201
|
Berg CN, Sinha N, Gluck MA. The Effects of APOE and ABCA7 on Cognitive Function and Alzheimer's Disease Risk in African Americans: A Focused Mini Review. Front Hum Neurosci 2019; 13:387. [PMID: 31749691 PMCID: PMC6848225 DOI: 10.3389/fnhum.2019.00387] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/16/2019] [Indexed: 01/12/2023] Open
Abstract
African Americans have double the prevalence of Alzheimer's disease (AD), as compared to European Americans. However, the underlying causes of this health disparity are due to a multitude of environmental, lifestyle, and genetic factors that are not yet fully understood. Here, we review the effects of the two largest genetic risk factors for AD in African Americans: Apolipoprotein E (APOE) and ABCA7. We will describe the direct effects of genetic variation on neural correlates of cognitive function and report the indirect modulating effects of genetic variation on modifiable AD risk factors, such as aerobic fitness. As a means of integrating previous findings, we present a novel schematic diagram to illustrate the many factors that contribute to AD risk and impaired cognitive function in older African Americans. Finally, we discuss areas that require further inquiry, and stress the importance of racially diverse and representative study populations.
Collapse
Affiliation(s)
- Chelsie N. Berg
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, United States
| | | | - Mark A. Gluck
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, United States
| |
Collapse
|
202
|
Tau deposition is associated with functional isolation of the hippocampus in aging. Nat Commun 2019; 10:4900. [PMID: 31653847 PMCID: PMC6814780 DOI: 10.1038/s41467-019-12921-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023] Open
Abstract
The tau protein aggregates in aging and Alzheimer disease and may lead to memory loss through disruption of medial temporal lobe (MTL)-dependent memory systems. Here, we investigated tau-mediated mechanisms of hippocampal dysfunction that underlie the expression of episodic memory decline using fMRI measures of hippocampal local coherence (regional homogeneity; ReHo), distant functional connectivity and tau-PET. We show that age and tau pathology are related to higher hippocampal ReHo. Functional disconnection between the hippocampus and other components of the MTL memory system, particularly an anterior-temporal network specialized for object memory, is also associated with higher hippocampal ReHo and greater tau burden in anterior-temporal regions. These associations are not observed in the posteromedial network, specialized for context/spatial information. Higher hippocampal ReHo predicts worse memory performance. These findings suggest that tau pathology plays a role in disconnecting the hippocampus from specific MTL memory systems leading to increased local coherence and memory decline. Deposition of tau protein aggregates occurs during aging and Alzheimer disease. Here, the authors show that tau burden in the anterior-temporal memory network is associated with disrupted fMRI connectivity and functional isolation of the hippocampus from other memory network components.
Collapse
|
203
|
Stark SM, Kirwan CB, Stark CEL. Mnemonic Similarity Task: A Tool for Assessing Hippocampal Integrity. Trends Cogn Sci 2019; 23:938-951. [PMID: 31597601 DOI: 10.1016/j.tics.2019.08.003] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/25/2022]
Abstract
The hippocampus is critical for learning and memory, relying in part on pattern separation processes supported by the dentate gyrus (DG) to prevent interference from overlapping memory representations. In 2007, we designed the Mnemonic Similarity Task (MST), a modified object recognition memory task, to be highly sensitive to hippocampal function by placing strong demands on pattern separation. The MST is now a widely used behavioral task, repeatedly shown to be sensitive to age-related memory decline, hippocampal connectivity, and hippocampal function, with specificity to the DG. Here, we review the utility of the MST, its relationship to hippocampal function, its utility in detecting hippocampal-based memory alterations across the lifespan, and impairments associated with clinical pathology from a variety of disorders.
Collapse
Affiliation(s)
- Shauna M Stark
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - C Brock Kirwan
- Department of Psychology, Neuroscience Center, Brigham Young University, Provo, UT, USA
| | - Craig E L Stark
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| |
Collapse
|
204
|
Aguirre N, Costumero V, Marin-Marin L, Escudero J, Belloch V, Parcet MA, Ávila C. Activity in Memory Brain Networks During Encoding Differentiates Mild Cognitive Impairment Converters from Non-Converters. J Alzheimers Dis 2019; 71:1049-1061. [DOI: 10.3233/jad-190421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Naiara Aguirre
- Department of Basic and Clinical Psychology and Psychobiology, Jaume I University, Castelló de la Plana, Spain
| | - Víctor Costumero
- Department of Basic and Clinical Psychology and Psychobiology, Jaume I University, Castelló de la Plana, Spain
- Center for Brain and Cognition, Pompeu Fabra University, Barcelona, Spain
| | - Lidón Marin-Marin
- Department of Basic and Clinical Psychology and Psychobiology, Jaume I University, Castelló de la Plana, Spain
| | - Joaquín Escudero
- Department of Neurology, General Hospital of Valencia, Valencia, Spain
| | | | - María Antonia Parcet
- Department of Basic and Clinical Psychology and Psychobiology, Jaume I University, Castelló de la Plana, Spain
| | - César Ávila
- Department of Basic and Clinical Psychology and Psychobiology, Jaume I University, Castelló de la Plana, Spain
| |
Collapse
|
205
|
VGF in Cerebrospinal Fluid Combined With Conventional Biomarkers Enhances Prediction of Conversion From MCI to AD. Alzheimer Dis Assoc Disord 2019; 33:307-314. [PMID: 31305322 DOI: 10.1097/wad.0000000000000328] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Previous work has suggested that the brain and cerebrospinal fluid (CSF) levels of a neural protein involved in synaptic transmission, VGF (a noninitialism), may be altered in mild cognitive impairment (MCI) and Alzheimer Disease (AD). The objective of the current work is to examine the potential of CSF levels of a peptide derived from VGF to predict conversion from MCI to AD. MATERIALS AND METHODS Using multivariate analytical approaches, the performance of the conventional biomarkers (CSF Aβ1-42 and phosphorylated tau +/- hippocampal volume) was compared with the same biomarkers combined with CSF VGF peptide levels in a large publicly available data set from human subjects. RESULTS It was observed that VGF peptides are lowered in CSF of patients with AD compared with controls and that combinations of CSF Aβ1-42 and phosphorylated tau, hippocampal volume, and VGF peptide levels outperformed conventional biomarkers alone (hazard ratio=2.2 vs. 3.9), for predicting MCI to AD conversion. CONCLUSIONS CSF VGF enhances the ability of conventional biomarkers to predict MCI to AD conversion. Future work will be needed to determine the specificity of VGF for AD versus other neurodegenerative diseases.
Collapse
|
206
|
Chai Z, Ma C, Jin X. Homeostatic activity regulation as a mechanism underlying the effect of brain stimulation. Bioelectron Med 2019; 5:16. [PMID: 32232105 PMCID: PMC7098242 DOI: 10.1186/s42234-019-0032-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/23/2019] [Indexed: 01/10/2023] Open
Abstract
Hyperexcitability of the neural network often occurs after brain injuries or degeneration and is a key pathophysiological feature in certain neurological diseases such as epilepsy, neuropathic pain, and tinnitus. Although the standard approach of pharmacological treatments is to directly suppress the hyperexcitability through reducing excitation or enhancing inhibition, different techniques for stimulating brain activity are often used to treat refractory neurological conditions. However, it is unclear why stimulating brain activity would be effective for controlling hyperexcitability. Recent studies suggest that the pathogenesis in these disorders exhibits a transition from an initial activity loss after acute injury or progressive neurodegeneration to subsequent development of hyperexcitability. This process mimics homeostatic activity regulation and may contribute to developing network hyperexcitability that underlies neurological symptoms. This hypothesis also predicts that stimulating brain activity should be effective in reducing hyperexcitability due to homeostatic activity regulation and in relieving symptoms. Here we review current evidence of homeostatic plasticity in the development of hyperexcitability in some neurological diseases and the effects of brain stimulation. The homeostatic plasticity hypothesis may provide new insights into the pathophysiology of neurological diseases and may guide the use of brain stimulation techniques for treating them.
Collapse
Affiliation(s)
- Zhi Chai
- Neurobiology Research Center, College of Basic Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619 China
| | - Cungen Ma
- Neurobiology Research Center, College of Basic Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030619 China
| | - Xiaoming Jin
- Department of Anatomy, Cell Biology and Physiology, Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB 500C, Indianapolis, IN 46202 USA
| |
Collapse
|
207
|
Amyloid β-Induced Upregulation of Na v1.6 Underlies Neuronal Hyperactivity in Tg2576 Alzheimer's Disease Mouse Model. Sci Rep 2019; 9:13592. [PMID: 31537873 PMCID: PMC6753212 DOI: 10.1038/s41598-019-50018-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/31/2019] [Indexed: 12/19/2022] Open
Abstract
Hyperexcitability and alterations in neuronal networks contribute to cognitive impairment in Alzheimer’s Disease (AD). Voltage-gated sodium channels (NaV), which are crucial for regulating neuronal excitability, have been implicated in AD-related hippocampal hyperactivity and higher incidence of spontaneous non-convulsive seizures. Here, we show by using primary hippocampal neurons exposed to amyloid-β1–42 (Aβ1–42) oligomers and from Tg2576 mouse embryos, that the selective upregulation of NaV1.6 subtype contributes to membrane depolarization and to the increase of spike frequency, thereby resulting in neuronal hyperexcitability. Interestingly, we also found that NaV1.6 overexpression is responsible for the aberrant neuronal activity observed in hippocampal slices from 3-month-old Tg2576 mice. These findings identify the NaV1.6 channels as a determinant of the hippocampal neuronal hyperexcitability induced by Aβ1–42 oligomers. The selective blockade of NaV1.6 overexpression and/or hyperactivity might therefore offer a new potential therapeutic approach to counteract early hippocampal hyperexcitability and subsequent cognitive deficits in the early stages of AD.
Collapse
|
208
|
Nguyen T, Kim M, Gwak J, Lee JJ, Choi KY, Lee KH, Kim JG. Investigation of brain functional connectivity in patients with mild cognitive impairment: A functional near-infrared spectroscopy (fNIRS) study. JOURNAL OF BIOPHOTONICS 2019; 12:e201800298. [PMID: 30963713 DOI: 10.1002/jbio.201800298] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 03/17/2019] [Accepted: 04/04/2019] [Indexed: 06/09/2023]
Abstract
This study examines brain functional connectivity in both cognitively normal seniors and patients with mild cognitive impairment (MCI) to elucidate prospective markers of MCI. A homemade four-channel functional near-infrared spectroscopy (fNIRS) system was employed to measure hemodynamic responses in the subjects' prefrontal cortex during a resting state, an oddball task, a 1-back task, and a verbal fluency task. Brain functional connectivity was calculated as the Pearson correlation coefficients between fNIRS channels. The results show that during the verbal fluency task, while the healthy control (HC) group presents a significantly stronger inter-hemispheric connectivity compared to intra-hemispheric connectivity, there is no difference between the inter- and intra-hemispheric connectivity in the MCI group. In addition, a comparison between the MCI and HC connectivity reveals that the MCI group has a statistically higher right and inter-hemispheric connectivity during the resting state, but a significantly lower left and inter-hemispheric connectivity during the verbal fluency test. These findings demonstrate the potential of fNIRS to study brain functional connectivity in neurodegenerative diseases.
Collapse
Affiliation(s)
- Thien Nguyen
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Minhee Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jeonghwan Gwak
- Department of Software, Korea National University of Transportation, Chungju, Republic of Korea
| | - Jang J Lee
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
| | - Kyu Y Choi
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
| | - Kun H Lee
- Department of Biomedical Science, Chosun University, Gwangju, Republic of Korea
- National Research Center for Dementia, Chosun University, Gwangju, Republic of Korea
| | - Jae G Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
209
|
Zhang W, Johndro H, Budson AE, Gutchess A. Influence of self-referential mode on memory for aMCI patients. Cogn Neuropsychol 2019; 37:46-57. [PMID: 31437095 DOI: 10.1080/02643294.2019.1657390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
One's concept of the self can act as a schema, in some cases facilitating memory performance but in other cases making memory more prone to errors. In this study, we attempted to induce long-lasting self-reference effects through an autobiographical mode in younger and older adults, as well as patients with amnestic mild cognitive impairment (aMCI), predicting that a self-referential mode of thought would benefit memory. Participants first either recalled autobiographical memories or described three neutral photographs in a narrative condition. This was followed by a conventional self-referencing task. At retrieval, participants completed a recognition task. Contrary to our prediction, the self-referencing benefit emerged consistently under autobiographical and narrative modes across all groups. Although our findings indicate that self-referencing can benefit memory, it carries the risk of increasing false alarm rates when induced through an autobiographical mode and consequently the strategy should be utilized with caution.
Collapse
Affiliation(s)
- Wanbing Zhang
- Department of Psychology, Brandeis University, Waltham, MA, USA
| | - Hunter Johndro
- Department of Psychology, Brandeis University, Waltham, MA, USA
| | - Andrew E Budson
- VA Boston Healthcare System, Boston, MA, USA.,Boston University School of Medicine, Boston, MA, USA
| | - Angela Gutchess
- Department of Psychology, Brandeis University, Waltham, MA, USA
| |
Collapse
|
210
|
Foster JB, Lashley R, Zhao F, Wang X, Kung N, Askwith CC, Lin L, Shultis MW, Hodgetts KJ, Lin CLG. Enhancement of tripartite synapses as a potential therapeutic strategy for Alzheimer's disease: a preclinical study in rTg4510 mice. Alzheimers Res Ther 2019; 11:75. [PMID: 31439023 PMCID: PMC6706914 DOI: 10.1186/s13195-019-0530-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The lack of effective treatment options for Alzheimer's disease (AD) is of momentous societal concern. Synaptic loss is the hallmark of AD that correlates best with impaired memory and occurs early in the disease process, before the onset of clinical symptoms. We have developed a small-molecule, pyridazine-based series that enhances the structure and function of both the glial processes and the synaptic boutons that form the tripartite synapse. Previously, we have shown that these pyridazine derivatives exhibit profound efficacy in an amyloid precursor protein AD model. Here, we evaluated the efficacy of an advanced compound, LDN/OSU-0215111, in rTg4510 mice-an aggressive tauopathy model. METHODS rTg4510 mice were treated orally with vehicle or LDN/OSU-0215111 (10 mg/kg) daily from the early symptomatic stage (2 months old) to moderate (4 months old) and severe (8 months old) disease stages. At each time point, mice were subjected to a battery of behavioral tests to assess the activity levels and cognition. Also, tissue collections were performed on a subset of mice to analyze the tripartite synaptic changes, neurodegeneration, gliosis, and tau phosphorylation as assessed by immunohistochemistry and Western blotting. At 8 months of age, a subset of rTg4510 mice treated with compound was switched to vehicle treatment and analyzed behaviorally and biochemically 30 days after treatment cessation. RESULTS At both the moderate and severe disease stages, compound treatment normalized cognition and behavior as well as reduced synaptic loss, neurodegeneration, tau hyperphosporylation, and neuroinflammation. Importantly, after 30 days of treatment cessation, the benefits of compound treatment were sustained, indicating disease modification. We also found that compound treatment rapidly and robustly reduced tau hyperphosphorylation/deposition possibly via the inhibition of GSK3β. CONCLUSIONS The results show that LDN/OSU-0215111 provides benefits for multiple aspects of tauopathy-dependent pathology found in Alzheimer's disease including tripartite synapse normalization and reduction of toxic tau burden, which, in turn, likely accounted for normalized cognition and activity levels in compound-treated rTg4510 mice. This study, in combination with our previous work regarding the benefit of pyridazine derivatives against amyloid-dependent pathology, strongly supports pyridazine derivatives as a viable, clinically relevant, and disease-modifying treatment for many of the facets of Alzheimer's disease.
Collapse
Affiliation(s)
- Joshua B. Foster
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Rashelle Lashley
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Fangli Zhao
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Xueqin Wang
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Nydia Kung
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Candice C. Askwith
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Lin Lin
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA USA
| | - Michael W. Shultis
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA USA
| | - Kevin J. Hodgetts
- Department of Neurology, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH USA
| |
Collapse
|
211
|
Pusil S, Dimitriadis SI, López ME, Pereda E, Maestú F. Aberrant MEG multi-frequency phase temporal synchronization predicts conversion from mild cognitive impairment-to-Alzheimer's disease. Neuroimage Clin 2019; 24:101972. [PMID: 31522127 PMCID: PMC6745514 DOI: 10.1016/j.nicl.2019.101972] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/19/2019] [Accepted: 08/03/2019] [Indexed: 11/15/2022]
Abstract
Many neuroimaging studies focus on a frequency-specific or a multi-frequency network analysis showing that functional brain networks are disrupted in patients with Alzheimer's disease (AD). Although those studies enriched our knowledge of the impact of AD in brain's functionality, our goal is to test the effectiveness of combining neuroimaging with network neuroscience to predict with high accuracy subjects with mild cognitive impairment (MCI) that will convert to AD. In this study, eyes-closed resting-state magnetoencephalography (MEG) recordings from 27 stable MCI (sMCI) and 27 progressive MCI (pMCI) from two scan sessions (baseline and follow-up after approximately 3 years) were projected via beamforming onto an atlas-based set of regions of interest (ROIs). Dynamic functional connectivity networks were constructed independently for the five classical frequency bands while a multivariate phase-based coupling metric was adopted. Thus, computing the distance between the fluctuation of functional strength of every pair of ROIs between the two conditions with dynamic time wrapping (DTW), a large set of features was extracted. A machine learning algorithm revealed 30 DTW-based features in the five frequency bands that can distinguish the sMCI from pMCI with absolute accuracy (100%). Further analysis of the selected links revealed that most of the connected ROIs were part of the default mode network (DMN), the cingulo-opercular (CO), the fronto-parietal and the sensorimotor network. Overall, our dynamic network multi-frequency analysis approach provides an effective framework of constructing a sensitive MEG-based connectome biomarker for the prediction of conversion from MCI to Alzheimer's disease.
Collapse
Affiliation(s)
- Sandra Pusil
- Laboratory of Neuropsychology, University of the Balearic Islands, Spain; Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain.
| | - Stavros I Dimitriadis
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, United Kingdom; Neuroinformatics Group, Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, United Kingdom; Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; School of Psychology, Cardiff University, Cardiff, United Kingdom; Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom; MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - María Eugenia López
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Ernesto Pereda
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Electrical Engineering and Bioengineering Lab, Department of Industrial Engineering, IUNE Universidad de La Laguna, Tenerife, Spain
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Universidad Complutense and Universidad Politécnica de Madrid, Madrid, Spain; Department of Experimental Psychology, Universidad Complutense de Madrid, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
212
|
Pignataro A, Meli G, Pagano R, Fontebasso V, Battistella R, Conforto G, Ammassari-Teule M, Middei S. Activity-Induced Amyloid-β Oligomers Drive Compensatory Synaptic Rearrangements in Brain Circuits Controlling Memory of Presymptomatic Alzheimer's Disease Mice. Biol Psychiatry 2019; 86:185-195. [PMID: 30528194 DOI: 10.1016/j.biopsych.2018.10.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND A consistent proportion of individuals at risk for Alzheimer's disease show intact cognition regardless of the extensive accumulation of amyloid-β (Aβ) peptide in their brain. Several pieces of evidence indicate that overactivation of brain regions negative for Aβ can compensate for the underactivation of Aβ-positive ones to preserve cognition, but the underlying synaptic changes are still unexplored. METHODS Using Golgi staining, we investigate how dendritic spines rearrange following contextual fear conditioning (CFC) in the hippocampus and amygdala of presymptomatic Tg2576 mice, a genetic model for Aβ accumulation. A molecular biology approach combined with intrahippocampal injection of a γ-secretase inhibitor evaluates the impact of Aβ fluctuations on spine rearrangements. RESULTS Encoding of CFC increases Aβ oligomerization in the hippocampus but not in the amygdala of Tg2576 mice. The presence of Aβ oligomers predicts vulnerability to network dysfunctions, as low c-Fos activation and spine maturation are detected in the hippocampus of Tg2576 mice upon recall of CFC memory. Rather, enhanced c-Fos activation and new spines are evident in the amygdala of Tg2576 mice compared with wild-type control mice. Preventing Aβ increase in the hippocampus of Tg2576 mice restores CFC-associated spine changes to wild-type levels in both the hippocampus and amygdala. CONCLUSIONS Our study provides the first evidence of neural compensation consisting of enhanced synaptic activity in brain regions spared by Aβ load. Furthermore, it unravels an activity-mediated feedback loop through which neuronal activation during CFC encoding favors Aβ oligomerization in the hippocampus and prevents synaptic rearrangements in this region.
Collapse
Affiliation(s)
- Annabella Pignataro
- Laboratory of Psychobiology, Department of Experimental Neurology, Santa Lucia Foundation, Rome, Italy; Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Giovanni Meli
- European Brain Research Institute-Fondazione Rita Levi Montalcini, Rome, Italy.
| | - Roberto Pagano
- Department of Molecular and Cellular Neurobiology Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Veronica Fontebasso
- Department of Pharmacology & Toxicology and Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, Austria
| | - Roberta Battistella
- Department of Experimental Biomedical Sciences, Lund University, Lund, Sweden
| | - Giulia Conforto
- Laboratory of Psychobiology, Department of Experimental Neurology, Santa Lucia Foundation, Rome, Italy
| | - Martine Ammassari-Teule
- Laboratory of Psychobiology, Department of Experimental Neurology, Santa Lucia Foundation, Rome, Italy; Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy
| | - Silvia Middei
- Institute of Cell Biology and Neurobiology, National Research Council, Rome, Italy.
| |
Collapse
|
213
|
Scherr M, Pasquini L, Benson G, Nuttall R, Gruber M, Neitzel J, Brandl F, Sorg C. Decoupling of Local Metabolic Activity and Functional Connectivity Links to Amyloid in Alzheimer's Disease. J Alzheimers Dis 2019; 64:405-415. [PMID: 29843243 DOI: 10.3233/jad-180022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Both ongoing local metabolic activity (LMA) and corresponding functional connectivity (FC) with remote brain regions are progressively impaired in Alzheimer's disease (AD), particularly in the posterior default mode network (pDMN); however, it is unknown how these impairments interact. It is well known that decreasing mean synaptic activity of a region, i.e., decreasing LMA, reduces the region's sensitivity to afferent input from other regions, i.e., FC. OBJECTIVE We hypothesized progressive decoupling between LMA and FC in AD, which is linked to amyloid-β pathology (Aβ). METHODS Healthy adults (n=20) and Aβ+patients without memory impairment (n=9), early MCI (n=21), late MCI (n=18) and AD (n=22) were assessed by resting-state fMRI, FDG-PET, and AV-45-PET to measure FC, LMA, and Aβ of the pDMN. Coupling between LMA and FC (rLA/FC) was estimated by voxelwise correlation. RESULTS RLMA/FC decreased with disease severity (F=20.09, p<0.001). This decrease was specifically associated with pDMN Aβ (r=-0.273, p=0.029) but not global Aβ (r=-0.112, p=0.378) and with the impact of Aβ on FC (i.e., rAβ/FC,r=-0.339; p=0.006). In multiple regression models rLMA/FC was also associated with memory impairment, reduced cognitive speed and flexibility, outperforming global Aβ, pDMN Aβ, pDMN LMA, and pDMN FC, respectively. CONCLUSION Results demonstrate increasing decoupling of LMA from its FC in AD. Data suggest that decoupling is driven by local Aβ and contributes to memory decline.
Collapse
Affiliation(s)
- Martin Scherr
- Department of Neurology, Paracelsus Medical University Salzburg, and Christian Doppler Medical Centre, Salzburg, Austria; Centre for Cognitive Neurosciences Salzburg, Salzburg, Austria.,Department of Psychiatry and Psychotherapy, Technische Universität München, Klinikum rechts der Isar, Munich, Germany
| | - Lorenzo Pasquini
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Gloria Benson
- Department of Neurology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Rachel Nuttall
- Department of Psychology, University of Salzburg, Salzburg, Austria.,TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,General and Experimental Psychology, Department of Psychology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Gruber
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Julia Neitzel
- General and Experimental Psychology, Department of Psychology, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Felix Brandl
- TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christian Sorg
- Department of Psychiatry and Psychotherapy, Technische Universität München, Klinikum rechts der Isar, Munich, Germany.,TUM Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | |
Collapse
|
214
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
215
|
Dietlin S, Soto M, Kiyasova V, Pueyo M, de Mauleon A, Delrieu J, Ousset PJ, Vellas B. Neuropsychiatric Symptoms and Risk of Progression to Alzheimer’s Disease Among Mild Cognitive Impairment Subjects. J Alzheimers Dis 2019; 70:25-34. [DOI: 10.3233/jad-190025] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Simon Dietlin
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| | - Maria Soto
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| | - Vera Kiyasova
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Maria Pueyo
- Institut de Recherches Internationales Servier, Suresnes, France
| | - Adelaïde de Mauleon
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| | - Julien Delrieu
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| | - Pierre Jean Ousset
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| | - Bruno Vellas
- Gerontopôle, INSERM U 1027, Alzheimer’s Disease Research and Clinical Center, Toulouse University Hospital, France
| |
Collapse
|
216
|
Delhaye E, Mechanic-Hamilton D, Saad L, Das SR, Wisse LEM, Yushkevich PA, Wolk DA, Bastin C. Associative memory for conceptually unitized word pairs in mild cognitive impairment is related to the volume of the perirhinal cortex. Hippocampus 2019; 29:630-638. [PMID: 30588714 PMCID: PMC6565465 DOI: 10.1002/hipo.23063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 11/06/2022]
Abstract
Unitization, that is, the encoding of an association as one integrated entity, has been shown to improve associative memory in populations presenting with associative memory deficit due to hippocampal dysfunction, such as amnesic patients with focal hippocampal lesions and healthy older adults. One reason for this benefit is that encoding of unitized associations would rely on the perirhinal cortex (PrC) and thus minimize the need for hippocampal recruitment. Mild cognitive impairment (MCI) is accompanied by a deficit in associative memory. However, unitization has never been studied to explore the potential benefit in associative memory in MCI, maybe because MCI is characterized by PrC pathology. However, the PrC may potentially still function sufficiently to allow for the successful adoption of unitization. In this study, we aimed at assessing whether unitization could attenuate MCI patients' associative memory deficit, and whether the ability to remember unitized associations would be modulated by the integrity of the PrC in MCI patients. Unitization was manipulated at a conceptual level, by encouraging participants to encode unrelated word pairs as new compound words. Participants also underwent a structural MRI exam, and measures of PrC were extracted (Brodmann Areas [BA] 35 and 36). Results showed that, contrary to healthy controls, MCI patients did not benefit from unitization. Moreover, their memory performance for unitized associations was related to the measure of PrC integrity (BA35), while it was not the case in controls. This finding thus suggests that unitization does not help to attenuate the associative deficit in MCI patients, and brings support to the literature linking unitization to the PrC function.
Collapse
Affiliation(s)
- Emma Delhaye
- GIGA-CRC In-Vivo Imaging, Liege University, Liège, Belgium
- PsyNCog, Faculty of Psychology, Liege, Belgium
| | - Dawn Mechanic-Hamilton
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Memory Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laura Saad
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Memory Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sandhitsu R. Das
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laura E. M. Wisse
- Penn Memory Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul A. Yushkevich
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A. Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Memory Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Image Computing and Science Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christine Bastin
- GIGA-CRC In-Vivo Imaging, Liege University, Liège, Belgium
- PsyNCog, Faculty of Psychology, Liege, Belgium
| |
Collapse
|
217
|
Vico Varela E, Etter G, Williams S. Excitatory-inhibitory imbalance in Alzheimer's disease and therapeutic significance. Neurobiol Dis 2019; 127:605-615. [DOI: 10.1016/j.nbd.2019.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 11/29/2022] Open
|
218
|
Zott B, Busche MA, Sperling RA, Konnerth A. What Happens with the Circuit in Alzheimer's Disease in Mice and Humans? Annu Rev Neurosci 2019; 41:277-297. [PMID: 29986165 DOI: 10.1146/annurev-neuro-080317-061725] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A major mystery of many types of neurological and psychiatric disorders, such as Alzheimer's disease (AD), remains the underlying, disease-specific neuronal damage. Because of the strong interconnectivity of neurons in the brain, neuronal dysfunction necessarily disrupts neuronal circuits. In this article, we review evidence for the disruption of large-scale networks from imaging studies of humans and relate it to studies of cellular dysfunction in mouse models of AD. The emerging picture is that some forms of early network dysfunctions can be explained by excessively increased levels of neuronal activity. The notion of such neuronal hyperactivity receives strong support from in vivo and in vitro cellular imaging and electrophysiological recordings in the mouse, which provide mechanistic insights underlying the change in neuronal excitability. Overall, some key aspects of AD-related neuronal dysfunctions in humans and mice are strikingly similar and support the continuation of such a translational strategy.
Collapse
Affiliation(s)
- Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; .,Center for Integrated Protein Sciences, Technical University of Munich, 80802 Munich, Germany.,Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany
| | - Marc Aurel Busche
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA.,Department of Neurology and Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, 80802 Munich, Germany; .,Center for Integrated Protein Sciences, Technical University of Munich, 80802 Munich, Germany.,Munich Cluster for Systems Neurology, Technical University of Munich, 80802 Munich, Germany
| |
Collapse
|
219
|
Najm R, Jones EA, Huang Y. Apolipoprotein E4, inhibitory network dysfunction, and Alzheimer's disease. Mol Neurodegener 2019; 14:24. [PMID: 31186040 PMCID: PMC6558779 DOI: 10.1186/s13024-019-0324-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/23/2019] [Indexed: 02/08/2023] Open
Abstract
Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer's disease (AD), increasing risk and decreasing age of disease onset. Many studies have demonstrated the detrimental effects of apoE4 in varying cellular contexts. However, the underlying mechanisms explaining how apoE4 leads to cognitive decline are not fully understood. Recently, the combination of human induced pluripotent stem cell (hiPSC) modeling of neurological diseases in vitro and electrophysiological studies in vivo have begun to unravel the intersection between apoE4, neuronal subtype dysfunction or loss, subsequent network deficits, and eventual cognitive decline. In this review, we provide an overview of the literature describing apoE4's detrimental effects in the central nervous system (CNS), specifically focusing on its contribution to neuronal subtype dysfunction or loss. We focus on γ-aminobutyric acid (GABA)-expressing interneurons in the hippocampus, which are selectively vulnerable to apoE4-mediated neurotoxicity. Additionally, we discuss the importance of the GABAergic inhibitory network to proper cognitive function and how dysfunction of this network manifests in AD. Finally, we examine how apoE4-mediated GABAergic interneuron loss can lead to inhibitory network deficits and how this deficit results in cognitive decline. We propose the following working model: Aging and/or stress induces neuronal expression of apoE. GABAergic interneurons are selectively vulnerable to intracellularly produced apoE4, through a tau dependent mechanism, which leads to their dysfunction and eventual death. In turn, GABAergic interneuron loss causes hyperexcitability and dysregulation of neural networks in the hippocampus and cortex. This dysfunction results in learning, memory, and other cognitive deficits that are the central features of AD.
Collapse
Affiliation(s)
- Ramsey Najm
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, 94143, USA
| | - Emily A Jones
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94143, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA, 94143, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, CA, 94143, USA.
- Department of Neurology, University of California, San Francisco, CA, 94143, USA.
- Department of Pathology, University of California, San Francisco, CA, 94143, USA.
| |
Collapse
|
220
|
Márquez F, Yassa MA. Neuroimaging Biomarkers for Alzheimer's Disease. Mol Neurodegener 2019; 14:21. [PMID: 31174557 PMCID: PMC6555939 DOI: 10.1186/s13024-019-0325-5] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
Currently, over five million Americans suffer with Alzheimer's disease (AD). In the absence of a cure, this number could increase to 13.8 million by 2050. A critical goal of biomedical research is to establish indicators of AD during the preclinical stage (i.e. biomarkers) allowing for early diagnosis and intervention. Numerous advances have been made in developing biomarkers for AD using neuroimaging approaches. These approaches offer tremendous versatility in terms of targeting distinct age-related and pathophysiological mechanisms such as structural decline (e.g. volumetry, cortical thinning), functional decline (e.g. fMRI activity, network correlations), connectivity decline (e.g. diffusion anisotropy), and pathological aggregates (e.g. amyloid and tau PET). In this review, we survey the state of the literature on neuroimaging approaches to developing novel biomarkers for the amnestic form of AD, with an emphasis on combining approaches into multimodal biomarkers. We also discuss emerging methods including imaging epigenetics, neuroinflammation, and synaptic integrity using PET tracers. Finally, we review the complementary information that neuroimaging biomarkers provide, which highlights the potential utility of composite biomarkers as suitable outcome measures for proof-of-concept clinical trials with experimental therapeutics.
Collapse
Affiliation(s)
- Freddie Márquez
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, 92697, USA.
| | - Michael A Yassa
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
221
|
Cacace R, Heeman B, Van Mossevelde S, De Roeck A, Hoogmartens J, De Rijk P, Gossye H, De Vos K, De Coster W, Strazisar M, De Baets G, Schymkowitz J, Rousseau F, Geerts N, De Pooter T, Peeters K, Sieben A, Martin JJ, Engelborghs S, Salmon E, Santens P, Vandenberghe R, Cras P, P. De Deyn P, C. van Swieten J, M. van Duijn C, van der Zee J, Sleegers K, Van Broeckhoven C. Loss of DPP6 in neurodegenerative dementia: a genetic player in the dysfunction of neuronal excitability. Acta Neuropathol 2019; 137:901-918. [PMID: 30874922 PMCID: PMC6531610 DOI: 10.1007/s00401-019-01976-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Emerging evidence suggested a converging mechanism in neurodegenerative brain diseases (NBD) involving early neuronal network dysfunctions and alterations in the homeostasis of neuronal firing as culprits of neurodegeneration. In this study, we used paired-end short-read and direct long-read whole genome sequencing to investigate an unresolved autosomal dominant dementia family significantly linked to 7q36. We identified and validated a chromosomal inversion of ca. 4 Mb, segregating on the disease haplotype and disrupting the coding sequence of dipeptidyl-peptidase 6 gene (DPP6). DPP6 resequencing identified significantly more rare variants-nonsense, frameshift, and missense-in early-onset Alzheimer's disease (EOAD, p value = 0.03, OR = 2.21 95% CI 1.05-4.82) and frontotemporal dementia (FTD, p = 0.006, OR = 2.59, 95% CI 1.28-5.49) patient cohorts. DPP6 is a type II transmembrane protein with a highly structured extracellular domain and is mainly expressed in brain, where it binds to the potassium channel Kv4.2 enhancing its expression, regulating its gating properties and controlling the dendritic excitability of hippocampal neurons. Using in vitro modeling, we showed that the missense variants found in patients destabilize DPP6 and reduce its membrane expression (p < 0.001 and p < 0.0001) leading to a loss of protein. Reduced DPP6 and/or Kv4.2 expression was also detected in brain tissue of missense variant carriers. Loss of DPP6 is known to cause neuronal hyperexcitability and behavioral alterations in Dpp6-KO mice. Taken together, the results of our genomic, genetic, expression and modeling analyses, provided direct evidence supporting the involvement of DPP6 loss in dementia. We propose that loss of function variants have a higher penetrance and disease impact, whereas the missense variants have a variable risk contribution to disease that can vary from high to low penetrance. Our findings of DPP6, as novel gene in dementia, strengthen the involvement of neuronal hyperexcitability and alteration in the homeostasis of neuronal firing as a disease mechanism to further investigate.
Collapse
Affiliation(s)
- Rita Cacace
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Bavo Heeman
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Sara Van Mossevelde
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Arne De Roeck
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Julie Hoogmartens
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Peter De Rijk
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Helena Gossye
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Kristof De Vos
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Wouter De Coster
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Mojca Strazisar
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Greet De Baets
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Centre for Brain and Disease Research, Louvain, Belgium
- Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Nathalie Geerts
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Tim De Pooter
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Karin Peeters
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Anne Sieben
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- Department of Neurology, University Hospital Ghent and University of Ghent, Ghent, Belgium
| | | | - Sebastiaan Engelborghs
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Eric Salmon
- Department of Neurology, Centre Hospitalier Universitaire de Liège and University of Liège, Liège, Belgium
| | - Patrick Santens
- Department of Neurology, University Hospital Ghent and University of Ghent, Ghent, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Faculty of Medicine, KU Leuven, Louvain, Belgium
- Laboratory of Cognitive Neurology, Department of Neurology, University Hospitals Leuven, Louvain, Belgium
| | - Patrick Cras
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Peter P. De Deyn
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA), Middelheim and Hoge Beuken, Antwerp, Belgium
| | - John C. van Swieten
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Julie van der Zee
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Kristel Sleegers
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Center for Molecular Neurology, VIB, Antwerp, Belgium
- Institute Born-Bunge, Antwerp, Belgium
- University of Antwerp, Antwerp, Belgium
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, University of Antwerp, CDE, Universiteitsplein 1, 2610 Antwerp, Belgium
| |
Collapse
|
222
|
Novelty processing and memory impairment in Alzheimer's disease: A review. Neurosci Biobehav Rev 2019; 100:237-249. [DOI: 10.1016/j.neubiorev.2019.02.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/24/2018] [Accepted: 02/28/2019] [Indexed: 01/09/2023]
|
223
|
Jacobson A, Green E, Haase L, Szajer J, Murphy C. Differential Effects of BMI on Brain Response to Odor in Olfactory, Reward and Memory Regions: Evidence from fMRI. Nutrients 2019; 11:E926. [PMID: 31022978 PMCID: PMC6520683 DOI: 10.3390/nu11040926] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 04/18/2019] [Accepted: 04/18/2019] [Indexed: 01/10/2023] Open
Abstract
:Obesity has reached epidemic proportions, motivating research into the underlying mechanisms. Olfaction is a powerful mediator of food consumption, and obesity has been associated with altered olfactory sensitivity. The current study used an event-related functional magnetic resonance imaging (fMRI) to examine the central processing of odor in humans to gain insight into the effect of the body mass index (BMI) on the neural processes involved in rating the pleasantness of a food odor during a hunger state and in a satiety state. We hypothesized that, during the hedonic evaluation of food odor, BMI would be associated with differences in brain activation within olfactory and higher order processing areas important for perception, reward, and memory. We report novel findings of a dissociation between the relationship between BMI and activation in reward areas and in olfactory and odor memory areas, i.e., activation in reward areas decreased as BMI increased, whereas activation in primary olfactory and memory regions increased as BMI increased. A greater BMI is associated with decreased activation in the reward and frontal regions, supporting a blunted reward response in obesity. These findings have important potential implications for decision making, response inhibition, and reward-based behaviors that may play key roles as causal and maintenance factors in obesity. In contrast, a greater BMI is associated with an increased activation in the primary olfactory and memory areas, which was observed during a hunger state. These results raise the speculative hypothesis that high BMI may be associated with hyperactivation in the olfactory and memory areas, and that over time, the resulting excitotoxic effects may contribute to neurodegenerative changes in these areas.
Collapse
Affiliation(s)
- Aaron Jacobson
- Department of Psychology, San Diego State University, San Diego, CA 92182, USA.
| | - Erin Green
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA.
| | - Lori Haase
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA.
- Department of Psychiatry, University of California School of Medicine, San Diego, CA 92093, USA.
| | - Jacquelyn Szajer
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA.
| | - Claire Murphy
- Department of Psychology, San Diego State University, San Diego, CA 92182, USA.
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA.
- Department of Psychiatry, University of California School of Medicine, San Diego, CA 92093, USA.
| |
Collapse
|
224
|
Wu M, Thurston RC, Tudorascu DL, Karim HT, Mathis CA, Lopresti BJ, Kamboh MI, Cohen AD, Snitz BE, Klunk WE, Aizenstein HJ. Amyloid deposition is associated with different patterns of hippocampal connectivity in men versus women. Neurobiol Aging 2019; 76:141-150. [PMID: 30711677 PMCID: PMC6584958 DOI: 10.1016/j.neurobiolaging.2018.11.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 10/04/2018] [Accepted: 11/18/2018] [Indexed: 01/26/2023]
Abstract
Compared to men, women are disproportionally affected by Alzheimer's disease (AD) and have an accelerated trajectory of cognitive decline and disease progression. Neurobiological factors underlying gender differences in AD remain unclear. This study investigated brain beta-amyloid (Aβ)-related neural system differences in cognitively normal older men and women (N = 61; 41 females, 65-93 years old). We found that men and women showed different associations between Aβ load and hippocampal functional connectivity. During associative memory encoding, in men greater Aβ burden was accompanied by greater hippocampus-prefrontal connectivity (i.e., more synchronized activities), whereas in women hippocampal connectivity did not vary by Aβ burden. For resting-state data, the interaction of gender × Aβ on hippocampal connectivity did not survive multiple comparison in the whole-brain analyses. In the region of interest-based analyses, resting-state hippocampal-prefrontal connectivity was positively correlated with Aβ load in men and was negatively correlated with Aβ load in women. The observed Aβ-related neural differences may explain the accelerated trajectory of cognitive decline and AD progression in women.
Collapse
Affiliation(s)
- Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Departments of Epidemiology and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L Tudorascu
- Departments of Medicine and Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Helmet T Karim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
225
|
Popiolek M, Mandelblat-Cerf Y, Young D, Garst-Orozco J, Lotarski SM, Stark E, Kramer M, Butler CR, Kozak R. In Vivo Modulation of Hippocampal Excitability by M4 Muscarinic Acetylcholine Receptor Activator: Implications for Treatment of Alzheimer's Disease and Schizophrenic Patients. ACS Chem Neurosci 2019; 10:1091-1098. [PMID: 30335349 DOI: 10.1021/acschemneuro.8b00496] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Abnormal hippocampal activity has been linked to impaired cognitive performance in Alzheimer's disease and schizophrenia, leading to a hypothesis that normalization of this activity may be therapeutically beneficial. Our work suggests that one approach for hippocampal normalization may be through activation of the M4 muscarinic acetylcholine receptor. We used a brain penetrant M4 muscarinic acetylcholine receptor selective activator, PT-3763, to show dose-dependent attenuation of field potentials in Schaffer collateral (CA3-CA1) and recurrent associational connections (CA3-CA3) ex vivo in hippocampal slices. In vivo, systemic administration of PT-3763 led to attenuation of glutamate release in CA3 as measured by amperometry and to a dose-dependent decrease in population CA1 pyramidal activity as measured by fiber photometry. This decrease in population activity was also evident with a localized administration of the compound to the recorded site. Finally, PT-3763 reversed scopolamine-induced deficit in Morris water maze. Our results suggest that M4 muscarinic acetylcholine receptor activation may be a suitable therapeutic treatment in diseases associated with hyperactive hippocampal activity.
Collapse
|
226
|
Zhuang X, Yang Z, Sreenivasan KR, Mishra VR, Curran T, Nandy R, Cordes D. Multivariate group-level analysis for task fMRI data with canonical correlation analysis. Neuroimage 2019; 194:25-41. [PMID: 30894332 DOI: 10.1016/j.neuroimage.2019.03.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/26/2019] [Accepted: 03/13/2019] [Indexed: 11/25/2022] Open
Abstract
Task-based functional Magnetic Resonance Imaging (fMRI) has been widely used to determine population-based brain activations for cognitive tasks. Popular group-level analysis in fMRI is based on the general linear model and constitutes a univariate method. However, univariate methods are known to suffer from low sensitivity for a given specificity because the spatial covariance structure at each voxel is not taken entirely into account. In this study, a spatially constrained local multivariate model is introduced for group-level analysis to improve sensitivity at a given specificity for activation detection. The proposed model is formulated in terms of a multivariate constrained optimization problem based on the maximum log likelihood method and solved efficiently with numerical optimization techniques. Both simulated data mimicking real fMRI time series at multiple noise fractions and real fMRI episodic memory data have been used to evaluate the performance of the proposed method. For simulated data, the area under the receiver operating characteristic curves in detecting group activations increases for the subject and group level multivariate method by 20%, as compared to the univariate method. Results from real fMRI data indicate a significant increase in group-level activation detection, particularly in hippocampus, para-hippocampal area and nearby medial temporal lobe regions with the proposed method.
Collapse
Affiliation(s)
- Xiaowei Zhuang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA
| | - Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA
| | | | - Virendra R Mishra
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA
| | - Tim Curran
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, 80309, USA
| | - Rajesh Nandy
- School of Public Health, University of North Texas, Fort Worth, TX, 76107, USA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA; Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, 80309, USA.
| |
Collapse
|
227
|
Unsupervised excitation: GABAergic dysfunctions in Alzheimer’s disease. Brain Res 2019; 1707:216-226. [DOI: 10.1016/j.brainres.2018.11.042] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/22/2022]
|
228
|
Wang X, Zhou W, Ye T, Lin X, Zhang J. The Relationship Between Hippocampal Volumes and Delayed Recall Is Modified by APOE ε4 in Mild Cognitive Impairment. Front Aging Neurosci 2019; 11:36. [PMID: 30863302 PMCID: PMC6399520 DOI: 10.3389/fnagi.2019.00036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 11/13/2022] Open
Abstract
Objective: To investigate whether APOE ε4 affects the association of verbal memory with neurodegeneration presented by the hippocampal volume/intracranial volume ratio (HpVR). Methods: The study sample included 371 individuals with normal cognition (NC), 725 subjects with amnestic mild cognitive impairment (aMCI), and 251 patients with mild Alzheimer’s disease (AD) from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) who underwent the rey auditory verbal learning test (RAVLT). Multiple linear regression models were conducted to assess the effect of the APOE ε4∗HpVR interaction on RAVLT in all subjects and in each diagnostic group adjusting for age, gender and educational attainment, and global cognition. Results: In all subjects, there was no significant APOE ε4 × HpVR interaction for immediate recall or delayed recall (p > 0.05). However, in aMCI subjects, there was a significant APOE ε4 × HpVR interaction for delayed recall (p = 0.008), but not immediate recall (p = 0.15). More specifically, the detrimental effect of APOE ε4 on delayed recall altered by HpVR such that this effect was most evident among subjects with small to moderate HpVR, but this disadvantage was absent or even reversed among subjects with larger HpVR. No significant interaction was observed in the NC or AD group. Conclusion: These findings highlight a potential role of APOE ε4 status in affecting the association of hippocampus size with delayed recall memory in the early stage of AD.
Collapse
Affiliation(s)
- Xiwu Wang
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Wenjun Zhou
- Department of Pathology, Hangzhou Normal University, Hangzhou, China
| | - Teng Ye
- Department of Ultrasound, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Jie Zhang
- Independent Researcher, Hangzhou, China
| | | |
Collapse
|
229
|
Mohanty R, Nair VA, Tellapragada N, Williams LM, Kang TJ, Prabhakaran V. Identification of Subclinical Language Deficit Using Machine Learning Classification Based on Poststroke Functional Connectivity Derived from Low Frequency Oscillations. Brain Connect 2019; 9:194-208. [PMID: 30398379 DOI: 10.1089/brain.2018.0597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Post-stroke neuropsychological evaluation is time-intensive in assessing impairments in subjects without overt clinical deficits. We utilized functional connectivity (FC) from ten-minute non-invasive resting-state functional MRI (rs-fMRI) to identify stroke subjects at risk for subclinical language deficit (SLD) using machine learning. Discriminative ability of FC derived from slow-5 (0.01-0.027 Hz), slow-4 (0.027-0.073 Hz) and low frequency oscillations (LFO; 0.01-0.1 Hz) was compared. Sixty clinically non-aphasic right-handed subjects were categorized into three subgroups based on stroke status and normalized verbal fluency (NVF) score: 20 ischemic early-stage stroke subjects at higher risk for SLD (LD+; mean VFS=-1.77), 20 ischemic early-stage stroke subjects with at risk for SLD (LD-; mean VFS=-0.05), 20 healthy controls (HC; mean VFS=0.29). T1-weighted and rs-fMRI were acquired within 30 days of stroke onset. Blood-oxygen-level-dependent signal was extracted within the language network. FC was evaluated and used by a multiclass support vector machine to classify test subject into a subgroup which was assessed by nested leave-one-out cross-validation. FC derived from slow-4 (70%) provided the best accuracy relative to LFO (65%) and slow-5 (50%), reasonably higher than random chance (33.33%). Using subgroup-specific accuracy, classification was best realized within slow-4 for LD+ (81.6%) and LD- (78.3%) and slow-4/LFO for HC (80%), i.e., early-stage stroke subjects showed a slow-4 FC dominance whereas HC also indicated the normalized involvement within LFO. While frontal FC differentiated stroke from healthy, occipital FC differentiated between the two stroke subgroups. Thus, stroke subjects at risk for SLD can be identified using rs-fMRI reasonably in an expedited manner.
Collapse
Affiliation(s)
- Rosaleena Mohanty
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin.,2 Department of Electrical Engineering, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin
| | - Veena A Nair
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin
| | - Neelima Tellapragada
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin
| | - Leroy M Williams
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin
| | - Theresa J Kang
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin
| | - Vivek Prabhakaran
- 1 Department of Radiology, Wisconsin Institute of Medical Research (WIMR), University of Wisconsin-Madison, Madison, Wisconsin.,3 Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin.,4 Department of Psychiatry, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
230
|
Abstract
Mild cognitive impairment (MCI) represents a transitional stage between healthy aging and dementia, and affects 10-15% of the population over the age of 65. The failure of drug trials in Alzheimer's disease (AD) treatment has shifted researchers' focus toward delaying progression from MCI to dementia, which would reduce the prevalence and costs of dementia profoundly. Diagnostic criteria for MCI increasingly emphasize the need for positive biomarkers to detect preclinical AD. The phenomenology of MCI comprises lower quality-of-life, greater symptoms of depression, and avoidant coping strategies including withdrawal from social engagement. Neurobiological features of MCI are hypoperfusion and hypometabolism in temporoparietal cortices, medial temporal lobe atrophy particularly in rhinal cortices, elevated tau and phosphorylated tau and decreased Aβ42 in cerebrospinal fluid, and brain Aβ42 deposition. Elevated tau can be identified in MCI, particularly in the entorhinal cortex, using positron emission tomography, and analysis of signal complexity using electroencephalography or magnetoencephalography holds promise as a biomarker. Assessment of MCI also relies on cognitive screening and neuropsychological assessment, but there is an urgent need for standardized cognitive tests to capitalize on recent discoveries in cognitive neuroscience that may lead to more sensitive measures of MCI. Cholinesterase inhibitors are frequently prescribed for MCI, despite the lack of evidence for their efficacy. Exercise and diet interventions hold promise for increasing reserve in MCI, and group psychoeducational programs teaching practical memory strategies appear effective. More work is needed to better understand the phenomenology and neurobiology of MCI, and how best to assess it and delay progression to dementia.
Collapse
|
231
|
Siano G, Varisco M, Caiazza MC, Quercioli V, Mainardi M, Ippolito C, Cattaneo A, Di Primio C. Tau Modulates VGluT1 Expression. J Mol Biol 2019; 431:873-884. [PMID: 30664870 DOI: 10.1016/j.jmb.2019.01.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 01/09/2019] [Indexed: 02/07/2023]
Abstract
Tau displacement from microtubules is the first step in the onset of tauopathies and is followed by toxic protein aggregation. However, other non-canonical functions of Tau might have a role in these pathologies. Here, we demonstrate that a small amount of Tau localizes in the nuclear compartment and accumulates in both the soluble and chromatin-bound fractions. We show that favoring Tau nuclear translocation and accumulation, by Tau overexpression or detachment from MTs, increases the expression of VGluT1, a disease-relevant gene directly involved in glutamatergic synaptic transmission. Remarkably, the P301L mutation, related to frontotemporal dementia FTDP-17, impairs this mechanism leading to a loss of function. Altogether, our results provide the demonstration of a direct physiological role of Tau on gene expression. Alterations of this mechanism may be at the basis of the onset of neurodegeneration.
Collapse
Affiliation(s)
- Giacomo Siano
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Martina Varisco
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | | | | | - Marco Mainardi
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | - Chiara Ippolito
- Unit of Histology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Antonino Cattaneo
- Laboratory of Biology, BIO@SNS, Scuola Normale Superiore, Pisa, Italy
| | | |
Collapse
|
232
|
Spyrou L, Parra M, Escudero J. Complex Tensor Factorization With PARAFAC2 for the Estimation of Brain Connectivity From the EEG. IEEE Trans Neural Syst Rehabil Eng 2019; 27:1-12. [DOI: 10.1109/tnsre.2018.2883514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
233
|
Risacher SL, Saykin AJ. Neuroimaging in aging and neurologic diseases. HANDBOOK OF CLINICAL NEUROLOGY 2019; 167:191-227. [PMID: 31753134 DOI: 10.1016/b978-0-12-804766-8.00012-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuroimaging biomarkers for neurologic diseases are important tools, both for understanding pathology associated with cognitive and clinical symptoms and for differential diagnosis. This chapter explores neuroimaging measures, including structural and functional measures from magnetic resonance imaging (MRI) and molecular measures primarily from positron emission tomography (PET), in healthy aging adults and in a number of neurologic diseases. The spectrum covers neuroimaging measures from normal aging to a variety of dementias: late-onset Alzheimer's disease [AD; including mild cognitive impairment (MCI)], familial and nonfamilial early-onset AD, atypical AD syndromes, posterior cortical atrophy (PCA), logopenic aphasia (lvPPA), cerebral amyloid angiopathy (CAA), vascular dementia (VaD), sporadic and familial behavioral-variant frontotemporal dementia (bvFTD), semantic dementia (SD), progressive nonfluent aphasia (PNFA), frontotemporal dementia with motor neuron disease (FTD-MND), frontotemporal dementia with amyotrophic lateral sclerosis (FTD-ALS), corticobasal degeneration (CBD), progressive supranuclear palsy (PSP), dementia with Lewy bodies (DLB), Parkinson's disease (PD) with and without dementia, and multiple systems atrophy (MSA). We also include a discussion of the appropriate use criteria (AUC) for amyloid imaging and conclude with a discussion of differential diagnosis of neurologic dementia disorders in the context of neuroimaging.
Collapse
Affiliation(s)
- Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
234
|
Tau impairs neural circuits, dominating amyloid-β effects, in Alzheimer models in vivo. Nat Neurosci 2018; 22:57-64. [PMID: 30559471 DOI: 10.1038/s41593-018-0289-8] [Citation(s) in RCA: 296] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/09/2018] [Indexed: 01/22/2023]
Abstract
The coexistence of amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the neocortex is linked to neural system failure and cognitive decline in Alzheimer's disease. However, the underlying neuronal mechanisms are unknown. By employing in vivo two-photon Ca2+ imaging of layer 2/3 cortical neurons in mice expressing human Aβ and tau, we reveal a dramatic tau-dependent suppression of activity and silencing of many neurons, which dominates over Aβ-dependent neuronal hyperactivity. We show that neurofibrillary tangles are neither sufficient nor required for the silencing, which instead is dependent on soluble tau. Surprisingly, although rapidly effective in tau mice, suppression of tau gene expression was much less effective in rescuing neuronal impairments in mice containing both Aβ and tau. Together, our results reveal how Aβ and tau synergize to impair the functional integrity of neural circuits in vivo and suggest a possible cellular explanation contributing to disappointing results from anti-Aβ therapeutic trials.
Collapse
|
235
|
Torrealba E, Garcia-Morales P, Cejudo JC, Diaz M, Rodriguez-Esparragon F, Fabre O, Mesa-Herrera F, Marin R, Sanchez-Garcia F, Rodriguez-Perez A, Gramunt N. In-Out-Test: A New Paradigm for Sorting the Wheat from the Chaff in Prodromal Alzheimer's Disease. J Alzheimers Dis 2018; 67:265-277. [PMID: 30530971 PMCID: PMC7836051 DOI: 10.3233/jad-171007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Assessment of hippocampal amnesia is helpful to distinguish between normal cognition and mild cognitive impairment (MCI), but not for identifying converters to dementia. Here biomarkers are useful but novel neuropsychological approaches are needed in their absence. The In-out-test assesses episodic memory using a new paradigm hypothesized to avoid reliance on executive function, which may compensate for damaged memory networks. OBJECTIVE To assess the validity of the In-out-test in identifying prodromal Alzheimer's disease (PAD) in a clinical setting, by comparing this to the Free and Cued Selective Reminding Test (FCSRT) and cerebrospinal fluid biomarkers. METHODS A cross-sectional study of 32 cognitively healthy, 32 MCI, and 30 progressive dementia subjects. All participants were given both the In-out-test and the FCSRT; 40 of them also received a lumbar puncture. RESULTS Internal consistency was demonstrated using Cronbach Alpha (r = 0.81) and Inter-rater reliability with Kappa (k = 0.94). Intraclass correlation (ICC) for test-retest reliability: r = 0.57 (p = 0.57). ICC between the In-out-test and FCSRT r = 0.87 (p = 0.001). ICC between the In-out-test and Aβ42 and P-tau/Aβ42 for controls: 0.73 and 0.75, respectively; P-tau for MCI: 0.77 and total sample: 0.70; Aβ42 for dementia: 0.71. All ICC measures between FCSRT and biomarkers were ≤0.264. AD diagnosis: In-out-test k = 0.71; FCSRT k = 0.49. PAD diagnosis (N = 35): In-out-test k = 0.69; FCSRT k = 0.44. CONCLUSIONS The In-out-test detected prodromal AD with a higher degree of accuracy than a conventional hippocampal-based memory test. These results suggest that this new paradigm could be of value in clinical settings, predicting which patients with MCI will go on to develop AD.
Collapse
Affiliation(s)
- Eduardo Torrealba
- Department of Neurology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Pilar Garcia-Morales
- Department of Psychiatry, Complejo Hospitalario Universitario Insular Materno-Infantil, Las Palmas de Gran Canaria, Spain
| | - Juan Carlos Cejudo
- Cognitive Impairment and Dementia Unit, Hospital Sagrat Cor. Hermanas Hospitalarias, Martorell, Barcelona, Spain
| | - Mario Diaz
- Department of Animal Biology, Laboratory of Membrane Physiology and Biophysics, Edaphology and Geology, University of La Laguna, Tenerife, Spain
| | | | - Oscar Fabre
- Department of Neurology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Fatima Mesa-Herrera
- Department of Animal Biology, Laboratory of Membrane Physiology and Biophysics, Edaphology and Geology, University of La Laguna, Tenerife, Spain
| | - Raquel Marin
- Department of Basic Medical Sciences, Laboratory of Cellular Neurobiology, Section Medicine, Faculty of Health Sciences, University of La Laguna, Tenerife, Spain
| | - Florentino Sanchez-Garcia
- Department of Immunology, Hospital Universitario de gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Aurelio Rodriguez-Perez
- Department of Anesthesia, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Nina Gramunt
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| |
Collapse
|
236
|
Foster CM, Kennedy KM, Horn MM, Hoagey DA, Rodrigue KM. Both hyper- and hypo-activation to cognitive challenge are associated with increased beta-amyloid deposition in healthy aging: A nonlinear effect. Neuroimage 2018; 166:285-292. [PMID: 29108941 DOI: 10.1016/j.neuroimage.2017.10.068] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/26/2017] [Accepted: 10/31/2017] [Indexed: 11/29/2022] Open
Abstract
Beta-amyloid (Aβ) positive individuals hyper-activate brain regions compared to those not at-risk; however, hyperactivation is then thought to diminish as Alzheimer's disease symptomatology begins, evidencing eventual hypoactivation. It remains unclear when in the disease staging this transition occurs. We hypothesized that differential levels of amyloid burden would be associated with both increased and decreased activation (i.e., a quadratic trajectory) in cognitively-normal adults. Participants (N = 62; aged 51-94) underwent an fMRI spatial distance-judgment task and Amyvid-PET scanning. Voxelwise regression modeled age, linear-Aβ, and quadratic-Aβ as predictors of BOLD activation to difficult spatial distance-judgments. A significant quadratic-Aβ effect on BOLD response explained differential activation in bilateral angular/temporal and medial prefrontal cortices, such that individuals with slightly elevated Aβ burden exhibited hyperactivation whereas even higher Aβ burden was then associated with hypoactivation. Importantly, in high-Aβ individuals, Aβ load moderated the effect of BOLD activation on behavioral task performance, where in lower-elevation, greater deactivation was associated with better accuracy, but in higher-elevation, greater deactivation was associated with poorer accuracy during the task. This study reveals a dose-response, quadratic relationship between increasing Aβ burden and alterations in BOLD activation to cognitive challenge in cognitively-normal individuals that suggests 1) the shift from hyper-to hypo-activation may begin early in disease staging, 2) depends, in part, on degree of Aβ burden, and 3) tracks cognitive performance.
Collapse
Affiliation(s)
- Chris M Foster
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Marci M Horn
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - David A Hoagey
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX 75235, USA.
| |
Collapse
|
237
|
Kennedy KM, Foster CM, Rodrigue KM. Increasing beta-amyloid deposition in cognitively healthy aging predicts nonlinear change in BOLD modulation to difficulty. Neuroimage 2018; 183:142-149. [PMID: 30102997 PMCID: PMC6197922 DOI: 10.1016/j.neuroimage.2018.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/23/2018] [Accepted: 08/09/2018] [Indexed: 11/24/2022] Open
Abstract
Recent evidence indicates that the relationship between increased beta-amyloid (Aβ) deposition and functional task-activation can be characterized by a non-linear trajectory of change in functional activation (Foster et al., 2017), explaining mixed results in prior literature showing both increases and decreases in activation as a function of beta-amyloid burden in cognitively normal adults. Here we sought to replicate this nonlinear effect in the same sample using a different functional paradigm to test the generalizability of this phenomenon. Participants (N = 68 healthy adults aged 49-94) underwent fMRI (0-, 2-, 3-, 4-back working memory task; WM) and 18F-Florbetapir PET scanning. A parametric WM load contrast was used as the dependent variable in a model with age, mean cortical Aβ, and Aβ2 as predictors. Results revealed that nonlinear amyloid (Aβ2) was a significant negative predictor of modulation of activation to WM load in two large inferior clusters: bilateral subcortical nuclei and bilateral lateral cerebellum. Individuals with slightly elevated Aβ burden evidenced greater modulation as compared to individuals with little or no Aβ burden, whereas individuals with the greatest Aβ burden evidenced lesser modulation as compared to individuals with slightly elevated Aβ. Increased modulation to WM load predicted better task accuracy and executive function measured outside the scanner. The current study provides further evidence for a dose-response, nonlinear relationship between increasing Aβ burden and alteration in brain activation in cognitively healthy adults, extending the existing evidence to dynamic range of activation to task difficulty, and reconciling seemingly discrepant effects of amyloid on brain function.
Collapse
Affiliation(s)
- Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, 75235, USA.
| | - Chris M Foster
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, 75235, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, 75235, USA
| |
Collapse
|
238
|
Simon SS, Hampstead BM, Nucci MP, Duran FLS, Fonseca LM, Martin MDGM, Ávila R, Porto FHG, Brucki SMD, Martins CB, Tascone LS, Amaro E, Busatto GF, Bottino CMC. Cognitive and Brain Activity Changes After Mnemonic Strategy Training in Amnestic Mild Cognitive Impairment: Evidence From a Randomized Controlled Trial. Front Aging Neurosci 2018; 10:342. [PMID: 30483113 PMCID: PMC6243115 DOI: 10.3389/fnagi.2018.00342] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Background: Mnemonic strategy training (MST) has been shown to improve cognitive performance in amnestic mild cognitive impairment (a-MCI), however, several questions remain unresolved. The goal of the present study was to replicate earlier pilot study findings using a randomized controlled design and to evaluate transfer effects and changes in brain activation. Methods: Thirty patients with a-MCI were randomized into MST or education program. At baseline, participants completed clinical and neuropsychological assessments as well as structural and functional magnetic resonance imaging (fMRI). Interventions were administered individually and comprised four sessions, over 2 weeks. MST taught patients to use a three-step process to learn and recall face-name associations. Post-treatment assessment included fMRI, a separate face-name association task, neuropsychological tests, and measures of metamemory. Behavioral (i.e., non-fMRI) measures were repeated after one and 3-months. Results: Participants in the MST condition showed greater improvement on measures of face-name memory, and increased associative strategy use; effects that were accompanied by increased fMRI activation in the left anterior temporal lobe. While all participants reported greater contentment with their everyday memory following intervention, only the MST group reported significant improvements in their memory abilities. There was no clear indication of far-transfer effects to other neuropsychological tests. Conclusion: Results demonstrate that patients with a-MCI not only show stimulus specific benefits of MST, but that they appear capable of transferring training to at least some other cognitive tasks. MST also facilitated the use of brain regions that are involved in face processing, episodic and semantic memory, and social cognition, which are consonant with the cognitive processes engaged by training.
Collapse
Affiliation(s)
- Sharon S. Simon
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Benjamin M. Hampstead
- Division of Neuropsychology, Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
- VA Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Mariana P. Nucci
- Neuroimagem Funcional – Laboratory of Medical Investigations on Magnetic Resonance Imaging (LIM-44), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Fábio L. S. Duran
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Luciana M. Fonseca
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Maria da Graça M. Martin
- Neuroimagem Funcional – Laboratory of Medical Investigations on Magnetic Resonance Imaging (LIM-44), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Renata Ávila
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Fábio H. G. Porto
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Sônia M. D. Brucki
- Department of Neurology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Camila B. Martins
- Department of Preventive Medicine, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Lyssandra S. Tascone
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Edson Amaro
- Neuroimagem Funcional – Laboratory of Medical Investigations on Magnetic Resonance Imaging (LIM-44), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Geraldo F. Busatto
- Laboratory of Psychiatric Neuroimaging (LIM-21), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Cássio M. C. Bottino
- Old Age Research Group (PROTER), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
239
|
Eavani H, Habes M, Satterthwaite TD, An Y, Hsieh MK, Honnorat N, Erus G, Doshi J, Ferrucci L, Beason-Held LL, Resnick SM, Davatzikos C. Heterogeneity of structural and functional imaging patterns of advanced brain aging revealed via machine learning methods. Neurobiol Aging 2018; 71:41-50. [PMID: 30077821 PMCID: PMC6162110 DOI: 10.1016/j.neurobiolaging.2018.06.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 12/19/2022]
Abstract
Disentangling the heterogeneity of brain aging in cognitively normal older adults is challenging, as multiple co-occurring pathologic processes result in diverse functional and structural changes. Capitalizing on machine learning methods applied to magnetic resonance imaging data from 400 participants aged 50 to 96 years in the Baltimore Longitudinal Study of Aging, we constructed normative cross-sectional brain aging trajectories of structural and functional changes. Deviations from typical trajectories identified individuals with resilient brain aging and multiple subtypes of advanced brain aging. We identified 5 distinct phenotypes of advanced brain aging. One group included individuals with relatively extensive structural and functional loss and high white matter hyperintensity burden. Another subgroup showed focal hippocampal atrophy and lower posterior-cingulate functional coherence, low white matter hyperintensity burden, and higher medial-temporal connectivity, potentially reflecting high brain tissue reserve counterbalancing brain loss that is consistent with early stages of Alzheimer's disease. Other subgroups displayed distinct patterns. These results indicate that brain changes should not be measured seeking a single signature of brain aging but rather via methods capturing heterogeneity and subtypes of brain aging. Our findings inform future studies aiming to better understand the neurobiological underpinnings of brain aging imaging patterns.
Collapse
Affiliation(s)
- Harini Eavani
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Theodore D Satterthwaite
- Department of Psychiatry, Brain Behavior Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Yang An
- National Institute on Aging, Baltimore, MD, USA
| | - Meng-Kang Hsieh
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Honnorat
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
240
|
Altimus CM, Keller K, Brannelly P, Ross E, Lin CT, Riley EAU, Briggs L, Smith J, Stevens M. Analysis of tauopathy research funding between 2006 and 2016 reveals critical gaps in research priorities. Alzheimers Dement 2018; 15:42-54. [PMID: 30314799 DOI: 10.1016/j.jalz.2018.07.218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/09/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
Neurodegenerative diseases encompass a range of diagnoses, such as Alzheimer's disease and Parkinson's disease. Despite decades of advancements in understanding the neurobiology of individual diseases, this class has few disease-modifying therapeutics and a paucity of biomarkers for diagnosis or progression. However, tau protein aggregation has emerged as a potential unifying factor across several neurodegenerative diseases, which has prompted a rapid growth in tau-related funding. In spite of this growth, research funding in this area is not in line with the immense magnitude of disease burden, and drug discovery and clinical research remain underfunded. Coordinated, collaborative efforts are key to making an impact, which can and should be led by the major funding bodies within the tau space. Here we describe the development and analysis of a tau-focused neurodegeneration funding database, which captures data from 2040 grants from 2006 to 2016. This database was developed as a public resource to allow funders, researchers, and policy makers to better understand tau funding patterns and to identify key funders and potential collaborations. This database can be used in conjunction with other neurodegenerative disease databases, such as the International Alzheimer's Disease Research Portfolio to gain specific insight into tau-research funding. Over the study period, overall tau funding rose dramatically; however, changes in capital distribution also changed. Specifically, the field experienced a strong bias toward funding tau in the context of Alzheimer's disease, while at the same time generally decreasing the overall proportion of funding for basic research, treatment development, and evaluation. As funding organizations look forward, this resource can both inform future funding strategies and priority areas and identify potential collaborative efforts with complementary funding organizations.
Collapse
Affiliation(s)
| | | | | | - Erin Ross
- Milken Institute, Washington, DC, USA
| | | | | | | | - Jeremy Smith
- Rainwater Charitable Foundation, Fort Worth, TX, USA
| | | |
Collapse
|
241
|
Eglen RM, Reisine T. Human iPS Cell-Derived Patient Tissues and 3D Cell Culture Part 1: Target Identification and Lead Optimization. SLAS Technol 2018; 24:3-17. [PMID: 30286296 DOI: 10.1177/2472630318803277] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human-induced pluripotent stem cells (HiPSCs), and new technologies to culture them into functional cell types and tissues, are now aiding drug discovery. Patient-derived HiPSCs can provide disease models that are more clinically relevant and so more predictive than the currently available animal-derived or tumor cell-derived cells. These cells, consequently, exhibit disease phenotypes close to the human pathology, particularly when cultured under conditions that allow them to recapitulate the tissue architecture in three-dimensional (3D) systems. A key feature of HiPSCs is that they can be cultured under conditions that favor formation of multicellular spheroids or organoids. By culturing and differentiating in systems mimicking the human tissue in vivo, the HiPSC microenvironment further reflects patient in vivo physiology, pathophysiology, and ultimately pharmacological responsiveness. We assess the rationale for using HiPSCs in several phases of preclinical drug discovery, specifically in disease modeling, target identification, and lead optimization. We also discuss the growing use of HiPSCs in compound lead optimization, particularly in profiling compounds for their potential metabolic liability and off-target toxicities. Collectively, we contend that both approaches, HiPSCs and 3D cell culture, when used in concert, have exciting potential for the development of novel medicines.
Collapse
|
242
|
Delhaye E, Bahri MA, Salmon E, Bastin C. Impaired perceptual integration and memory for unitized representations are associated with perirhinal cortex atrophy in Alzheimer's disease. Neurobiol Aging 2018; 73:135-144. [PMID: 30342274 DOI: 10.1016/j.neurobiolaging.2018.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 11/27/2022]
Abstract
Unitization, the capacity to encode associations as one integrated entity, can enhance associative memory in populations with an associative memory deficit by promoting familiarity-based associative recognition. Patients with Alzheimer's disease (AD) are typically impaired in associative memory compared with healthy controls but do not benefit from unitization strategies. Using fragmented pictures of objects, this study aimed at assessing which of the cognitive processes that compose unitization is actually affected in AD: the retrieval of unitized representations itself, or some earlier stages of processing, such as the integration process at a perceptual or conceptual stage of representation. We also intended to relate patients' object unitization capacity to the integrity of their perirhinal cortex (PrC), as the PrC is thought to underlie unitization and is also one of the first affected regions in AD. We evaluated perceptual integration capacity and subsequent memory for those items that have supposedly been unitized in 23 mild AD patients and 20 controls. We systematically manipulated the level of perceptual integration during encoding by presenting object pictures that were either left intact, separated into 2 fragments, or separated into 4 fragments. Subjects were instructed to unitize the fragments into a single representation. Success of integration was assessed by a question requiring the identification of the object. Participants also underwent a structural magnetic resonance imaging examination, and measures of PrC, posterior cingulate cortex volume and thickness, and hippocampal volume, were extracted. The results showed that patients' perceptual integration performance decreased with the increased fragmentation level and that their memory for unitized representations was impaired whatever the demands in terms of perceptual integration at encoding. Both perceptual integration and memory for unitized representations were related to the integrity of the PrC, and memory for unitized representations was also related to the volume of the hippocampus. We argue that, globally, this supports representational theories of memory that hold that the role of the PrC is not only perceptual nor mnemonic but instead underlies complex object representation.
Collapse
Affiliation(s)
- Emma Delhaye
- GIGA-CRC In-Vivo Imaging, University in Liège, Liège, Belgium; PsyNCog, Faculty of Psychology, University in Liège, Liège, Belgium.
| | | | - Eric Salmon
- GIGA-CRC In-Vivo Imaging, University in Liège, Liège, Belgium; PsyNCog, Faculty of Psychology, University in Liège, Liège, Belgium; Memory Clinic, CHU Liege, University in Liège, Liège, Belgium
| | - Christine Bastin
- GIGA-CRC In-Vivo Imaging, University in Liège, Liège, Belgium; PsyNCog, Faculty of Psychology, University in Liège, Liège, Belgium
| |
Collapse
|
243
|
Sinha N, Berg CN, Tustison NJ, Shaw A, Hill D, Yassa MA, Gluck MA. APOE ε4 status in healthy older African Americans is associated with deficits in pattern separation and hippocampal hyperactivation. Neurobiol Aging 2018; 69:221-229. [PMID: 29909179 PMCID: PMC6392014 DOI: 10.1016/j.neurobiolaging.2018.05.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 12/30/2022]
Abstract
African Americans are 1.4 times more likely than European Americans to carry the apolipoprotein E (APOE) ε4 allele, a risk factor for Alzheimer's disease (AD). However, little is known about the neural correlates of cognitive function in older African Americans and how they relate to genetic risk for AD. In particular, no past study on African Americans has examined the effect of APOE ε4 status on pattern separation-mnemonic discrimination performance and its corresponding neural computations in the hippocampus. Previous work using the mnemonic discrimination paradigm has localized increased activation in the DG/CA3 hippocampal subregions as being correlated with discrimination deficits. In a case-control high-resolution functional magnetic resonance imaging study of 30 healthy African Americans, aged 60 years and older, we observed APOE ε4-related impairments in mnemonic discrimination, coincident with dysfunctional hyperactivation in the DG/CA3, and CA1 regions, despite no evidence of structural differences in the hippocampus between carriers and noncarriers. Our results add to the growing body of evidence that deficits in pattern separation may be an early marker for AD-related neuronal dysfunction.
Collapse
Affiliation(s)
- Neha Sinha
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, USA.
| | - Chelsie N Berg
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, USA
| | - Nicholas J Tustison
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA; Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ashlee Shaw
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, USA
| | - Diane Hill
- Office of University-Community Partnerships, Rutgers University-Newark, Newark, NJ, USA
| | - Michael A Yassa
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA, USA
| | - Mark A Gluck
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, NJ, USA.
| |
Collapse
|
244
|
Suchy Y, Franchow EI, Niermeyer MA, Ziemnik RE, Williams PG, Pennington NN. Exaggerated reaction to novelty as a predictor of incipient cognitive decline among community-dwelling older adults. J Clin Exp Neuropsychol 2018; 40:987-999. [PMID: 30112978 DOI: 10.1080/13803395.2018.1457627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The ability to detect covert markers of incipient cognitive decline among older adults before cognitive decline becomes overtly evident on traditional cognitive tests represents an important topic of research. Exaggerated reactions to novelty, reflected in novelty-induced increases in action planning latencies ("novelty effect"; NE) and low openness to experience (openness), have been previously associated with incipient cognitive decline among older adults who appeared cognitively normal at baseline. The purposes of the present study were to extend prior research on the utility of these markers by examining whether (a) NE and openness each predict cognitive change uniquely, and (b) whether these indices predict cognitive change above and beyond measures of memory, executive functions, processing speed/efficiency, premorbid IQ, and depressive symptoms. METHOD Sixty-one cognitively normal community-dwelling older adults were administered a battery of tests assessing the relevant constructs at baseline and one-year follow up. Changes in cognitive status were assessed using the Demetia Rating Scale, 2nd Edition, NE was assessed using the Push-Turn-Taptap task (an electronic motor sequence learning task), and openness using the NEO Personality Inventory-Revised. The Test of Premorbid Functioning, and subtests from Repeatable Battery for the Assessment of Neuropsychological Status, Delis-Kaplan Executive Function System, and Wechsler Adult Intelligence Scale, 4th Edition, were used for additional assessment of baseline cognition. Depressive symptoms were assessed using the Geriatric Depression Scale. RESULTS Results confirmed our hypothesis that both NE and openness contribute to prediction of cognitive change beyond baseline cognition and depressive symptoms, but none of the covariates (i.e., depression, executive functions, processing efficiency, or memory) themselves contributed to the model. NE and openness each contributed unique variance and were independent of each other. CONCLUSIONS Openness and NE have the potential to provide evidence-based methods for estimating risk of future cognitive change in persons with currently normal standardized test scores.
Collapse
Affiliation(s)
- Yana Suchy
- a Department of Psychology , University of Utah , Salt Lake City , UT , USA
| | - Emilie I Franchow
- a Department of Psychology , University of Utah , Salt Lake City , UT , USA.,b Behavioral Healthcare Line , New Mexico Veterans Affairs Health Care System , Albuquerque , NM , USA
| | | | - Rosemary E Ziemnik
- a Department of Psychology , University of Utah , Salt Lake City , UT , USA
| | - Paula G Williams
- a Department of Psychology , University of Utah , Salt Lake City , UT , USA
| | | |
Collapse
|
245
|
Merlo S, Spampinato SF, Sortino MA. Early compensatory responses against neuronal injury: A new therapeutic window of opportunity for Alzheimer's Disease? CNS Neurosci Ther 2018; 25:5-13. [PMID: 30101571 DOI: 10.1111/cns.13050] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by extensive neurodegeneration and inflammation in selective brain areas, linked to severely disabling cognitive deficits. Before full manifestation, different stages appear with progressively increased brain pathology and cognitive impairment. This significantly extends the time lag between initial molecular triggers and appearance of detectable symptoms. Notably, a number of studies in the last decade have revealed that in the early stage of mild cognitive impairment, events that appear in contrast with neuronal distress may occur. These have been reproduced in vitro and in animal models and include increase in synaptic elements, increase in synaptic and metabolic activity, enhancement of neurotrophic milieu and changes in glial cell reactivity and inflammation. They have been interpreted as compensatory responses that could either delay disease progression or, in the long run, result detrimental. For this reason, these mechanisms define a new and previously undervalued window of opportunity for intervention. Their importance resides especially in their early appearance. Directing efforts to better characterize this stage, in order to identify new pharmacological targets, is an exciting new avenue to future advances in AD research.
Collapse
Affiliation(s)
- Sara Merlo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| |
Collapse
|
246
|
Jin N, Lipponen A, Koivisto H, Gurevicius K, Tanila H. Increased cortical beta power and spike-wave discharges in middle-aged APP/PS1 mice. Neurobiol Aging 2018; 71:127-141. [PMID: 30138766 DOI: 10.1016/j.neurobiolaging.2018.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/12/2018] [Accepted: 07/14/2018] [Indexed: 12/11/2022]
Abstract
Amyloid plaque-forming transgenic mice display neuronal hyperexcitability, epilepsy, and sudden deaths in early adulthood. However, it is unknown whether hyperexcitability persists until middle ages when memory impairment manifests. We recorded multichannel video electroencephalography (EEG), local field potentials, and auditory evoked potentials in transgenic mice carrying mutated human amyloid precursor protein (APP) and presenilin-1 (PS1) genes and wild-type littermates at 14-16 months and compared the results with data we have earlier collected from 4-month-old mice. Furthermore, we monitored acoustic startle responses in other APP/PS1 and wild-type mice from 3 to 11 months of age. Independent of the age APP/PS1 mice demonstrated increased cortical power at 8-60 Hz. They also displayed over 5-fold increase in the occurrence of spike-wave discharges and augmented auditory evoked potentials compared with nontransgenic littermates. In contrast to evoked potentials, APP/PS1 mice showed normalization of acoustic startle responses with aging. Increased cortical power and spike-wave discharges provide powerful new biomarkers to monitor progression of amyloid pathology in preclinical intervention studies.
Collapse
Affiliation(s)
- Nanxiang Jin
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Arto Lipponen
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | | | | | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
247
|
Bråthen ACS, de Lange AMG, Rohani DA, Sneve MH, Fjell AM, Walhovd KB. Multimodal cortical and hippocampal prediction of episodic-memory plasticity in young and older adults. Hum Brain Mapp 2018; 39:4480-4492. [PMID: 30004603 DOI: 10.1002/hbm.24287] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/20/2018] [Accepted: 06/16/2018] [Indexed: 12/31/2022] Open
Abstract
Episodic memory can be trained in both early and late adulthood, but there is considerable variation in cognitive improvement across individuals. Which brain characteristics make some individuals benefit more than others? We used a multimodal approach to investigate whether volumetric magnetic resonance imaging (MRI) and resting-state functional MRI characteristics of the cortex and hippocampus, brain regions involved in episodic-memory function, were predictive of cognitive improvement after memory training. We hypothesized that these brain characteristics would differentially predict memory improvement in young and older adults, given the vulnerability of cortical regions as well as the hippocampus to healthy aging. Following structural and resting-state activity magnetic resonance scans, 50 young and 76 older participants completed 10 weeks of strategic episodic-memory training. Both age groups improved their memory performance, but the young adults more so than the older. Vertex-wise analyses of cortical volume showed no significant relation to memory benefit. When analyzing the two age groups separately, hippocampal volume was predictive of memory improvement in the group of older participants only. In this age group, the lower resting-state activity of the hippocampus was also predictive of memory improvement. Both volumetric and resting-state characteristics of the hippocampus explained unique variance of the improvement in the older participants suggesting that a multimodal imaging approach is valuable for the understanding of mechanisms underlying memory plasticity in aging.
Collapse
Affiliation(s)
- Anne Cecilie Sjøli Bråthen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Ann-Marie Glasø de Lange
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Darius A Rohani
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Markus H Sneve
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
248
|
Apple AC, Schroeder MP, Ryals AJ, Wagner LI, Cella D, Shih PA, Reilly J, Penedo FJ, Voss JL, Wang L. Hippocampal functional connectivity is related to self-reported cognitive concerns in breast cancer patients undergoing adjuvant therapy. NEUROIMAGE-CLINICAL 2018; 20:110-118. [PMID: 30094161 PMCID: PMC6077172 DOI: 10.1016/j.nicl.2018.07.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 06/28/2018] [Accepted: 07/10/2018] [Indexed: 02/06/2023]
Abstract
Nearly three out of four survivors experience Cancer-Related Cognitive Impairment (CRCI) for months or years following treatment. Both clinical and animal studies point to the hippocampus as a likely brain region affected in CRCI, however no previous study has investigated the functional connectivity of the hippocampus in CRCI. We compared hippocampal connectivity in cancer survivors and healthy controls and tested the relationship between functional connectivity differences and measures of objective and subjective cognition. Exploratory analysis of inflammatory markers was conducted in a small subset of participants as well. FMRI data were acquired during a memory task from 16 breast cancer survivors and 17 controls. The NIH Toolbox was used to assess cognitive performance and Neuro-QoL was used to measure self-reported cognitive concerns. Whole-brain group-level comparisons identified clusters with different connectivity to the hippocampus in survivors versus controls during task. Average connectivity was extracted from clusters of significant difference between the groups and correlated with cognitive performance and subjective report. Survivors performed worse on a test of episodic memory and reported greater cognitive concern than controls. Exploratory analysis found higher IL6 in cancer survivors compared to controls. Cancer survivors demonstrated higher connectivity of hippocampus with left cuneus, left lingual, left precuneus, and right middle prefrontal gyrus compared with controls. In survivors, higher task-related hippocampal-cortical connectivity was related to worse subjective measures of cognitive concern. Of the four significant clusters, higher connectivity of the precuneus with hippocampus was significantly associated with worse cognitive concern in survivors. The observed greater hippocampal-cortical connectivity in survivors compared to controls is the first reported fMRI biomarker of subjective concern, and may represent a compensatory response to cancer and its treatments. This compensation could explain, in part, the subjective feelings of cognitive impairment that were reported by survivors. Cancer survivors performed worse on a test of episodic memory and reported greater cognitive concern than controls Cancer survivors demonstrated significantly higher hippocampal-cortical connectivity Higher functional connectivity was associated with worse self-reported cognitive functioning in cancer survivors
Collapse
Affiliation(s)
- Alexandra C Apple
- Division of Clinical Psychology, Northwestern University Feinberg School of Medicine, United States; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States.
| | - Matthew P Schroeder
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States
| | - Anthony J Ryals
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States
| | - Lynne I Wagner
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, United States
| | - David Cella
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States; Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, United States; Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, United States
| | - Pei-An Shih
- Department of Psychiatry, University of California, San Diego, United States
| | - James Reilly
- Division of Clinical Psychology, Northwestern University Feinberg School of Medicine, United States; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States
| | - Frank J Penedo
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, United States
| | - Joel L Voss
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States; Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, United States; Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, United States
| | - Lei Wang
- Division of Clinical Psychology, Northwestern University Feinberg School of Medicine, United States; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, United States; Department of Radiology, Northwestern University Feinberg School of Medicine, United States
| |
Collapse
|
249
|
Bayram E, Caldwell JZK, Banks SJ. Current understanding of magnetic resonance imaging biomarkers and memory in Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2018; 4:395-413. [PMID: 30229130 PMCID: PMC6140335 DOI: 10.1016/j.trci.2018.04.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Alzheimer's disease (AD) is caused by a cascade of changes to brain integrity. Neuroimaging biomarkers are important in diagnosis and monitoring the effects of interventions. As memory impairments are among the first symptoms of AD, the relationship between imaging findings and memory deficits is important in biomarker research. The most established magnetic resonance imaging (MRI) finding is hippocampal atrophy, which is related to memory decline and currently used as a diagnostic criterion for AD. While the medial temporal lobes are impacted early by the spread of neurofibrillary tangles, other networks and regional changes can be found quite early in the progression. Atrophy in several frontal and parietal regions, cortical thinning, and white matter alterations correlate with memory deficits in early AD. Changes in activation and connectivity have been detected by functional MRI (fMRI). Task-based fMRI studies have revealed medial temporal lobe hypoactivation, parietal hyperactivation, and frontal hyperactivation in AD during memory tasks, and activation patterns of these regions are also altered in preclinical and prodromal AD. Resting state fMRI has revealed alterations in default mode network activity related to memory in early AD. These studies are limited in part due to the historic inclusion of patients who had suspected AD but likely did not have the disorder. Modern biomarkers allow for more diagnostic certainty, allowing better understanding of neuroimaging markers in true AD, even in the preclinical stage. Larger patient cohorts, comparison of candidate imaging biomarkers to more established biomarkers, and inclusion of more detailed neuropsychological batteries to assess multiple aspects of memory are needed to better understand the memory deficit in AD and help develop new biomarkers. This article reviews MRI findings related to episodic memory impairments in AD and introduces a new study with multimodal imaging and comprehensive neuropsychiatric evaluation to overcome current limitations.
Collapse
Affiliation(s)
- Ece Bayram
- Department of Neurology, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Jessica Z K Caldwell
- Department of Neurology, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Sarah J Banks
- Department of Neurology, Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| |
Collapse
|
250
|
Novel interaction between Alzheimer's disease-related protein presenilin 1 and glutamate transporter 1. Sci Rep 2018; 8:8718. [PMID: 29880815 PMCID: PMC5992168 DOI: 10.1038/s41598-018-26888-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/18/2018] [Indexed: 12/28/2022] Open
Abstract
Neuronal hyperactivity is one of the earliest events observed in Alzheimer’s disease (AD). Moreover, alterations in the expression of glutamate transporters have been reported to exacerbate amyloid pathology and cognitive deficits in transgenic AD mouse models. However, the molecular links between these pathophysiological changes remain largely unknown. Here, we report novel interaction between presenilin 1 (PS1), the catalytic component of the amyloid precursor protein-processing enzyme, γ-secretase, and a major glutamate transporter-1 (GLT-1). Our data demonstrate that the interaction occurs between PS1 and GLT-1 expressed at their endogenous levels in vivo and in vitro, takes place in both neurons and astrocytes, and is independent of the PS1 autoproteolysis and γ-secretase activity. This intriguing discovery may shed light on the molecular crosstalk between the proteins linked to the maintenance of glutamate homeostasis and Aβ pathology.
Collapse
|