID: PMC8085397 DOI: 10.3389/fneur.2021.644317] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/12/2021] [Indexed: 01/11/2023] Open
Abstract
Agitation is a behavioral syndrome characterized by increased, often undirected, motor activity, restlessness, aggressiveness, and emotional distress. According to several observations, agitation prevalence ranges from 30 to 50% in Alzheimer's disease, 30% in dementia with Lewy bodies, 40% in frontotemporal dementia, and 40% in vascular dementia (VaD). With an overall prevalence of about 30%, agitation is the third most common neuropsychiatric symptoms (NPS) in dementia, after apathy and depression, and it is even more frequent (80%) in residents of nursing homes. The pathophysiological mechanism underlying agitation is represented by a frontal lobe dysfunction, mostly involving the anterior cingulate cortex (ACC) and the orbitofrontal cortex (OFC), respectively, meaningful in selecting the salient stimuli and subsequent decision-making and behavioral reactions. Furthermore, increased sensitivity to noradrenergic signaling has been observed, possibly due to a frontal lobe up-regulation of adrenergic receptors, as a reaction to the depletion of noradrenergic neurons within the locus coeruleus (LC). Indeed, LC neurons mainly project toward the OFC and ACC. These observations may explain the abnormal reactivity to weak stimuli and the global arousal found in many patients who have dementia. Furthermore, agitation can be precipitated by several factors, e.g., the sunset or low lighted environments as in the sundown syndrome, hospitalization, the admission to nursing residencies, or changes in pharmacological regimens. In recent days, the global pandemic has increased agitation incidence among dementia patients and generated higher distress levels in patients and caregivers. Hence, given the increasing presence of this condition and its related burden on society and the health system, the present point of view aims at providing an extensive guide to facilitate the identification, prevention, and management of acute and chronic agitation in dementia patients.
Collapse
Affiliation(s)
- Claudia Carrarini
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Mirella Russo
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Fedele Dono
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Filomena Barbone
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Marianna G Rispoli
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Ferri
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Martina Di Pietro
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Anna Digiovanni
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Paola Ajdinaj
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Rino Speranza
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Alberto Granzotto
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology-CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Institute for Mind Impairments and Neurological Disorders-iMIND, University of California, Irvine, Irvine, CA, United States
| | - Valerio Frazzini
- Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology-CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Institut du Cerveau et de la Moelle épinière, ICM, INSERM UMRS 1127, CNRS UMR 7225, Pitié Salpêtrière Hospital, Paris, France.,AP-HP, GH Pitie-Salpêtrière-Charles Foix, Epilepsy Unit and Neurophysiology Department, Paris, France
| | - Astrid Thomas
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Parkinson's Disease Rehabilitation Centre, FERB ONLUS-S. Isidoro Hospital, Trescore Balneario, Italy
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology-CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Stefano L Sensi
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology-CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Bonanni
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy.,Behavioral Neurology and Molecular Neurology Units, Center for Advanced Studies and Technology-CAST, University G. d'Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
47
|
Combi R, Salsone M, Villa C, Ferini-Strambi L. Genetic Architecture and Molecular, Imaging and Prodromic Markers in Dementia with Lewy Bodies: State of the Art, Opportunities and Challenges. Int J Mol Sci 2021; 22:3960. [PMID: 33921279 PMCID: PMC8069386 DOI: 10.3390/ijms22083960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 11/16/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is one of the most common causes of dementia and belongs to the group of α-synucleinopathies. Due to its clinical overlap with other neurodegenerative disorders and its high clinical heterogeneity, the clinical differential diagnosis of DLB from other similar disorders is often difficult and it is frequently underdiagnosed. Moreover, its genetic etiology has been studied only recently due to the unavailability of large cohorts with a certain diagnosis and shows genetic heterogeneity with a rare contribution of pathogenic mutations and relatively common risk factors. The rapid increase in the reported cases of DLB highlights the need for an easy, efficient and accurate diagnosis of the disease in its initial stages in order to halt or delay the progression. The currently used diagnostic methods proposed by the International DLB consortium rely on a list of criteria that comprises both clinical observations and the use of biomarkers. Herein, we summarize the up-to-now reported knowledge on the genetic architecture of DLB and discuss the use of prodromal biomarkers as well as recent promising candidates from alternative body fluids and new imaging techniques.
Collapse
Affiliation(s)
- Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Maria Salsone
- Institute of Molecular Bioimaging and Physiology, National Research Council, 20054 Segrate (MI), Italy;
- Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Luigi Ferini-Strambi
- Department of Clinical Neurosciences, Neurology-Sleep Disorder Center, IRCCS San Raffaele Scientific Institute, 20127 Milan, Italy
- Department of Clinical Neurosciences, “Vita-Salute” San Raffaele University, 20127 Milan, Italy
| |
Collapse
|
48
|
Slater C, Wang Q. Alzheimer's disease: An evolving understanding of noradrenergic involvement and the promising future of electroceutical therapies. Clin Transl Med 2021; 11:e397. [PMID: 33931975 PMCID: PMC8087948 DOI: 10.1002/ctm2.397] [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: 11/11/2020] [Revised: 04/05/2021] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) poses a significant global health concern over the next several decades. Multiple hypotheses have been put forth that attempt to explain the underlying pathophysiology of AD. Many of these are briefly reviewed here, but to-date no disease-altering therapy has been achieved. Despite this, recent work expanding on the role of noradrenergic system dysfunction in both the pathogenesis and symptomatic exacerbation of AD has shown promise. The role norepinephrine (NE) plays in AD remains complicated but pre-tangle tau has consistently been shown to arise in the locus coeruleus (LC) of patients with AD decades before symptom onset. The current research reviewed here indicates NE can facilitate neuroprotective and memory-enhancing effects through β adrenergic receptors, while α2A adrenergic receptors may exacerbate amyloid toxicity through a contribution to tau hyperphosphorylation. AD appears to involve a disruption in the balance between these two receptors and their various subtypes. There is also a poorly characterized interplay between the noradrenergic and cholinergic systems. LC deterioration leads to maladaptation in the remaining LC-NE system and subsequently inhibits cholinergic neuron function, eventually leading to the classic cholinergic disruption seen in AD. Understanding AD as a dysfunctional noradrenergic system, provides new avenues for the use of advanced neural stimulation techniques to both study and therapeutically target the earliest stages of neuropathology. Direct LC stimulation and non-invasive vagus nerve stimulation (VNS) have both demonstrated potential use as AD therapeutics. Significant work remains, though, to better understand the role of the noradrenergic system in AD and how electroceuticals can provide disease-altering treatments.
Collapse
Affiliation(s)
- Cody Slater
- Department of Biomedical EngineeringColumbia UniversityNew YorkNew YorkUSA
- Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Qi Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
49
|
Kelly SC, Nelson PT, Counts SE. Pontine Arteriolosclerosis and Locus Coeruleus Oxidative Stress Differentiate Resilience from Mild Cognitive Impairment in a Clinical Pathologic Cohort. J Neuropathol Exp Neurol 2021; 80:325-335. [PMID: 33709107 DOI: 10.1093/jnen/nlab017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Locus coeruleus (LC) neurodegeneration is associated with cognitive deterioration during the transition from normal cognition to mild cognitive impairment (MCI) and Alzheimer disease (AD). However, the extent to which LC degenerative processes differentiate cognitively normal, "resilient" subjects bearing a high AD pathological burden from those with MCI or AD remains unclear. We approached this problem by quantifying the number of LC neurons and the percentage of LC neurons bearing AT8 tau pathology, TDP-43 pathology, or a marker for DNA/RNA oxidative damage, in well-characterized subjects diagnosed as normal cognition-low AD pathology (NC-LP), NC-high AD pathology (NC-HP), MCI, or mild/moderate AD. In addition, the severity of pontine arteriolosclerosis in each subject was compared across the groups. There was a trend for a step-wise ∼20% loss of LC neuron number between the NC-LP, NC-HP and MCI subjects despite a successive, significant ∼80%-100% increase in tau pathology between these groups. In contrast, increasing pontine arteriolosclerosis severity scores and LC oxidative stress burden significantly separated the NC-LP/HP and MCI/AD groups via comparative, correlation, and regression analysis. Pontine perfusion, as well as LC neuronal metabolic and redox function, may impact noradrenergic LC modulation of cognition during the preclinical and prodromal stages of AD.
Collapse
Affiliation(s)
- Sarah C Kelly
- From the Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan
| | - Peter T Nelson
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky.,University of Kentucky Alzheimer's Disease Research Center, Lexington, Kentucky
| | - Scott E Counts
- From the Department of Translational Neuroscience, Michigan State University, Grand Rapids, Michigan.,Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
50
|
Noradrenergic correlates of chronic cocaine craving: neuromelanin and functional brain imaging. Neuropsychopharmacology 2021; 46:851-859. [PMID: 33408330 PMCID: PMC8027452 DOI: 10.1038/s41386-020-00937-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Preclinical studies have implicated noradrenergic (NA) dysfunction in cocaine addiction. In particular, the NA system plays a central role in motivated behavior and may partake in the regulation of craving and drug use. Yet, human studies of the NA system are scarce, likely hampered by the difficulty in precisely localizing the locus coeruleus (LC). Here, we used neuromelanin imaging to localize the LC and quantified LC neuromelanin signal (NMS) intensity in 44 current cocaine users (CU; 37 men) and 59 nondrug users (NU; 44 men). We also employed fMRI to investigate cue-induced regional responses and LC functional connectivities, as quantified by generalized psychophysiological interaction (gPPI), in CU. Imaging data were processed by published routines and the findings were evaluated with a corrected threshold. We examined how these neural measures were associated with chronic cocaine craving, as assessed by the Cocaine Craving Questionnaire (CCQ). Compared to NU, CU demonstrated higher LC NMS for all probabilistic thresholds defined of 50-90% of the peak. In contrast, NMS of the ventral tegmental area/substantia nigra (VTA/SN) did not show significant group differences. Drug as compared to neutral cues elicited higher activations of many cortical and subcortical regions, none of which were significantly correlated with CCQ score. Drug vs. neutral cues also elicited "deactivation" of bilateral parahippocampal gyri (PHG) and PHG gPPI with a wide array of cortical and subcortical regions, including the ventral striatum and, with small volume correction, the LC. Less deactivation of the PHG (r = 0.40, p = 0.008) and higher PHG-LC gPPI (r = 0.44, p = 0.003) were positively correlated with the CCQ score. In contrast, PHG-VTA/SN connectivity did not correlate with the CCQ score. Together, chronic cocaine exposure may induce higher NMS intensity, suggesting neurotoxic effects on the LC. The correlation of cue-elicited PHG LC connectivity with CCQ score suggests a noradrenergic correlate of chronic cocaine craving. Potentially compensating for memory functions as in neurodegenerative conditions, cue-elicited PHG LC circuit connectivity plays an ill-adaptive role in supporting cocaine craving.
Collapse
|