1
|
Deng F, Dounavi ME, Plini ERG, Ritchie K, Muniz-Terrera G, Hutchinson S, Malhotra P, Ritchie CW, Lawlor B, Naci L. Cardiovascular risk of dementia is associated with brain-behaviour changes in cognitively healthy, middle-aged individuals. Neurobiol Aging 2024; 144:78-92. [PMID: 39293163 DOI: 10.1016/j.neurobiolaging.2024.09.006] [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: 09/22/2022] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Alzheimer's Disease (AD) neuropathology start decades before clinical manifestations, but whether risk factors are associated with early cognitive and brain changes in midlife remains poorly understood. We examined whether AD risk factors were associated with cognition and functional connectivity (FC) between the Locus Coeruleus (LC) and hippocampus - two key brain structures in AD neuropathology - cross-sectionally and longitudinally in cognitively healthy midlife individuals. Neuropsychological assessments and functional Magnetic Resonance Imaging were obtained at baseline (N=210), and two-years follow-up (N=188). Associations of cognition and FC with apolipoprotein ε4 (APOE ε4) genotype, family history of dementia, and the Cardiovascular Risk Factors, Aging, and Incidence of Dementia (CAIDE) score were investigated. Cross-sectionally, higher CAIDE scores were associated with worse cognition. Menopausal status interacted with the CAIDE risk on cognition. Furthermore, the CAIDE score significantly moderated the relationship between cognition and LC-Hippocampus FC. Longitudinally, the LC-Hippocampus FC decreased significantly over 2 years. These results suggest that cardiovascular risk of dementia is associated with brain-behaviour changes in cognitively healthy, middle-aged individuals.
Collapse
Affiliation(s)
- Feng Deng
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK
| | - Emanuele R G Plini
- Trinity College Institute of Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Karen Ritchie
- U1061 Neuropsychiatry, INSERM, University of Montpellier, Montpellier, France
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention, University of Edinburgh, Edinburgh, UK; Department of Social medicine, Ohio University, USA
| | | | - Paresh Malhotra
- Department of Brain Science, Imperial College Healthcare NHS Trust, UK
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, University of Edinburgh, Edinburgh, UK
| | - Brian Lawlor
- Trinity College Institute of Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorina Naci
- Trinity College Institute of Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Wengler K, Trujillo P, Cassidy CM, Horga G. Neuromelanin-sensitive MRI for mechanistic research and biomarker development in psychiatry. Neuropsychopharmacology 2024; 50:137-152. [PMID: 39160355 PMCID: PMC11526017 DOI: 10.1038/s41386-024-01934-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/21/2024] [Accepted: 07/15/2024] [Indexed: 08/21/2024]
Abstract
Neuromelanin-sensitive MRI is a burgeoning non-invasive neuroimaging method with an increasing number of applications in psychiatric research. This MRI modality is sensitive to the concentration of neuromelanin, which is synthesized from intracellular catecholamines and accumulates in catecholaminergic nuclei including the dopaminergic substantia nigra and the noradrenergic locus coeruleus. Emerging data suggest the utility of neuromelanin-sensitive MRI as a proxy measure for variability in catecholamine metabolism and function, even in the absence of catecholaminergic cell loss. Given the importance of catecholamine function to several psychiatric disorders and their treatments, neuromelanin-sensitive MRI is ideally positioned as an informative and easy-to-acquire catecholaminergic index. In this review paper, we examine basic aspects of neuromelanin and neuromelanin-sensitive MRI and focus on its psychiatric applications in the contexts of mechanistic research and biomarker development. We discuss ongoing debates and state-of-the-art research into the mechanisms of the neuromelanin-sensitive MRI contrast, standardized protocols and optimized analytic approaches, and application of cutting-edge methods such as machine learning and artificial intelligence to enhance the feasibility and predictive power of neuromelanin-sensitive-MRI-based tools. We finally lay out important future directions to allow neuromelanin-sensitive-MRI to fulfill its potential as a key component of the research, and ultimately clinical, toolbox in psychiatry.
Collapse
Affiliation(s)
- Kenneth Wengler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Vanderbilt, TN, USA
| | - Clifford M Cassidy
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Guillermo Horga
- New York State Psychiatric Institute, New York, NY, USA.
- Department of Psychiatry, Columbia University, New York, NY, USA.
| |
Collapse
|
3
|
Olchanyi MD, Augustinack J, Haynes RL, Lewis LD, Cicero N, Li J, Destrieux C, Folkerth RD, Kinney HC, Fischl B, Brown EN, Iglesias JE, Edlow BL. Histology-guided MRI segmentation of brainstem nuclei critical to consciousness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.26.24314117. [PMID: 39399006 PMCID: PMC11469455 DOI: 10.1101/2024.09.26.24314117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
While substantial progress has been made in mapping the connectivity of cortical networks responsible for conscious awareness, neuroimaging analysis of subcortical arousal networks that modulate arousal (i.e., wakefulness) has been limited by a lack of a robust segmentation procedures for brainstem arousal nuclei. Automated segmentation of brainstem arousal nuclei is an essential step toward elucidating the physiology of arousal in human consciousness and the pathophysiology of disorders of consciousness. We created a probabilistic atlas of brainstem arousal nuclei built on diffusion MRI scans of five ex vivo human brain specimens scanned at 750 μm isotropic resolution. Labels of arousal nuclei used to generate the probabilistic atlas were manually annotated with reference to nucleus-specific immunostaining in two of the five brain specimens. We then developed a Bayesian segmentation algorithm that utilizes the probabilistic atlas as a generative model and automatically identifies brainstem arousal nuclei in a resolution- and contrast-agnostic manner. The segmentation method displayed high accuracy in both healthy and lesioned in vivo T1 MRI scans and high test-retest reliability across both T1 and T2 MRI contrasts. Finally, we show that the segmentation algorithm can detect volumetric changes and differences in magnetic susceptibility within brainstem arousal nuclei in Alzheimer's disease and traumatic coma, respectively. We release the probabilistic atlas and Bayesian segmentation tool in FreeSurfer to advance the study of human consciousness and its disorders.
Collapse
|
4
|
Sinakevitch IT, McDermott KE, Gray DT, Barnes CA. A combined MRI, histological and immunohistochemical rendering of the rhesus macaque locus coeruleus (LC) enables the differentiation of three distinct LC subcompartments. J Chem Neuroanat 2024; 140:102449. [PMID: 39084478 PMCID: PMC11392618 DOI: 10.1016/j.jchemneu.2024.102449] [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: 05/22/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
Locus coeruleus (LC) neurons send their noradrenergic axons across multiple brain regions, including neocortex, subcortical regions, and spinal cord. Many aspects of cognition are known to be dependent on the noradrenergic system, and it has been suggested that dysfunction in this system may play central roles in cognitive decline associated with both normative aging and neurodegenerative disease. While basic anatomical and biochemical features of the LC have been examined in many species, detailed characterizations of the structure and function of the LC across the lifespan are not currently available. This includes the rhesus macaque, which is an important model of human brain function because of their striking similarities in brain architecture and behavioral capacities. In the present study, we describe a method to combine structural MRI, Nissl, and immunofluorescent histology from individual monkeys to reconstruct, in 3 dimensions, the entire macaque LC nucleus. Using these combined methods, a standardized volume of the LC was determined, and high-resolution confocal images of tyrosine hydroxylase-positive neurons were mapped into this volume. This detailed representation of the LC allows definitions to be proposed for three distinct subnuclei, including a medial region and a lateral region (based on location with respect to the central gray, inside or outside, respectively), and a compact region (defined by densely packed neurons within the medial compartment). This enabled the volume to be estimated and cell density to be calculated independently in each LC subnucleus for the first time. This combination of methods should allow precise characterization of the LC and has the potential to do the same for other nuclei with distinct molecular features.
Collapse
Affiliation(s)
- Irina T Sinakevitch
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States.
| | - Kelsey E McDermott
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States.
| | - Daniel T Gray
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States.
| | - Carol A Barnes
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ 85721, United States; Departments of Psychology, Neurology and Neuroscience, University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
5
|
Saggu S, Bai A, Aida M, Rehman H, Pless A, Ware D, Deak F, Jiao K, Wang Q. Monoamine alterations in Alzheimer's disease and their implications in comorbid neuropsychiatric symptoms. GeroScience 2024:10.1007/s11357-024-01359-x. [PMID: 39331291 DOI: 10.1007/s11357-024-01359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by relentless cognitive decline and the emergence of profoundly disruptive neuropsychiatric symptoms. As the disease progresses, it unveils a formidable array of neuropsychiatric manifestations, including debilitating depression, anxiety, agitation, and distressing episodes of psychosis. The intricate web of the monoaminergic system, governed by serotonin, dopamine, and norepinephrine, significantly influences our mood, cognition, and behavior. Emerging evidence suggests that dysregulation and degeneration of this system occur early in AD, leading to notable alterations in these critical neurotransmitters' levels, metabolism, and receptor function. However, how the degeneration of monoaminergic neurons and subsequent compensatory changes contribute to the presentation of neuropsychiatric symptoms observed in Alzheimer's disease remains elusive. This review synthesizes current findings on monoamine alterations in AD and explores how these changes contribute to the neuropsychiatric symptomatology of the disease. By elucidating the biological underpinnings of AD-related psychiatric symptoms, we aim to underscore the complexity and inform innovative approaches for treating neuropsychiatric symptoms in AD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| | - Ava Bai
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Mae Aida
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hasibur Rehman
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Andrew Pless
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Destany Ware
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Kai Jiao
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
6
|
Bennett IJ, Langley J, Sun A, Solis K, Seitz AR, Hu XP. Locus coeruleus contrast and diffusivity metrics differentially relate to age and memory performance. Sci Rep 2024; 14:15372. [PMID: 38965363 PMCID: PMC11224383 DOI: 10.1038/s41598-024-66238-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024] Open
Abstract
Neurocognitive aging researchers are increasingly focused on the locus coeruleus, a neuromodulatory brainstem structure that degrades with age. With this rapid growth, the field will benefit from consensus regarding which magnetic resonance imaging (MRI) metrics of locus coeruleus structure are most sensitive to age and cognition. To address this need, the current study acquired magnetization transfer- and diffusion-weighted MRI images in younger and older adults who also completed a free recall memory task. Results revealed significantly larger differences between younger and older adults for maximum than average magnetization transfer-weighted contrast (MTC), axial than mean or radial single-tensor diffusivity (DTI), and free than restricted multi-compartment diffusion (NODDI) metrics in the locus coeruleus; with maximum MTC being the best predictor of age group. Age effects for all imaging modalities interacted with sex, with larger age group differences in males than females for MTC and NODDI metrics. Age group differences also varied across locus coeruleus subdivision for DTI and NODDI metrics, and across locus coeruleus hemispheres for MTC. Within older adults, however, there were no significant effects of age on MTC or DTI metrics, only an interaction between age and sex for free diffusion. Finally, independent of age and sex, higher restricted diffusion in the locus coeruleus was significantly related to better (lower) recall variability, but not mean recall. Whereas MTC has been widely used in the literature, our comparison between the average and maximum MTC metrics, inclusion of DTI and NODDI metrics, and breakdowns by locus coeruleus subdivision and hemisphere make important and novel contributions to our understanding of the aging of locus coeruleus structure.
Collapse
Affiliation(s)
- Ilana J Bennett
- Department of Psychology, University of California, 900 University Avenue, 2127 Psychology Building, Riverside, CA, 92521-0426, USA.
| | - Jason Langley
- Center for Advanced Neuroimaging, University of California Riverside, Riverside, CA, USA
| | - Andrew Sun
- Department of Psychology, Northeastern University, Boston, MA, USA
| | - Kitzia Solis
- Department of Psychology, University of California, 900 University Avenue, 2127 Psychology Building, Riverside, CA, 92521-0426, USA
| | - Aaron R Seitz
- Department of Psychology, University of California, 900 University Avenue, 2127 Psychology Building, Riverside, CA, 92521-0426, USA
- Department of Psychology, Northeastern University, Boston, MA, USA
| | - Xiaoping P Hu
- Center for Advanced Neuroimaging, University of California Riverside, Riverside, CA, USA
- Department of Bioengineering, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
7
|
Li Z, Chen X, He W, Chen H, Chen D. The causal effect of Alzheimer's disease and family history of Alzheimer's disease on non-ischemic cardiomyopathy and left ventricular structure and function: a Mendelian randomization study. Front Genet 2024; 15:1379865. [PMID: 38903751 PMCID: PMC11188370 DOI: 10.3389/fgene.2024.1379865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Background Previous studies have shown that Alzheimer's disease (AD) can cause myocardial damage. However, whether there is a causal association between AD and non-ischemic cardiomyopathy (NICM) remains unclear. Using a comprehensive two-sample Mendelian randomization (MR) method, we aimed to determine whether AD and family history of AD (FHAD) affect left ventricular (LV) structure and function and lead to NICM, including hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Methods The summary statistics for exposures [AD, paternal history of AD (PH-AD), and maternal history of AD (MH-AD)] and outcomes (NICM, HCM, DCM, and LV traits) were obtained from the large European genome-wide association studies. The causal effects were estimated using inverse variance weighted, MR-Egger, and weighted median methods. Sensitivity analyses were conducted, including Cochran's Q test, MR-Egger intercept test, MR pleiotropy residual sum and outlier, MR Steiger test, leave-one-out analysis, and the funnel plot. Results Genetically predicted AD was associated with a lower risk of NICM [odds ratio (OR) 0.9306, 95% confidence interval (CI) 0.8825-0.9813, p = 0.0078], DCM (OR 0.8666, 95% CI 0.7752-0.9689, p = 0.0119), and LV remodeling index (OR 0.9969, 95% CI 0.9940-0.9998, p = 0.0337). Moreover, genetically predicted PH-AD was associated with a decreased risk of NICM (OR 0.8924, 95% CI 0.8332-0.9557, p = 0.0011). MH-AD was also strongly associated with a decreased risk of NICM (OR 0.8958, 95% CI 0.8449-0.9498, p = 0.0002). Different methods of sensitivity analysis demonstrated the robustness of the results. Conclusion Our study revealed that AD and FHAD were associated with a decreased risk of NICM, providing a new genetic perspective on the pathogenesis of NICM.
Collapse
Affiliation(s)
| | | | | | | | - Dehai Chen
- Department of Cardiovascular Surgery, The First People’s Hospital of Zhaoqing, The First Affiliated Hospital of Zhaoqing Medical College, Zhaoqing, China
| |
Collapse
|
8
|
Kim AJ, Nguyen K, Mather M. Eye movements reveal age differences in how arousal modulates saliency priority but not attention processing speed. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592619. [PMID: 38766110 PMCID: PMC11100628 DOI: 10.1101/2024.05.06.592619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The arousal-biased competition theory posits that inducing arousal increases attentional priority of salient stimuli while reducing priority of non-pertinent stimuli. However, unlike in young adults, older adults rarely exhibit shifts in priority under increased arousal, and prior studies have proposed different neural mechanisms to explain how arousal differentially modulates selective attention in older adults. Therefore, we investigated how the threat of unpredictable shock differentially modulates attentional control mechanisms in young and older adults by observing eye movements. Participants completed two oculomotor search tasks in which the salient distractor was typically captured by attention (singleton search) or proactively suppressed (feature search). We found that arousal did not modulate attentional priority for any stimulus among older adults nor affect the speed of attention processing in either age group. Furthermore, we observed that arousal modulated pupil sizes and found a correlation between evoked pupil responses and oculomotor function. Our findings suggest age differences in how the locus coeruleus-noradrenaline system interacts with neural networks of attention and oculomotor function.
Collapse
Affiliation(s)
- Andy Jeesu Kim
- University of Southern California, School of Gerontology
| | | | - Mara Mather
- University of Southern California, School of Gerontology
| |
Collapse
|
9
|
Trujillo P, Aumann MA, Claassen DO. Neuromelanin-sensitive MRI as a promising biomarker of catecholamine function. Brain 2024; 147:337-351. [PMID: 37669320 PMCID: PMC10834262 DOI: 10.1093/brain/awad300] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/17/2023] [Accepted: 08/20/2023] [Indexed: 09/07/2023] Open
Abstract
Disruptions to dopamine and noradrenergic neurotransmission are noted in several neurodegenerative and psychiatric disorders. Neuromelanin-sensitive (NM)-MRI offers a non-invasive approach to visualize and quantify the structural and functional integrity of the substantia nigra and locus coeruleus. This method may aid in the diagnosis and quantification of longitudinal changes of disease and could provide a stratification tool for predicting treatment success of pharmacological interventions targeting the dopaminergic and noradrenergic systems. Given the growing clinical interest in NM-MRI, understanding the contrast mechanisms that generate this signal is crucial for appropriate interpretation of NM-MRI outcomes and for the continued development of quantitative MRI biomarkers that assess disease severity and progression. To date, most studies associate NM-MRI measurements to the content of the neuromelanin pigment and/or density of neuromelanin-containing neurons, while recent studies suggest that the main source of the NM-MRI contrast is not the presence of neuromelanin but the high-water content in the dopaminergic and noradrenergic neurons. In this review, we consider the biological and physical basis for the NM-MRI contrast and discuss a wide range of interpretations of NM-MRI. We describe different acquisition and image processing approaches and discuss how these methods could be improved and standardized to facilitate large-scale multisite studies and translation into clinical use. We review the potential clinical applications in neurological and psychiatric disorders and the promise of NM-MRI as a biomarker of disease, and finally, we discuss the current limitations of NM-MRI that need to be addressed before this technique can be utilized as a biomarker and translated into clinical practice and offer suggestions for future research.
Collapse
Affiliation(s)
- Paula Trujillo
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Megan A Aumann
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| | - Daniel O Claassen
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37212, USA
| |
Collapse
|
10
|
Galgani A, Giorgi FS. Exploring the Role of Locus Coeruleus in Alzheimer's Disease: a Comprehensive Update on MRI Studies and Implications. Curr Neurol Neurosci Rep 2023; 23:925-936. [PMID: 38064152 PMCID: PMC10724305 DOI: 10.1007/s11910-023-01324-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2023] [Indexed: 12/18/2023]
Abstract
PURPOSE OF REVIEW Performing a thorough review of magnetic resonance imaging (MRI) studies assessing locus coeruleus (LC) integrity in ageing and Alzheimer's disease (AD), and contextualizing them with current preclinical and neuropathological literature. RECENT FINDINGS MRI successfully detected LC alterations in ageing and AD, identifying degenerative phenomena involving this nucleus even in the prodromal stages of the disorder. The degree of LC disruption was also associated with the severity of AD cortical pathology, cognitive and behavioral impairment, and the risk of clinical progression. Locus coeruleus-MRI has proved to be a useful tool to assess the integrity of the central noradrenergic system in vivo in humans. It allowed to test in patients preclinical and experimental hypothesis, thus confirming the specific and marked involvement of the LC in AD and its key pathogenetic role. Locus coeruleus-MRI-related data might represent the theoretical basis on which to start developing noradrenergic drugs to target AD.
Collapse
Affiliation(s)
- Alessandro Galgani
- Department of Translational Research and of New Surgical and Medical Technologies School of Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies School of Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| |
Collapse
|
11
|
Tang Y, Cao M, Li Y, Lin Y, Wu X, Chen M. Altered structural covariance of locus coeruleus in individuals with significant memory concern and patients with mild cognitive impairment. Cereb Cortex 2023; 33:8523-8533. [PMID: 37130822 PMCID: PMC10321106 DOI: 10.1093/cercor/bhad137] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/31/2023] [Accepted: 04/02/2023] [Indexed: 05/04/2023] Open
Abstract
The locus coeruleus (LC) is the site where tau accumulation is preferentially observed pathologically in Alzheimer's disease (AD) patients, but the changes in gray matter co-alteration patterns between the LC and the whole brain in the predementia phase of AD remain unclear. In this study, we estimated and compared the gray matter volume of the LC and its structural covariance (SC) with the whole brain among 161 normal healthy controls (HCs), 99 individuals with significant memory concern (SMC) and 131 patients with mild cognitive impairment (MCI). We found that SC decreased in MCI groups, which mainly involved the salience network and default mode network. These results imply that seeding from LC, the gray matter network disruption and disconnection appears early in the MCI group. The altered SC network seeding from the LC can serve as an imaging biomarker for discriminating the patients in the potential predementia phase of AD from the normal subjects.
Collapse
Affiliation(s)
- Yingmei Tang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang Road West, Guangzhou 510120, Guangdong, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang Road West, Guangzhou 510120, Guangdong, China
| | - Yunhua Li
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang Road West, Guangzhou 510120, Guangdong, China
| | - Yuting Lin
- School of Psychology, Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, No.55 Zhongshan Avenue West, Guangzhou 510631, Guangdong, China
| | - Xiaoyan Wu
- School of Psychology, Key Laboratory of Brain, Cognition and Education Sciences (South China Normal University), Ministry of Education, Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, No.55 Zhongshan Avenue West, Guangzhou 510631, Guangdong, China
| | - Meiwei Chen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang Road West, Guangzhou 510120, Guangdong, China
| |
Collapse
|
12
|
Arulchelvan E, Vanneste S. Promising neurostimulation routes for targeting the hippocampus to improve episodic memory: A review. Brain Res 2023:148457. [PMID: 37315722 DOI: 10.1016/j.brainres.2023.148457] [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: 03/30/2023] [Revised: 05/24/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
This review aims to highlight modern neurostimulation approaches that are effectively activating the hippocampus and enhancing episodic memory performance. The hippocampus is a brain region known to play an essential role in episodic memory processes. However, as it is nestled deep within the brain, it has been a challenging target for traditional neurostimulation approaches, with studies reporting inconsistent memory effects. Recent studies suggest more than half of the electrical current from non-invasive transcranial electrical stimulation (tES) methods may be attenuated by the human scalp, skull, and cerebral spinal fluid. Thus, this review aims to highlight novel neurostimulation approaches that are showing promise as alternative routes for activating hippocampal circuitry. Early evidence suggests temporal interference, closed-loop and individualized protocols, sensory stimulation and peripheral nerve-targeted tES protocols warrant further investigation. These approaches each provide promising routes for activating the hippocampus by a) increasing its functional connectiveness to key brain regions, b) strengthening synaptic plasticity mechanisms, or c) enhancing neural entrainment specifically within and between theta and gamma frequencies in these regions. Importantly, these three functional mechanisms and the hippocampus' structural integrity are negatively impacted throughout the progression of Alzheimer's Disease, with episodic memory deficits likewise evident in early stages. Consequently, depending on further validation of the approaches reviewed here, these techniques could offer significant applied therapeutic value for patients suffering from memory deficits or neurodegenerative diseases including amnestic Mild Cognitive Impairment or Alzheimer's disease.
Collapse
Affiliation(s)
- Elva Arulchelvan
- Lab for Clinical and Integrative Neuroscience, Trinity Institute for Neuroscience, School of Psychology, Trinity College Dublin, Ireland
| | - Sven Vanneste
- Global Brain Health Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Yamazaki Y, Suwabe K, Nagano-Saito A, Saotome K, Kuwamizu R, Hiraga T, Torma F, Suzuki K, Sankai Y, Yassa MA, Soya H. A possible contribution of the locus coeruleus to arousal enhancement with mild exercise: evidence from pupillometry and neuromelanin imaging. Cereb Cortex Commun 2023; 4:tgad010. [PMID: 37323937 PMCID: PMC10267300 DOI: 10.1093/texcom/tgad010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 06/17/2023] Open
Abstract
Acute mild exercise has been observed to facilitate executive function and memory. A possible underlying mechanism of this is the upregulation of the ascending arousal system, including the catecholaminergic system originating from the locus coeruleus (LC). Prior work indicates that pupil diameter, as an indirect marker of the ascending arousal system, including the LC, increases even with very light-intensity exercise. However, it remains unclear whether the LC directly contributes to exercise-induced pupil-linked arousal. Here, we examined the involvement of the LC in the change in pupil dilation induced by very light-intensity exercise using pupillometry and neuromelanin imaging to assess the LC integrity. A sample of 21 young males performed 10 min of very light-intensity exercise, and we measured changes in the pupil diameters and psychological arousal levels induced by the exercise. Neuromelanin-weighted magnetic resonance imaging scans were also obtained. We observed that pupil diameter and psychological arousal levels increased during very light-intensity exercise, which is consistent with previous findings. Notably, the LC contrast, a marker of LC integrity, predicted the magnitude of pupil dilation and psychological arousal enhancement with exercise. These relationships suggest that the LC-catecholaminergic system is a potential a mechanism for pupil-linked arousal induced by very light-intensity exercise.
Collapse
Affiliation(s)
- Yudai Yamazaki
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tannoudai, Tsukuba, Ibaraki 305-8574, Japan
- Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Kazuya Suwabe
- Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
- Faculty of Health and Sport Sciences, Ryutsu Keizai University, 120 Ryugasaki, Ibaraki 301-0844, Japan
- Center for Cybernics Research, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
| | - Atsuko Nagano-Saito
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tannoudai, Tsukuba, Ibaraki 305-8574, Japan
- Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
- Department of Radiology, Ushiku Aiwa General Hospital, 896 Inoko-cho, Ushiku, Ibaraki 300-1296, Japan
| | - Kousaku Saotome
- Center for Cybernics Research, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
- Department of Radiological Sciences, School of Health Sciences, Fukushima Medical University, 1 Hikarigaoka, Fukushima, Fukushima 960-1295, Japan
| | - Ryuta Kuwamizu
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tannoudai, Tsukuba, Ibaraki 305-8574, Japan
- Graduate School of Letters, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Taichi Hiraga
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tannoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Ferenc Torma
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tannoudai, Tsukuba, Ibaraki 305-8574, Japan
- Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
| | - Kenji Suzuki
- Center for Cybernics Research, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
| | - Yoshiyuki Sankai
- Center for Cybernics Research, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8573, Japan
| | - Michael A Yassa
- Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92679-3800, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92679-3800, United States
| | - Hideaki Soya
- Corresponding author: Laboratory of Exercise Biochemistry and Neuroendocrinology; Sport Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8574, Japan.
| |
Collapse
|
14
|
The sensory and affective components of pain differentially shape pupillary dilatation during cold pressor tests. Auton Neurosci 2023; 246:103084. [PMID: 36934567 DOI: 10.1016/j.autneu.2023.103084] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Nociceptive and affective stimuli increase reflex sympathetic outflow to the pupils. To investigate effects of stimulus intensity, unpleasantness and distress on these pupillary reflexes, and to assess their stability, healthy participants immersed their hand in ice-water three times (for 20, 40 and 60 s; or 60, 40 and 20 s; or three times for 60 s) (N = 21 in each condition). Each ice-water immersion was preceded by a 90 s warm water immersion. To evaluate phasic sympathetic influences on pupil diameter, pupillary re-dilatation after 1 s of bright light was assessed during the last 10 s of each immersion. By-and-large, pain ratings and pupil diameter were greater during longer than shorter ice-water immersions, and ice-water immersions facilitated pupillary re-dilatation after the flash stimulus. However, mean pupil diameter during ice- and warm water immersions, minor ipsilateral amplification of the pupillary response, and ratings of pain unpleasantness and distress decreased across the experiment. Together, these findings suggest that nociceptive input increased sympathetic pupillary tone and amplified phasic increases in sympathetic activity after exposure to light. However, tonic sympathetic influences on pupil diameter and lateralization decreased across repeated immersions, possibly as novel or threatening aspects of the experience declined. Pupillary nociceptive and affective reflexes involve the locus coeruleus, an integral component of neural circuits that heighten cortical arousal and regulate pain. As these reflexes appear to reflect different aspects of sensory and affective processing, their combined assessment might increase the sensitivity and specificity of tests of locus coeruleus function in patients with suspected deficits.
Collapse
|
15
|
Wu Y, Chen L, Zhong F, Zhou K, Lu C, Cheng X, Wang S. Cognitive impairment in patients with heart failure: molecular mechanism and therapy. Heart Fail Rev 2023:10.1007/s10741-022-10289-9. [PMID: 36593370 DOI: 10.1007/s10741-022-10289-9] [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] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
Heart failure (HF) is associated with multiple organ dysfunction and many comorbidities. Its incidence is high among the elderly and is a major health burden worldwide. Cognitive impairment (CI) is highly prevalent in older patients with HF, which is an abnormality in one or more of the items of cognition, attention, memory, language, psychomotor function, and visual spatial acuity. Studies have shown that the incidence of CI in HF patients is between 13 and 54%, and patients with both conditions have poor self-care ability and prognosis, as well as increased mortality rates. However, the mechanisms of CI development in HF patients are still unclear. In this review, we describe the epidemiology and risk factors as well as measures of improving CI in HF patients. We update the latest pathophysiological mechanisms related to the neurocognitive changes in HF patients, expounding on the mechanisms associated with the development of CI in HF patients.
Collapse
Affiliation(s)
- Yanan Wu
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Liwen Chen
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, Guangdong, China
| | - Feng Zhong
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Kaiyi Zhou
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Chao Lu
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xiao Cheng
- Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sheng Wang
- Department of Anesthesiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Department of Anesthesiology, Guangdong Province, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China.
| |
Collapse
|
16
|
Engels-Domínguez N, Koops EA, Prokopiou PC, Van Egroo M, Schneider C, Riphagen JM, Singhal T, Jacobs HIL. State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: Challenges and opportunities. Neurosci Biobehav Rev 2023; 144:104998. [PMID: 36526031 PMCID: PMC9805533 DOI: 10.1016/j.neubiorev.2022.104998] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [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
|
17
|
Liebe T, Dordevic M, Kaufmann J, Avetisyan A, Skalej M, Müller N. Investigation of the functional pathogenesis of mild cognitive impairment by localisation-based locus coeruleus resting-state fMRI. Hum Brain Mapp 2022; 43:5630-5642. [PMID: 36441846 PMCID: PMC9704796 DOI: 10.1002/hbm.26039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/27/2022] [Accepted: 07/25/2022] [Indexed: 01/15/2023] Open
Abstract
Dementia as one of the most prevalent diseases urges for a better understanding of the central mechanisms responsible for clinical symptoms, and necessitates improvement of actual diagnostic capabilities. The brainstem nucleus locus coeruleus (LC) is a promising target for early diagnosis because of its early structural alterations and its relationship to the functional disturbances in the patients. In this study, we applied our improved method of localisation-based LC resting-state fMRI to investigate the differences in central sensory signal processing when comparing functional connectivity (fc) of a patient group with mild cognitive impairment (MCI, n = 28) and an age-matched healthy control group (n = 29). MCI and control participants could be differentiated in their Mini-Mental-State-Examination (MMSE) scores (p < .001) and LC intensity ratio (p = .010). In the fMRI, LC fc to anterior cingulate cortex (FDR p < .001) and left anterior insula (FDR p = .012) was elevated, and LC fc to right temporoparietal junction (rTPJ, FDR p = .012) and posterior cingulate cortex (PCC, FDR p = .021) was decreased in the patient group. Importantly, LC to rTPJ connectivity was also positively correlated to MMSE scores in MCI patients (p = .017). Furthermore, we found a hyperactivation of the left-insula salience network in the MCI patients. Our results and our proposed disease model shed new light on the functional pathogenesis of MCI by directing to attentional network disturbances, which could aid new therapeutic strategies and provide a marker for diagnosis and prediction of disease progression.
Collapse
Affiliation(s)
- Thomas Liebe
- Department of PsychiatryMedical University of ViennaViennaAustria
- Department of RadiologyUniversity Hospital JenaJenaGermany
- Department of PsychiatryUniversity Hospital JenaJenaGermany
- Clinical Affective Neuroimaging LaboratoryLeibniz Institute for NeurobiologyMagdeburgGermany
| | - Milos Dordevic
- Department of Degenerative and Chronic DiseasesUniversity PotsdamPotsdamGermany
| | - Jörn Kaufmann
- Department of NeurologyUniversity Hospital MagdeburgMagdeburgGermany
| | - Araks Avetisyan
- Neuroprotection LabGerman Center for Neurodegenerative Diseases (DZNE)MagdeburgGermany
| | - Martin Skalej
- Department of Neuroradiology, Clinic and Policlinic of RadiologyUniversity Hospital HalleHalleGermany
| | - Notger Müller
- Department of Degenerative and Chronic DiseasesUniversity PotsdamPotsdamGermany
| |
Collapse
|
18
|
Adcock KS, Lawlor B, Robertson IH, Vanneste S. Diminishing accelerated long-term forgetting in mild cognitive impairment: Study protocol for a prospective, double-blind, placebo-controlled, randomized controlled trial. Contemp Clin Trials Commun 2022; 30:100989. [PMID: 36117569 PMCID: PMC9478352 DOI: 10.1016/j.conctc.2022.100989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/12/2022] [Accepted: 08/28/2022] [Indexed: 11/24/2022] Open
Abstract
Background Harnessing the lifelong potential of the human brain for neuroplasticity may serve to maintain the viability of neural structures and postpone the onset of cognitive decline. The absence of effective pharmacological interventions to counter memory decline has encouraged scientists to test the possibility that noninvasive electrical stimulation may serve as an additional tool to improve memory abilities. Previous research showed that electrical stimulation of the greater occipital nerve enhances memory recall performance in young and older healthy subjects. This study aims to extend these findings to determine the effect of transcutaneous electrical stimulation of the greater occipital nerve on the improvement of episodic memory in individuals with amnestic Mild Cognitive Impairment (aMCI). Methods/design This study is a prospective, double-blind, placebo-controlled, randomized parallel-group study. A total of 100 individuals with a diagnosis of aMCI according to NIA/AA will be recruited. Participants will be randomly assigned to one of four groups. One group will receive active non-invasive transcutaneous electrical stimulation of greater occipital nerve (NITESGON), while three groups will serve as controls (i.e., sham NITESGON, active NITESGON with local anesthesia, and active NITESGON on the C5/C6 nerve). The primary outcome, i.e., memory recall, will be determined by a word association task, and will be recorded at baseline, 7 days after NITESGON, and 28 days after NITESGON. The secondary outcome is neurophysiological changes determined by resting state EEG and will be assessed immediately before and after NITESGON. Discussion The results will add new insights into improving episodic memory in individuals with aMCI. Trial registration #NCT05289804 (clinicaltrial.gov) Protocol approval id #SPREC102021-23 (Ethics Committee at Trinity College Dublin, School of Psychology)
Collapse
Affiliation(s)
- Katherine S Adcock
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Brian Lawlor
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Ian H Robertson
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Sven Vanneste
- Global Brain Health Institute, Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
Bachman SL, Nashiro K, Yoo H, Wang D, Thayer JF, Mather M. Associations between locus coeruleus MRI contrast and physiological responses to acute stress in younger and older adults. Brain Res 2022; 1796:148070. [PMID: 36088961 PMCID: PMC9805382 DOI: 10.1016/j.brainres.2022.148070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 01/03/2023]
Abstract
Acute stress activates the brain's locus coeruleus (LC)-noradrenaline system. Recent studies indicate that a magnetic resonance imaging (MRI)-based measure of LC structure is associated with better cognitive outcomes in later life. Yet despite the LC's documented role in promoting physiological arousal during acute stress, no studies have examined whether MRI-assessed LC structure is related to arousal responses to acute stress. In this study, 102 younger and 51 older adults completed an acute stress induction task while we assessed multiple measures of physiological arousal (heart rate, breathing rate, systolic and diastolic blood pressure, sympathetic tone, and heart rate variability, HRV). We used turbo spin echo MRI scans to quantify LC MRI contrast as a measure of LC structure. We applied univariate and multivariate approaches to assess how LC MRI contrast was associated with arousal at rest and during acute stress reactivity and recovery. In older participants, having higher caudal LC MRI contrast was associated with greater stress-related increases in systolic blood pressure and decreases in HRV, as well as lower HRV during recovery from acute stress. These results suggest that having higher caudal LC MRI contrast in older adulthood is associated with more pronounced physiological responses to acute stress. Further work is needed to confirm these patterns in larger samples of older adults.
Collapse
|
20
|
Li M, Liu S, Zhu H, Guo Z, Zhi Y, Liu R, Jiang Z, Liang X, Hu H, Zhu J. Decreased locus coeruleus signal associated with Alzheimer’s disease based on neuromelanin-sensitive magnetic resonance imaging technique. Front Neurosci 2022; 16:1014485. [PMID: 36278009 PMCID: PMC9580271 DOI: 10.3389/fnins.2022.1014485] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Objective Neuromelanin-sensitive magnetic resonance imaging (NM-MRI) technique was used to detect the changes of the locus coeruleus (LC) signals in Alzheimer’s disease patients (AD), and to analyze its correlation with cognitive function. Materials and methods A total of 27 patients with AD, 15 patients with mild cognitive impairment (MCI), and 25 healthy controls (HC) were examined by NM-MRI technique. ImageJ software was used to measure the LC signals. The locus coeruleus signal contrast ratios (LC-CRs) were calculated, along with the measurement of neuropsychological scales. Results The LC-CRs of AD patients were significantly different from that of HC (p = 0.007, 95% CI: −0.053∼−0.007). However, such significant differences were not observed between MCI and HC (p = 1.000, 95% CI: −0.030∼0.024), AD and MCI (p = 0.050, 95% CI: −0.054∼0.000). Furthermore, a significant positive correlation was identified between LC-CRs and MMSE sub item Drawing (r = 0.484, p = 0.011) in the AD group, MoCA sub item Attention (r = 0.519, p = 0.047) in the MCI group. The area under the curve of LC-CRs in the diagnosis of AD was 0.749 (p = 0.002, 95% CI: 0.618∼0.880), with a sensitivity of 85.2% and a specificity of 56.0%. Conclusion The NM-MRI technique could quantify the pathological degenerations of the LC in AD. Such LC degenerations can be employed to distinguish AD from healthy elderly.
Collapse
Affiliation(s)
- Meng Li
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shanwen Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongqin Zhu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Zhiwen Guo
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuqi Zhi
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rong Liu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhen Jiang
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoyun Liang
- Institute of Artificial Intelligence and Clinical Innovation, Neusoft Medical Systems Co., Ltd., Shanghai, China
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Hua Hu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Hua Hu,
| | - Jiangtao Zhu
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- *Correspondence: Jiangtao Zhu,
| |
Collapse
|
21
|
Elman JA, Puckett OK, Hagler DJ, Pearce RC, Fennema-Notestine C, Hatton SN, Lyons MJ, McEvoy LK, Panizzon MS, Reas ET, Dale AM, Franz CE, Kremen WS. Associations between MRI-assessed locus coeruleus integrity and cortical gray matter microstructure. Cereb Cortex 2022; 32:4191-4203. [PMID: 34969072 PMCID: PMC9528780 DOI: 10.1093/cercor/bhab475] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 01/27/2023] Open
Abstract
The locus coeruleus (LC) is one of the earliest sites of tau pathology, making it a key structure in early Alzheimer's disease (AD) progression. As the primary source of norepinephrine for the brain, reduced LC integrity may have negative consequences for brain health, yet macrostructural brain measures (e.g. cortical thickness) may not be sensitive to early stages of neurodegeneration. We therefore examined whether LC integrity was associated with differences in cortical gray matter microstructure among 435 men (mean age = 67.5; range = 62-71.7). LC structural integrity was indexed by contrast-to-noise ratio (LCCNR) from a neuromelanin-sensitive MRI scan. Restriction spectrum imaging (RSI), an advanced multi-shell diffusion technique, was used to characterize cortical microstructure, modeling total diffusion in restricted, hindered, and free water compartments. Higher LCCNR (greater integrity) was associated with higher hindered and lower free water diffusion in multiple cortical regions. In contrast, no associations between LCCNR and cortical thickness survived correction. Results suggest lower LC integrity is associated with patterns of cortical microstructure that may reflect a reduction in cytoarchitectural barriers due to broader neurodegenerative processes. These findings highlight the potential utility for LC imaging and advanced diffusion measures of cortical microstructure in assessing brain health and early identification of neurodegenerative processes.
Collapse
Affiliation(s)
- Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Rahul C Pearce
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
| | - Sean N Hatton
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Linda K McEvoy
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, CA 92093, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - Emilie T Reas
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA 92093, USA
- Center of Excellence for Stress and Mental Health, VA San Diego Health Care System, La Jolla, CA 92161, USA
| |
Collapse
|
22
|
Matti N, Javanshiri K, Haglund M, Saenz-Sardá X, Englund E. Locus Coeruleus Degeneration Differs Between Frontotemporal Lobar Degeneration Subtypes. J Alzheimers Dis 2022; 89:463-471. [PMID: 35871340 PMCID: PMC9535600 DOI: 10.3233/jad-220276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Background: There are few studies on the locus coeruleus (LC) in frontotemporal lobar degeneration (FTLD) and the potential differences in the LC related to the underlying proteinopathy. Objective: The aim of this study was to investigate the LC in FTLD subgroups. Methods: Neuropathological cases diagnosed with FTLD were included. The subgroups consisted of FTLD with tau, transactive response DNA-binding protein 43 (TDP) and fused in sarcoma (FUS). Micro- and macroscopical degeneration of the LC were assessed with respect to the number of neurons and the degree of depigmentation. A group of cognitively healthy subjects and a group with vascular cognitive impairment (VCI) served as comparison groups. Results: A total of 85 FTLD cases were included, of which 44 had FTLD-TDP, 38 had FTLD-tau, and three had FTLD-FUS. The groups were compared with 25 VCI cases and 41 cognitively healthy control cases (N = 151 for the entire study). All FTLD groups had a statistically higher microscopical degeneration of the LC compared to the controls, but the FTLD-tau group had greater micro- and macroscopical degeneration than the FTLD-TDP group. Age correlated positively with the LC score in the FTLD-tau group, but not in the FTLD-TDP group. Conclusion: A greater microscopical degeneration of the LC was observed in all FTLD cases compared to healthy controls and those with VCI. The LC degeneration was more severe in FTLD-tau than in FTLD-TDP. The macroscopically differential degeneration of the LC in FTLD subgroups may facilitate differential diagnostics, potentially with imaging.
Collapse
Affiliation(s)
- Nathalie Matti
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Keivan Javanshiri
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Mattias Haglund
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Xavier Saenz-Sardá
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| |
Collapse
|
23
|
David MCB, Del Giovane M, Liu KY, Gostick B, Rowe JB, Oboh I, Howard R, Malhotra PA. Cognitive and neuropsychiatric effects of noradrenergic treatment in Alzheimer's disease: systematic review and meta-analysis. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2022-329136. [PMID: 35790417 PMCID: PMC9484390 DOI: 10.1136/jnnp-2022-329136] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/19/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Dysfunction of the locus coeruleus-noradrenergic system occurs early in Alzheimer's disease, contributing to cognitive and neuropsychiatric symptoms in some patients. This system offers a potential therapeutic target, although noradrenergic treatments are not currently used in clinical practice. OBJECTIVE To assess the efficacy of drugs with principally noradrenergic action in improving cognitive and neuropsychiatric symptoms in Alzheimer's disease. METHODS The MEDLINE, Embase and ClinicalTrials.gov databases were searched from 1980 to December 2021. We generated pooled estimates using random effects meta-analyses. RESULTS We included 19 randomised controlled trials (1811 patients), of which six were judged as 'good' quality, seven as 'fair' and six 'poor'. Meta-analysis of 10 of these studies (1300 patients) showed a significant small positive effect of noradrenergic drugs on global cognition, measured using the Mini-Mental State Examination or Alzheimer's Disease Assessment Scale-Cognitive Subscale (standardised mean difference (SMD): 0.14, 95% CI: 0.03 to 0.25, p=0.01; I2=0%). No significant effect was seen on measures of attention (SMD: 0.01, 95% CI: -0.17 to 0.19, p=0.91; I2=0). The apathy meta-analysis included eight trials (425 patients) and detected a large positive effect of noradrenergic drugs (SMD: 0.45, 95% CI: 0.16 to 0.73, p=0.002; I2=58%). This positive effect was still present following removal of outliers to account for heterogeneity across studies. DISCUSSION Repurposing of established noradrenergic drugs is most likely to offer effective treatment in Alzheimer's disease for general cognition and apathy. However, several factors should be considered before designing future clinical trials. These include targeting of appropriate patient subgroups and understanding the dose effects of individual drugs and their interactions with other treatments to minimise risks and maximise therapeutic effects. PROSPERO REGISTERATION NUMBER CRD42021277500.
Collapse
Affiliation(s)
- Michael C B David
- Imperial College London and the University of Surrey, UK Dementia Research Institute Care Research and Technology Centre, London, UK
- Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Clinical Neurosciences, Charing Cross Hospital, London, UK
| | - Martina Del Giovane
- Imperial College London and the University of Surrey, UK Dementia Research Institute Care Research and Technology Centre, London, UK
- Brain Sciences, Imperial College London, London, UK
| | - Kathy Y Liu
- Division of Psychiatry, University College London, London, UK
| | | | - James Benedict Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Imafidon Oboh
- South West London and St George's Mental Health NHS Trust, London, UK
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| | - Paresh A Malhotra
- Imperial College London and the University of Surrey, UK Dementia Research Institute Care Research and Technology Centre, London, UK
- Brain Sciences, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Clinical Neurosciences, Charing Cross Hospital, London, UK
| |
Collapse
|
24
|
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: 0.5] [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]
|
25
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
26
|
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: 13] [Impact Index Per Article: 6.5] [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
|
27
|
David M, Malhotra PA. New approaches for the quantification and targeting of noradrenergic dysfunction in Alzheimer's disease. Ann Clin Transl Neurol 2022; 9:582-596. [PMID: 35293158 PMCID: PMC8994981 DOI: 10.1002/acn3.51539] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/14/2022] Open
Abstract
There is clear, early noradrenergic dysfunction in Alzheimer's disease. This is likely secondary to pathological tau deposition in the locus coeruleus, the pontine nucleus that produces and releases noradrenaline, prior to involvement of cortical brain regions. Disruption of noradrenergic pathways affects cognition, especially attention, impacting memory and broader functioning. Additionally, it leads to autonomic and neuropsychiatric symptoms. Despite the strong evidence of noradrenergic involvement in Alzheimer's, there are no clear trial data supporting the clinical use of any noradrenergic treatments. Several approaches have been tried, including proof-of-principle studies and (mostly small scale) randomised controlled trials. Treatments have included pharmacotherapies as well as stimulation. The lack of clear positive findings is likely secondary to limitations in gauging locus coeruleus integrity and dysfunction at an individual level. However, the recent development of several novel biomarkers holds potential and should allow quantification of dysfunction. This may then inform inclusion criteria and stratification for future trials. Imaging approaches have improved greatly following the development of neuromelanin-sensitive sequences, enabling the use of structural MRI to estimate locus coeruleus integrity. Additionally, functional MRI scanning has the potential to quantify network dysfunction. As well as neuroimaging, EEG, fluid biomarkers and pupillometry techniques may prove useful in assessing noradrenergic tone. Here, we review the development of these biomarkers and how they might augment clinical studies, particularly randomised trials, through identification of patients most likely to benefit from treatment. We outline the biomarkers with most potential, and how they may transform symptomatic therapy for people living with Alzheimer's disease.
Collapse
Affiliation(s)
- Michael David
- Imperial College London and the University of SurreyUK Dementia Research Institute Care Research and Technology CentreSir Michael Uren Hub, 86 Wood LaneLondonW12 0BZUK
- Imperial College London, Brain SciencesSouth KensingtonLondonSW7 2AZUK
- Imperial College Healthcare NHS Trust, Clinical NeurosciencesCharing Cross HospitalLondonW2 1NYUK
| | - Paresh A. Malhotra
- Imperial College London and the University of SurreyUK Dementia Research Institute Care Research and Technology CentreSir Michael Uren Hub, 86 Wood LaneLondonW12 0BZUK
- Imperial College London, Brain SciencesSouth KensingtonLondonSW7 2AZUK
- Imperial College Healthcare NHS Trust, Clinical NeurosciencesCharing Cross HospitalLondonW2 1NYUK
| |
Collapse
|
28
|
Dahl MJ, Mather M, Werkle-Bergner M, Kennedy BL, Guzman S, Hurth K, Miller CA, Qiao Y, Shi Y, Chui HC, Ringman JM. Locus coeruleus integrity is related to tau burden and memory loss in autosomal-dominant Alzheimer's disease. Neurobiol Aging 2022; 112:39-54. [PMID: 35045380 PMCID: PMC8976827 DOI: 10.1016/j.neurobiolaging.2021.11.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 11/17/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022]
Abstract
Abnormally phosphorylated tau, an indicator of Alzheimer's disease, accumulates in the first decades of life in the locus coeruleus (LC), the brain's main noradrenaline supply. However, technical challenges in in-vivo assessments have impeded research into the role of the LC in Alzheimer's disease. We studied participants with or known to be at-risk for mutations in genes causing autosomal-dominant Alzheimer's disease (ADAD) with early onset, providing a unique window into the pathogenesis of Alzheimer's largely disentangled from age-related factors. Using high-resolution MRI and tau PET, we found lower rostral LC integrity in symptomatic participants. LC integrity was associated with individual differences in tau burden and memory decline. Post-mortem analyses in a separate set of carriers of the same mutation confirmed substantial neuronal loss in the LC. Our findings link LC degeneration to tau burden and memory in Alzheimer's, and highlight a role of the noradrenergic system in this neurodegenerative disease.
Collapse
Affiliation(s)
- Martin J Dahl
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany; Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| | - Mara Mather
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Markus Werkle-Bergner
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Briana L Kennedy
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; School of Psychological Science, University of Western Australia, Perth, Australia
| | - Samuel Guzman
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kyle Hurth
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yuchuan Qiao
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yonggang Shi
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
29
|
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: 12.5] [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
|
30
|
Chen Y, Chen T, Hou R. Locus coeruleus in the pathogenesis of Alzheimer's disease: A systematic review. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12257. [PMID: 35282658 PMCID: PMC8900465 DOI: 10.1002/trc2.12257] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 01/22/2023]
Abstract
The locus coeruleus (LC) is a nucleus in the brain stem producing noradrenaline. While cognitive decline in Alzheimer's disease (AD) has primarily been related to cholinergic depletion, evidence indicates extensive LC degeneration as its earliest pathological marker. The current study aimed to systematically evaluate current evidence investigating the role of the LC in the pathogenesis of AD. A systematic search of the literature was performed on electronic databases including PubMed and Web of Science. Twelve animal, human post mortem, and human imaging studies were included in this review. Screening, data extraction, and quality assessment were undertaken following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines for preferred reporting of systematic reviews. Significant associations were identified between LC changes and cognitive decline. Significant reductions in fiber density, neuronal number, and LC volume were seen to correlate with other pathological degenerative markers. Current evidence indicates an important role of the LC in pathogenesis of AD and suggests its potential in both diagnosis and treatment of AD. This systematic review advances our understanding of the role of the LC in AD by synthesizing available evidence, identifying research gaps, highlighting methodological challenges, and making recommendations for future work.
Collapse
Affiliation(s)
- Yuqing Chen
- School of Clinical MedicineAddenbrooke's HospitalUniversity of CambridgeCambridgeUK
| | - Teng Chen
- Department of NeurosurgeryQilu Hospital of Shandong UniversityJinanShandongChina
| | - Ruihua Hou
- Clinical and Experimental SciencesFaculty of MedicineUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
31
|
Berger A, Vespa S, Dricot L, Dumoulin M, Iachim E, Doguet P, Vandewalle G, El Tahry R. How Is the Norepinephrine System Involved in the Antiepileptic Effects of Vagus Nerve Stimulation? Front Neurosci 2021; 15:790943. [PMID: 34924947 PMCID: PMC8675889 DOI: 10.3389/fnins.2021.790943] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/11/2021] [Indexed: 01/09/2023] Open
Abstract
Vagus Nerve Stimulation (VNS) is an adjunctive treatment for patients suffering from inoperable drug-resistant epilepsy. Although a complete understanding of the mediators involved in the antiepileptic effects of VNS and their complex interactions is lacking, VNS is known to trigger the release of neurotransmitters that have seizure-suppressing effects. In particular, norepinephrine (NE) is a neurotransmitter that has been associated with the clinical effects of VNS by preventing seizure development and by inducing long-term plastic changes that could restore a normal function of the brain circuitry. However, the biological requisites to become responder to VNS are still unknown. In this review, we report evidence of the critical involvement of NE in the antiepileptic effects of VNS in rodents and humans. Moreover, we emphasize the hypothesis that the functional integrity of the noradrenergic system could be a determining factor to obtain clinical benefits from the therapy. Finally, encouraging avenues of research involving NE in VNS treatment are discussed. These could lead to the personalization of the stimulation parameters to maximize the antiepileptic effects and potentially improve the response rate to the therapy.
Collapse
Affiliation(s)
- Alexandre Berger
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Synergia Medical SA, Mont-Saint-Guibert, Belgium.,GIGA-Cyclotron Research Center-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Simone Vespa
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Laurence Dricot
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Manon Dumoulin
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
| | - Evelina Iachim
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Department of Pediatric Neurology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Gilles Vandewalle
- GIGA-Cyclotron Research Center-In Vivo Imaging, University of Liège, Liège, Belgium
| | - Riëm El Tahry
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium.,Center for Refractory Epilepsy, Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
32
|
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: 23] [Impact Index Per Article: 7.7] [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
|
33
|
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: 20] [Impact Index Per Article: 6.7] [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
|
34
|
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: 2.3] [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
|
35
|
Liu J, Tao J, Xia R, Li M, Huang M, Li S, Chen X, Wilson G, Park J, Zheng G, Chen L, Kong J. Mind-Body Exercise Modulates Locus Coeruleus and Ventral Tegmental Area Functional Connectivity in Individuals With Mild Cognitive Impairment. Front Aging Neurosci 2021; 13:646807. [PMID: 34194314 PMCID: PMC8236862 DOI: 10.3389/fnagi.2021.646807] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Mild cognitive impairment (MCI) is a common global health problem. Recently, the potential of mind-body intervention for MCI has drawn the interest of investigators. This study aims to comparatively explore the modulation effect of Baduanjin, a popular mind-body exercise, and physical exercise on the cognitive function, as well as the norepinephrine and dopamine systems using the resting state functional connectivity (rsFC) method in patients with MCI. 69 patients were randomized to the Baduanjin, brisk walking, or healthy education control group for 6 months. The Montreal Cognitive Assessment (MoCA) and magnetic resonance imaging (MRI) scans were applied at baseline and at the end of the experiment. Results showed that (1) compared to the brisk walking, the Baduanjin significantly increased MoCA scores; (2) Baduanjin significantly increased the right locus coeruleus (LC) and left ventral tegmental area (VTA) rsFC with the right insula and right amygdala compared to that of the control group; and the right anterior cingulate cortex (ACC) compared to that of the brisk walking group; (3) the increased right LC-right insula rsFC and right LC-right ACC rsFC were significantly associated with the corresponding MoCA score after 6-months of intervention; (4) both exercise groups experienced an increased effective connectivity from the right ACC to the left VTA compared to the control group; and (5) Baduanjin group experienced an increase in gray matter volume in the right ACC compared to the control group. Our results suggest that Baduanjin can significantly modulate intrinsic functional connectivity and the influence of the norepinephrine (LC) and dopamine (VTA) systems. These findings may shed light on the mechanisms of mind-body intervention and aid the development of new treatments for MCI.
Collapse
Affiliation(s)
- Jiao Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States.,Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation, Fujian University of Traditional Chinese Medicine, Ministry of Education, Fuzhou, China
| | - Rui Xia
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Moyi Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Maomao Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shuzhen Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiangli Chen
- Department of Rehabilitation Psychology and Special Education, University of Wisconsin, Madison, WI, United States
| | - Georgia Wilson
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Joe Park
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| | - Guohua Zheng
- School of Nursing and Health Management, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
36
|
Ekuban FA, Zong C, Takikawa M, Morikawa K, Sakurai T, Ichihara S, Itoh K, Yamamoto M, Ohsako S, Ichihara G. Genetic ablation of Nrf2 exacerbates neurotoxic effects of acrylamide in mice. Toxicology 2021; 456:152785. [PMID: 33872730 DOI: 10.1016/j.tox.2021.152785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/20/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Acrylamide (ACR), a recognized neurotoxicant in humans and experimental animals, is widely used in industry and in food generated through Maillard reaction. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of the cellular defense system and activates antioxidants and cytoprotective genes. The exact roles of Nrf2 in environmental electrophile-induced neurotoxicity is poorly understood. The aim of this study was to determine the roles of Nrf2 in ACR-induced neurotoxicity including degeneration of monoaminergic axons and sensorimotor dysfunction. Male 10-week-old C57BL/6JJcl Nrf2-knockout mice and wild type (WT) counterparts were each divided into four groups of 12 and provided with drinking water containing acrylamide at 0, 67, 110 or 200 ppm for four weeks. The effects of acrylamide were examined by landing foot spread test, immunohistochemistry for noradrenaline (NA) and serotonin (5-HT)-containing axons and Iba1-positive microglia in the prefrontal cortex as well as quantitative real-time polymerase chain reaction (qRT-PCR) on antioxidant, proinflammatory and anti-inflammatory genes in the prefrontal cortex. Relative to the wild type, exposure of Nrf2-knockout mice to acrylamide increased hindlimb splay length, microglial area and process length as well as decreasing the density of NA and 5-HT-immunoreactive axons to a greater extent. Moreover, deletion of Nrf2 gene suppressed acrylamide-induced mRNA upregulation of Nrf2-antioxidants, NAD(P): quinone oxidoreductase 1 (NQO1), superoxide dismutase-1 (SOD-1) and heme oxygenase-1 (HO-1) as well as anti-inflammatory markers such as, arginase-1 (Arg1), found in the inflammatory zone-1 (Fizz1), chitinase-like 3 (Chi3l3), interleukin-4 receptor alpha (IL-4Rα), cluster of differentiation 206 (CD206) and transforming growth factor beta-1 (TGFβ1) while enhancing acrylamide-induced upregulation of pro-inflammatory cytokines, interleukin-1 beta (IL-1β), tumor necrosis-alpha (TNF-α) and inducible nitric oxide synthase (iNOS) in the prefrontal cortex. The results demonstrate susceptibility of mice lacking the Nrf2 gene to acrylamide-induced neurotoxicity and neuroinflammation with the activation of microglia. Moreover, the results suggest the role of Nrf2 not only in induction of antioxidant gene expression, but also in suppression of proinflammatory cytokine gene expression.
Collapse
Affiliation(s)
- Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan
| | - Cai Zong
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan
| | - Madoka Takikawa
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan
| | - Kota Morikawa
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan
| | - Toshihiro Sakurai
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Ken Itoh
- Department of Stress Response Hirosaki University, Hirosaki, Japan
| | - Masayuki Yamamoto
- Department of Molecular Biochemistry Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Seiichiroh Ohsako
- Laboratory of Environmental Health Sciences, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo University of Science, Noda, Japan.
| |
Collapse
|
37
|
Liu KY, Acosta-Cabronero J, Hong YT, Yi YJ, Hämmerer D, Howard R. FDG-PET assessment of the locus coeruleus in Alzheimer's disease. NEUROIMAGE. REPORTS 2021; 1:100002. [PMID: 34396361 PMCID: PMC8262255 DOI: 10.1016/j.ynirp.2020.100002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Sensitive and reliable in vivo imaging of the locus coeruleus (LC) is important to develop and evaluate its potential as a biomarker in neurodegenerative diseases such as Alzheimer's disease (AD). It is not known whether AD-related alterations in LC integrity can be detected using 18F-labelled fluoro-2-deoxyglucose (FDG) positron emission tomography (PET). Mean FDG-PET images from AD patients (N = 193) and controls (N = 256) from the ADNI database were co-registered to a study-wise anatomical template. Regional LC median standardized uptake value ratio (SUVR) values were obtained using four previously published LC masks and normalized to values from pons and cerebellar vermis reference regions. To support the validity of our methods, other regions previously reported to be most and least affected metabolically in AD were also compared to controls. The LC did not show between-group differences in FDG-PET signal, whereas the mammillary bodies did, despite these regions having comparable volumes and SUVR ranges. Brain regions previously reported to be most and least affected metabolically in AD compared to controls showed medium-to-large and small effect sizes for SUVR differences respectively. The results do not support the current application of LC FDG-PET signal as an in vivo biomarker for AD. Methodological and demographic factors potentially contributing to these findings are discussed. Future research may investigate age-related differences in LC FDG-PET signal and higher resolution images to fully explore its biomarker potential.
Collapse
Affiliation(s)
- Kathy Y. Liu
- Division of Psychiatry, University College London, UK
| | - Julio Acosta-Cabronero
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, University College London, UK
- Tenoke Ltd., Cambridge, UK
| | - Young T. Hong
- Wolfson Brain Imaging Centre, University of Cambridge, UK
| | - Yeo-Jin Yi
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Dorothea Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, UK
| | - Robert Howard
- Division of Psychiatry, University College London, UK
| | | |
Collapse
|
38
|
Kusama M, Sato N, Kimura Y, Miyagi K. Quick MR Neuromelanin Imaging Using a Chemical Shift Selective Pulse. Magn Reson Med Sci 2021; 20:106-111. [PMID: 32074593 PMCID: PMC7952205 DOI: 10.2463/mrms.tn.2019-0167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Not only magnetization transfer contrast (MTC) pulse, but also chemical shift selective (CHESS) pulse would be a useful additional one for shortening the scan time of neuromelanin imaging. We compared three sequences among turbo-spin echo (TSE) images with CHESS, MTC, and without an additional pulse in the same short time, 3 min 20 s. The TSE with CHESS image was the most useful for the diagnosis of neuromelanin within the limited time.
Collapse
Affiliation(s)
- Midori Kusama
- Department of Radiology, National Center of Neurology and Psychiatry
| | - Noriko Sato
- Department of Radiology, National Center of Neurology and Psychiatry
| | - Yukio Kimura
- Department of Radiology, National Center of Neurology and Psychiatry
| | - Kenji Miyagi
- Department of Radiology, National Center of Neurology and Psychiatry
| |
Collapse
|
39
|
Guinea-Izquierdo A, Giménez M, Martínez-Zalacaín I, Del Cerro I, Canal-Noguer P, Blasco G, Gascón J, Reñé R, Rico I, Camins A, Aguilera C, Urretavizcaya M, Ferrer I, Menchón JM, Soria V, Soriano-Mas C. Lower Locus Coeruleus MRI intensity in patients with late-life major depression. PeerJ 2021; 9:e10828. [PMID: 33628639 PMCID: PMC7894108 DOI: 10.7717/peerj.10828] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/04/2021] [Indexed: 11/24/2022] Open
Abstract
Background The locus coeruleus (LC) is the major noradrenergic source in the central nervous system. Structural alterations in the LC contribute to the pathophysiology of different neuropsychiatric disorders, which may increase to a variable extent the likelihood of developing neurodegenerative conditions. The characterization of such alterations may therefore help to predict progression to neurodegenerative disorders. Despite the LC cannot be visualized with conventional magnetic resonance imaging (MRI), specific MRI sequences have been developed to infer its structural integrity. Methods We quantified LC signal Contrast Ratios (LCCRs) in late-life major depressive disorder (MDD) (n = 37, 9 with comorbid aMCI), amnestic Mild Cognitive Impairment (aMCI) (n = 21, without comorbid MDD), and healthy controls (HCs) (n = 31), and also assessed the putative modulatory effects of comorbidities and other clinical variables. Results LCCRs were lower in MDD compared to aMCI and HCs. While no effects of aMCI comorbidity were observed, lower LCCRs were specifically observed in patients taking serotonin/norepinephrine reuptake inhibitors (SNRIs). Conclusion Our results do not support the hypothesis that lower LCCRs characterize the different clinical groups that may eventually develop a neurodegenerative disorder. Conversely, our results were specifically observed in patients with late-life MDD taking SNRIs. Further research with larger samples is warranted to ascertain whether medication or particular clinical features of patients taking SNRIs are associated with changes in LC neurons.
Collapse
Affiliation(s)
- Andrés Guinea-Izquierdo
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain
| | - Mónica Giménez
- Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain
| | - Ignacio Martínez-Zalacaín
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain
| | - Inés Del Cerro
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
| | - Pol Canal-Noguer
- B2SLab/Departament d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, Barcelona, Spain.,Networking Biomedical Research Centre in the subject area of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Institut de Recerca Pediàtrica, Hospital Sant Joan de Déu, Esplugues de Llobregat (Barcelona), Spain
| | - Gerard Blasco
- Imaging Diagnostic Institute (IDI), Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Jordi Gascón
- Dementia Diagnostic and Treatment Unit/Department of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Ramon Reñé
- Dementia Diagnostic and Treatment Unit/Department of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Inmaculada Rico
- Dementia Diagnostic and Treatment Unit/Department of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Angels Camins
- Imaging Diagnostic Institute (IDI), Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Carlos Aguilera
- Imaging Diagnostic Institute (IDI), Bellvitge University Hospital, Hospitalet de Llobregat (Barcelona), Spain
| | - Mikel Urretavizcaya
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
| | - Isidre Ferrer
- Department of Pathology and Experimental Therapeutics/Institute of Neurosciences, University of Barcelona, Hospitalet de Llobregat (Barcelona), Spain.,Department of Pathologic Anatomy/Bellvitge University Hospital, Bellvitge Biomedical Research Institute-IDIBELL, Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Neurodegenerative diseases (CIBERNED), Madrid, Spain
| | - José Manuel Menchón
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
| | - Virginia Soria
- Department of Clinical Sciences/School of Medicine, University of Barcelona, Barcelona, Spain.,Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain
| | - Carles Soriano-Mas
- Department of Psychiatry/Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat (Barcelona), Spain.,Network Center for Biomedical Research on Mental Health (CIBERSAM), Madrid, Spain.,Department of Psychobiology and Methodology in Health Sciences, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| |
Collapse
|
40
|
Elman JA, Puckett OK, Beck A, Fennema-Notestine C, Cross LK, Dale AM, Eglit GML, Eyler LT, Gillespie NA, Granholm EL, Gustavson DE, Hagler DJ, Hatton SN, Hauger R, Jak AJ, Logue MW, McEvoy LK, McKenzie RE, Neale MC, Panizzon MS, Reynolds CA, Sanderson-Cimino M, Toomey R, Tu XM, Whitsel N, Williams ME, Xian H, Lyons MJ, Franz CE, Kremen WS. MRI-assessed locus coeruleus integrity is heritable and associated with multiple cognitive domains, mild cognitive impairment, and daytime dysfunction. Alzheimers Dement 2021; 17:1017-1025. [PMID: 33580733 PMCID: PMC8248066 DOI: 10.1002/alz.12261] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 12/22/2022]
Abstract
Introduction The locus coeruleus (LC) undergoes extensive neurodegeneration in early Alzheimer's disease (AD). The LC is implicated in regulating the sleep–wake cycle, modulating cognitive function, and AD progression. Methods Participants were 481 men (ages 62 to 71.7) from the Vietnam Era Twin Study of Aging. LC structural integrity was indexed by neuromelanin‐sensitive magnetic resonance imaging (MRI) contrast‐to‐noise ratio (LCCNR). We examined LCCNR, cognition, amnestic mild cognitive impairment (aMCI), and daytime dysfunction. Results Heritability of LCCNR was .48. Participants with aMCI showed greater daytime dysfunction. Lower LCCNR was associated with poorer episodic memory, general verbal fluency, semantic fluency, and processing speed, as well as increased odds of aMCI and greater daytime dysfunction. Discussion Reduced LC integrity is associated with widespread differences across cognitive domains, daytime sleep‐related dysfunction, and risk for aMCI. These findings in late‐middle‐aged adults highlight the potential of MRI‐based measures of LC integrity in early identification of AD risk.
Collapse
Affiliation(s)
- Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Asad Beck
- Graduate Program in Neuroscience, University of Washington, Seattle, Washington, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Latonya K Cross
- Department of Psychology, University of Hawaii Hilo, Hilo, Hawaii, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California, USA.,Department of Neuroscience, University of California San Diego, La Jolla, California, USA
| | - Graham M L Eglit
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Desert Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, California, USA
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eric L Granholm
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| | - Daniel E Gustavson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Sean N Hatton
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA.,Department of Neuroscience, University of California San Diego, La Jolla, California, USA
| | - Richard Hauger
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| | - Amy J Jak
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| | - Mark W Logue
- National Center for PTSD: Behavioral Science Division, VA Boston Healthcare System, Boston, Massachusetts, USA.,Department of Psychiatry and the Biomedical Genetics Section, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Linda K McEvoy
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Ruth E McKenzie
- School of Education and Public Policy, Merrimack College, Andover, Massachusetts, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - Chandra A Reynolds
- Department of Psychology, University of California Riverside, Riverside, California, USA
| | - Mark Sanderson-Cimino
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA.,Joint Doctoral Program in Clinical Psychology, San Diego State/University of California, San Diego, California, USA
| | - Rosemary Toomey
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Xin M Tu
- Family Medicine and Public Health, University of California San Diego, La Jolla, California, USA
| | - Nathan Whitsel
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - McKenna E Williams
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA.,Joint Doctoral Program in Clinical Psychology, San Diego State/University of California, San Diego, California, USA
| | - Hong Xian
- Department of Epidemiology & Biostatistics, St. Louis University, St. Louis, Missouri, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA.,Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California, USA.,VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
41
|
Hou R, Beardmore R, Holmes C, Osmond C, Darekar A. A case-control study of the locus coeruleus degeneration in Alzheimer's disease. Eur Neuropsychopharmacol 2021; 43:153-159. [PMID: 33431221 DOI: 10.1016/j.euroneuro.2020.12.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 12/16/2020] [Accepted: 12/31/2020] [Indexed: 01/04/2023]
Abstract
The locus coeruleus (LC) is the major source of noradrenaline, which plays a key role in cognition. We aimed to detect the extent of the LC signal attenuation in Alzheimer's disease (AD) patients using a neuromelanin (NM)-sensitive MRI and how it may correlate with inflammatory and autonomic measures. An individually matched case-control study design was employed. 24 patients with AD and 24 age and gender matched controls with no cognitive impairment were recruited. The primary outcome measure was the LC signal intensity indicated by the LC contrast ratio (CR) and measured by the NM-sensitive MRI. Secondary outcome measures included neuropsychometric tests of cognitive state, peripheral inflammatory and autonomic measures. Conditional logistic regression analysis revealed a significant 22% LC-CR reduction in the AD group compared with the control group. However, there was no statistical significance from inflammatory or autonomic measures. This is the largest individually-matched case-control study to visualise the LC degeneration in AD patients. The study revealed significant LC degeneration which holds promise to stratify patients who may benefit from treatment targeting noradrenergic dysfunction.
Collapse
Affiliation(s)
- Ruihua Hou
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, 4-12 Terminus Terrace, Southampton SO14 3DT, United Kingdom.
| | - Rebecca Beardmore
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, 4-12 Terminus Terrace, Southampton SO14 3DT, United Kingdom
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, 4-12 Terminus Terrace, Southampton SO14 3DT, United Kingdom
| | - Clive Osmond
- Medical Research Council Lifecourse Epidemiology Unit, University of Southampton, United Kingdom
| | - Angela Darekar
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| |
Collapse
|
42
|
Babić Leko M, Hof PR, Šimić G. Alterations and interactions of subcortical modulatory systems in Alzheimer's disease. PROGRESS IN BRAIN RESEARCH 2021; 261:379-421. [PMID: 33785136 DOI: 10.1016/bs.pbr.2020.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) is not fully understood. Here we summarize current knowledge on the involvement of the serotonergic, noradrenergic, dopaminergic, cholinergic, and opioid systems in AD, emphasizing the importance of interactions between the serotonergic and the other subcortical modulatory systems during the progression of AD. In physiological conditions, all neurotransmitter systems function in concert and are interdependent at both the neuroanatomical and molecular levels. Through their early involvement in AD, cognitive and behavioral abilities that rely on their interactions also become disrupted. Considering that serotonin (5HT) regulates the release of noradrenaline (NA), dopamine (DA) and acetylcholine (ACh), any alteration in 5HT levels leads to disturbance of NA, DA, and ACh homeostasis in the brain. One of the earliest pathological changes during the prodromal phase of AD is a decrease of serotonergic transmission throughout the brain, with serotonergic receptors being also affected. Additionally, serotonergic and noradrenergic as well as serotonergic and dopaminergic nuclei are reciprocally interconnected. As the serotonergic dorsal raphe nucleus (DRN) is affected by pathological changes early in AD, and the noradrenergic locus coeruleus (LC) and dopaminergic ventral tegmental area (VTA) exhibit AD-related pathological changes, their connectivity also becomes altered in AD. Such disrupted interactions among neurotransmitter systems in AD can be used in the development of multi-target drugs. Some of the potential AD therapeutics (such as ASS234, RS67333, tropisetron) target multiple neurotransmitter systems to achieve the best possible improvement of cognitive and behavioral deficits observed in AD. Here, we review how serotonergic system interacts with other subcortical modulatory systems (noradrenergic, dopaminergic, cholinergic, and opioid systems) during AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Goran Šimić
- Department for Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia.
| |
Collapse
|
43
|
Beardmore R, Hou R, Darekar A, Holmes C, Boche D. The Locus Coeruleus in Aging and Alzheimer's Disease: A Postmortem and Brain Imaging Review. J Alzheimers Dis 2021; 83:5-22. [PMID: 34219717 PMCID: PMC8461706 DOI: 10.3233/jad-210191] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 12/21/2022]
Abstract
The locus coeruleus (LC), a tiny nucleus in the brainstem and the principal site of noradrenaline synthesis, has a major role in regulating autonomic function, arousal, attention, and neuroinflammation. LC dysfunction has been linked to a range of disorders; however particular interest is given to the role it plays in Alzheimer's disease (AD). The LC undergoes significant neuronal loss in AD, thought to occur early in the disease process. While neuronal loss in the LC has also been suggested to occur in aging, this relationship is less clear as the findings have been contradictory. LC density has been suggested to be indicative of cognitive reserve and the evidence for these claims will be discussed. Recent imaging techniques allowing visualization of the LC in vivo using neuromelanin-sensitive MRI are developing our understanding of the role of LC in aging and AD. Tau pathology within the LC is evident at an early age in most individuals; however, the relationship between tau accumulation and neuronal loss and why some individuals then develop AD is not understood. Neuromelanin pigment accumulates within LC cells with age and is proposed to be toxic and inflammatory when released into the extracellular environment. This review will explore our current knowledge of the LC changes in both aging and AD from postmortem, imaging, and experimental studies. We will discuss the reasons behind the susceptibility of the LC to neuronal loss, with a focus on the role of extracellular neuromelanin and neuroinflammation caused by the dysfunction of the LC-noradrenaline pathway.
Collapse
Affiliation(s)
- Rebecca Beardmore
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Memory Assessment and Research Centre, Moorgreen Hospital, Southern Health Foundation Trust, Southampton, UK
| | - Ruihua Hou
- Clinical and Experimental Sciences, Department of Psychiatry, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Angela Darekar
- Department of Medical Physics, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Clive Holmes
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Memory Assessment and Research Centre, Moorgreen Hospital, Southern Health Foundation Trust, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
44
|
Galgani A, Lombardo F, Della Latta D, Martini N, Bonuccelli U, Fornai F, Giorgi FS. Locus Coeruleus Magnetic Resonance Imaging in Neurological Diseases. Curr Neurol Neurosci Rep 2020; 21:2. [PMID: 33313963 PMCID: PMC7732795 DOI: 10.1007/s11910-020-01087-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW Locus coeruleus (LC) is the main noradrenergic nucleus of the brain, and its degeneration is considered to be key in the pathogenesis of neurodegenerative diseases. In the last 15 years,MRI has been used to assess LC in vivo, both in healthy subjects and in patients suffering from neurological disorders. In this review, we summarize the main findings of LC-MRI studies, interpreting them in light of preclinical and histopathological data, and discussing its potential role as diagnostic and experimental tool. RECENT FINDINGS LC-MRI findings were largely in agreement with neuropathological evidences; LC signal showed to be not significantly affected during normal aging and to correlate with cognitive performances. On the contrary, a marked reduction of LC signal was observed in patients suffering from neurodegenerative disorders, with specific features. LC-MRI is a promising tool, which may be used in the future to explore LC pathophysiology as well as an early biomarker for degenerative diseases.
Collapse
Affiliation(s)
| | - Francesco Lombardo
- U.O.C. "Risonanza Magnetica Specialistica e Neuroradiologia", Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | - Daniele Della Latta
- Deep Health Unit, Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | - Nicola Martini
- Deep Health Unit, Fondazione "G. Monasterio"- National Research Council/Tuscany Region, Pisa, Italy
| | | | - Francesco Fornai
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Filippo Sean Giorgi
- Neurology Unit, Pisa University Hospital, Pisa, Italy.
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy.
| |
Collapse
|
45
|
Sun W, Tang Y, Qiao Y, Ge X, Mather M, Ringman JM, Shi Y. A probabilistic atlas of locus coeruleus pathways to transentorhinal cortex for connectome imaging in Alzheimer's disease. Neuroimage 2020; 223:117301. [PMID: 32861791 PMCID: PMC7797167 DOI: 10.1016/j.neuroimage.2020.117301] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
According to the latest Braak staging of Alzheimer's disease (AD), tau pathology occurs earliest in the brain in the locus coeruleus (LC) of the brainstem, then propagates to the transentorhinal cortex (TEC), and later to other neocortical regions. Recent animal and in vivo human brain imaging research also support the trans-axonal propagation of tau pathology. In addition, neurochemical studies link norepinephrine to behavioral symptoms in AD. It is thus critical to examine the integrity of the LC-TEC pathway in studying the early development of the disease, but there has been limited work in this direction. By leveraging the high-resolution and multi-shell diffusion MRI data from the Human Connectome Project (HCP), in this work we develop a novel method for the reconstruction of the LC-TEC pathway in a cohort of 40 HCP subjects carefully selected based on rigorous quality control of the residual distortion artifacts in the brainstem. A probabilistic atlas of the LC-TEC pathway of both hemispheres is then developed in the MNI152 space and distributed publicly on the NITRC website. To apply our atlas on clinical imaging data, we develop an automated approach to calculate the medial core of the LC-TEC pathway for localized analysis of connectivity changes. In a cohort of 138 subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI), we demonstrate the detection of the decreased fiber integrity in the LC-TEC pathways with increasing disease severity.
Collapse
Affiliation(s)
- Wei Sun
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave., Los Angeles 90033, CA, USA
| | - Yuchun Tang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave., Los Angeles 90033, CA, USA
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuchuan Qiao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave., Los Angeles 90033, CA, USA
| | - Xinting Ge
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave., Los Angeles 90033, CA, USA
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - John M. Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yonggang Shi
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, 2025 Zonal Ave., Los Angeles 90033, CA, USA
| | | |
Collapse
|
46
|
Ye R, Rua C, O'Callaghan C, Jones PS, Hezemans FH, Kaalund SS, Tsvetanov KA, Rodgers CT, Williams G, Passamonti L, Rowe JB. An in vivo probabilistic atlas of the human locus coeruleus at ultra-high field. Neuroimage 2020; 225:117487. [PMID: 33164875 PMCID: PMC7779564 DOI: 10.1016/j.neuroimage.2020.117487] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/22/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022] Open
Abstract
Early and profound pathological changes are evident in the locus coeruleus (LC) in dementia and Parkinson's disease, with effects on arousal, attention, cognitive and motor control. The LC can be identified in vivo using non-invasive magnetic resonance imaging techniques which have potential as biomarkers for detecting and monitoring disease progression. Technical limitations of existing imaging protocols have impaired the sensitivity to regional contrast variance or the spatial variability on the rostrocaudal extent of the LC, with spatial mapping consistent with post mortem findings. The current study employs a sensitive magnetisation transfer sequence using ultrahigh field 7T MRI to investigate the LC structure in vivo at high-resolution (0.4 × 0.4 × 0.5 mm). Magnetisation transfer images from 53 healthy older volunteers (52 - 84 years) clearly revealed the spatial features of the LC and were used to create a probabilistic LC atlas for older adults. This atlas may be especially relevant for studying disorders associated with older age. To use the atlas does not require use of the same MT sequence of 7T MRI, provided good co-registration and normalisation is achieved. Consistent rostrocaudal gradients of slice-wise volume, contrast and variance along the LC were observed, mirroring distinctive ex vivo spatial distributions of LC cells in its subregions. The contrast-to-noise ratios were calculated for the peak voxels, and for the averaged signals within the atlas, to accommodate the volumetric differences in estimated contrast. The probabilistic atlas is freely available, and the MRI dataset will be made available for non-commercial research, for replication or to facilitate accurate LC localisation and unbiased contrast extraction in future studies.
Collapse
Affiliation(s)
- Rong Ye
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK.
| | - Catarina Rua
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Claire O'Callaghan
- Brain and Mind Centre and School of Medical Sciences, Faculty of Medicine, University of Sydney, Sydney, Australia; Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Simon Jones
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Frank H Hezemans
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Sanne S Kaalund
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK
| | - Kamen A Tsvetanov
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Department of Psychology, University of Cambridge, Cambridge, UK
| | | | - Guy Williams
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Luca Passamonti
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; Istituto di Bioimmagini e Fisiologia Molecolare (IBFM), Consiglio Nazionale delle Ricerche (CNR), Milano, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Herchel Smith Building for Brain and Mind Sciences, Forvie Site, Robinson Way, Cambridge CB2 0SZ, UK; MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Priovoulos N, van Boxel SCJ, Jacobs HIL, Poser BA, Uludag K, Verhey FRJ, Ivanov D. Unraveling the contributions to the neuromelanin-MRI contrast. Brain Struct Funct 2020; 225:2757-2774. [PMID: 33090274 PMCID: PMC7674382 DOI: 10.1007/s00429-020-02153-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
The Locus Coeruleus (LC) and the Substantia Nigra (SN) are small brainstem nuclei that change with aging and may be involved in the development of various neurodegenerative and psychiatric diseases. Magnetization Transfer (MT) MRI has been shown to facilitate LC and the SN visualization, and the observed contrast is assumed to be related to neuromelanin accumulation. Imaging these nuclei may have predictive value for the progression of various diseases, but interpretation of previous studies is hindered by the fact that the precise biological source of the contrast remains unclear, though several hypotheses have been put forward. To inform clinical studies on the possible biological interpretation of the LC- and SN contrast, we examined an agar-based phantom containing samples of natural Sepia melanin and synthetic Cys-Dopa-Melanin and compared this to the in vivo human LC and SN. T1 and T2* maps, MT spectra and relaxation times of the phantom, the LC and the SN were measured, and a two-pool MT model was fitted. Additionally, Bloch simulations and a transient MT experiment were conducted to confirm the findings. Overall, our results indicate that Neuromelanin-MRI contrast in the LC likely results from a lower macromolecular fraction, thus facilitating interpretation of results in clinical populations. We further demonstrate that in older individuals T1 lengthening occurs in the LC.
Collapse
Affiliation(s)
- Nikos Priovoulos
- School for Mental Health and Neuroscience, Alzheimer Center Limburg, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, Netherlands.
| | - Stan C J van Boxel
- School for Mental Health and Neuroscience, Alzheimer Center Limburg, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, Netherlands
| | - Heidi I L Jacobs
- School for Mental Health and Neuroscience, Alzheimer Center Limburg, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, Netherlands.,Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands.,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benedikt A Poser
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Kamil Uludag
- Center for Neuroscience Imaging Research, Institute for Basic Science and Department of Biomedical Engineering, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon, Republic of Korea.,Techna Institute and Koerner Scientist in MR Imaging, University Health Network, 121-100 College Street, Toronto, M5G 1L5, Canada
| | - Frans R J Verhey
- School for Mental Health and Neuroscience, Alzheimer Center Limburg, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, Netherlands
| | - Dimo Ivanov
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands.
| |
Collapse
|
48
|
Kelberman M, Keilholz S, Weinshenker D. What's That (Blue) Spot on my MRI? Multimodal Neuroimaging of the Locus Coeruleus in Neurodegenerative Disease. Front Neurosci 2020; 14:583421. [PMID: 33122996 PMCID: PMC7573566 DOI: 10.3389/fnins.2020.583421] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/16/2020] [Indexed: 01/04/2023] Open
Abstract
The locus coeruleus (LC) has long been underappreciated for its role in the pathophysiology of Alzheimer’s disease (AD), Parkinson’s disease (PD), and other neurodegenerative disorders. While AD and PD are distinct in clinical presentation, both are characterized by prodromal protein aggregation in the LC, late-stage degeneration of the LC, and comorbid conditions indicative of LC dysfunction. Many of these early studies were limited to post-mortem histological techniques due to the LC’s small size and location deep in the brainstem. Thus, there is a growing interest in utilizing in vivo imaging of the LC as a predictor of preclinical neurodegenerative processes and biomarker of disease progression. Simultaneously, neuroimaging in animal models of neurodegenerative disease holds promise for identifying early alterations to LC circuits, but has thus far been underutilized. While still in its infancy, a handful of studies have reported effects of single gene mutations and pathology on LC function in disease using various neuroimaging techniques. Furthermore, combining imaging and optogenetics or chemogenetics allows for interrogation of network connectivity in response to changes in LC activity. The purpose of this article is twofold: (1) to review what magnetic resonance imaging (MRI) and positron emission tomography (PET) have revealed about LC dysfunction in neurodegenerative disease and its potential as a biomarker in humans, and (2) to explore how animal models can be used to test hypotheses derived from clinical data and establish a mechanistic framework to inform LC-focused therapeutic interventions to alleviate symptoms and impede disease progression.
Collapse
Affiliation(s)
- Michael Kelberman
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| | - Shella Keilholz
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University, Atlanta, GA, United States
| | - David Weinshenker
- Department of Human Genetics, Emory University, Atlanta, GA, United States
| |
Collapse
|
49
|
Giorgi FS, Galgani A, Puglisi-Allegra S, Limanaqi F, Busceti CL, Fornai F. Locus Coeruleus and neurovascular unit: From its role in physiology to its potential role in Alzheimer's disease pathogenesis. J Neurosci Res 2020; 98:2406-2434. [PMID: 32875628 DOI: 10.1002/jnr.24718] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 08/08/2020] [Indexed: 12/15/2022]
Abstract
Locus coeruleus (LC) is the main noradrenergic (NA) nucleus of the central nervous system. LC degenerates early during Alzheimer's disease (AD) and NA loss might concur to AD pathogenesis. Aside from neurons, LC terminals provide dense innervation of brain intraparenchymal arterioles/capillaries, and NA modulates astrocyte functions. The term neurovascular unit (NVU) defines the strict anatomical/functional interaction occurring between neurons, glial cells, and brain vessels. NVU plays a fundamental role in coupling the energy demand of activated brain regions with regional cerebral blood flow, it includes the blood-brain barrier (BBB), plays an active role in neuroinflammation, and participates also to the glymphatic system. NVU alteration is involved in AD pathophysiology through several mechanisms, mainly related to a relative oligoemia in activated brain regions and impairment of structural and functional BBB integrity, which contributes also to the intracerebral accumulation of insoluble amyloid. We review the existing data on the morphological features of LC-NA innervation of the NVU, as well as its contribution to neurovascular coupling and BBB proper functioning. After introducing the main experimental data linking LC with AD, which have repeatedly shown a key role of neuroinflammation and increased amyloid plaque formation, we discuss the potential mechanisms by which the loss of NVU modulation by LC might contribute to AD pathogenesis. Surprisingly, thus far not so many studies have tested directly these mechanisms in models of AD in which LC has been lesioned experimentally. Clarifying the interaction of LC with NVU in AD pathogenesis may disclose potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Filippo Sean Giorgi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Neurology Unit, Pisa University Hospital, Pisa, Italy
| | | | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,I.R.C.C.S. I.N.M. Neuromed, Pozzilli, Italy
| |
Collapse
|
50
|
Correlation between cognition and plasma noradrenaline level in Alzheimer's disease: a potential new blood marker of disease evolution. Transl Psychiatry 2020; 10:213. [PMID: 32620743 PMCID: PMC7335170 DOI: 10.1038/s41398-020-0841-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/31/2022] Open
Abstract
Recent evidence showing degeneration of the noradrenergic system in the locus coeruleus (LC) in Alzheimer's disease (AD) has motivated great interest in noradrenaline (NA) as a potential brain hallmark of the disease. Despite the current exploration of blood markers for AD, the deregulation of the plasma NA concentration ([NA]plasma) in AD is currently not well understood. This retrospective study includes a cohort of 71 patients (32 AD patients, 22 with other dementia and 17 without dementia) who were given consultations for memory complaints in the Cognitive Neurology Center of Lariboisière (Paris) between 2009 and 2014. As previously described in brain tissue, we show for the first time a linear correlation between [NA]plasma and Mini Mental State Examination (MMSE) score in AD patients. We observed that high [NA]plasma in AD patients was associated with higher [Aβ1-42]CSF than in other AD patients with [NA]plasma similar to NC patients. In parallel, we observed a lower (p-Tau/Tau)CSF in AD patients with low [NA]plasma than in non-AD patients with [NA]plasma similar to [NA]plasma in NC patients. Our data suggest that [NA]plasma could be a potential biomarker of disease evolution in the context of AD and could possibly improve early diagnosis.
Collapse
|