1
|
Cicali KA, Tapia-Rojas C. Synaptic mitochondria: A crucial factor in the aged hippocampus. Ageing Res Rev 2024; 101:102524. [PMID: 39369797 DOI: 10.1016/j.arr.2024.102524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
Aging is a multifaceted biological process characterized by progressive molecular and cellular damage accumulation. The brain hippocampus undergoes functional deterioration with age, caused by cellular deficits, decreased synaptic communication, and neuronal death, ultimately leading to memory impairment. One of the factors contributing to this dysfunction is the loss of mitochondrial function. In neurons, mitochondria are categorized into synaptic and non-synaptic pools based on their location. Synaptic mitochondria, situated at the synapses, play a crucial role in maintaining neuronal function and synaptic plasticity, whereas non-synaptic mitochondria are distributed throughout other neuronal compartments, supporting overall cellular metabolism and energy supply. The proper function of synaptic mitochondria is essential for synaptic transmission as they provide the energy required and regulate calcium homeostasis at the communication sites between neurons. Maintaining the structure and functionality of synaptic mitochondria involves intricate processes, including mitochondrial dynamics such as fission, fusion, transport, and quality control mechanisms. These processes ensure that mitochondria remain functional, replace damaged organelles, and sustain cellular homeostasis at synapses. Notably, deficiencies in these mechanisms have been increasingly associated with aging and the onset of age-related neurodegenerative diseases. Synaptic mitochondria from the hippocampus are particularly vulnerable to age-related changes, including alterations in morphology and a decline in functionality, which significantly contribute to decreased synaptic activity during aging. This review comprehensively explores the critical roles that mitochondrial dynamics and quality control mechanisms play in preserving synaptic activity and neuronal function. It emphasizes the emerging evidence linking the deterioration of synaptic mitochondria to the aging process and the development of neurodegenerative diseases, highlighting the importance of these organelles from hippocampal neurons as potential therapeutic targets for mitigating cognitive decline and synaptic degeneration associated with aging. The novelty of this review lies in its focus on the unique vulnerability of hippocampal synaptic mitochondria to aging, underscoring their importance in maintaining brain function across the lifespan.
Collapse
Affiliation(s)
- Karina A Cicali
- Laboratory of Neurobiology of Aging, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile.
| |
Collapse
|
2
|
Wang J, Liang X, Lu J, Zhang W, Chen Q, Li X, Chen J, Zhang X, Zhang B. Cortical and subcortical gray matter abnormalities in mild cognitive impairment. Neuroscience 2024; 557:81-88. [PMID: 39067683 DOI: 10.1016/j.neuroscience.2024.07.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Gray matter changes are thought to be closely related to cognitive decline in mild cognitive impairment (MCI) patients. The study aimed to explore cortical and subcortical structural alterations in MCI and their association with cognitive assessment. 24 MCI patients and 22 normal controls (NCs) were included. Voxel-based morphometry (VBM), vertex-based shape analysis and surface-based morphometry (SBM) analysis were applied to explore subcortical nuclei volume, shape and cortical morphology. Correlations between structural changes and cognition were explored using spearman correlation analysis. Support vector machine (SVM) classification evaluated MCI identification accuracy. MCI patients showed significant atrophy in the left thalamus, left hippocampus, left amygdala, right pallidum, right hippocampus, along with inward deformation in the left amygdala. SBM analysis revealed that MCI group exhibited shallower sulci depth in the left hemisphere and increased cortical gyrification index (GI) in the right frontal gyrus. Correlation analysis showed the positive correlation between right hippocampus volume and episodic memory, while negative correlation between the altered GI and memory performance in MCI group. SVM analysis demonstrated superior performance of sulci depth and GI derived from SBM in MCI identification. When combined with cortical and subcortical metrics, SVM achieved a peak accuracy of 89 % in distinguishing MCI from NC. The study reveals significant gray matter structural changes in MCI, suggesting their potential role in underlying functional differences and neural mechanisms behind memory impairment in MCI.
Collapse
Affiliation(s)
- Junxia Wang
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xue Liang
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Jiaming Lu
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wen Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Qian Chen
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xin Li
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Jiu Chen
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xin Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Bing Zhang
- Department of Radiology, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China; Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing 210008, China.
| |
Collapse
|
3
|
Keeler JL, Konyn CY, Treasure J, Cardi V, Himmerich H, Tchanturia K, Mycroft H. "Your mind doesn't have room for anything else": a qualitative study of perceptions of cognitive functioning during and after recovery from anorexia nervosa. J Eat Disord 2022; 10:201. [PMID: 36575533 PMCID: PMC9793561 DOI: 10.1186/s40337-022-00723-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Past research has indicated the presence of cognitive difficulties in individuals with anorexia nervosa (AN), although it is unclear how these are experienced in real life. Moreover, it is unclear how and whether the experience of cognitive difficulties changes in nature and intensity over the course of the illness and following recovery. METHODS Twenty-one female participants (AN = 11; recovered AN = 10) participated in online semi-structured interviews, utilising open-ended questions and exploring topics relating to their experiences of their cognitive function, changes over time and their perspectives on the future. Reflexive thematic analysis was conducted on the resulting transcripts. RESULTS Six themes were identified, centred around the effects of the illness on mental and physical function, cognition, adaptation to living with the illness, similarities between AN and other psychopathology, tentative optimism for the future and recovery as a representation of liberation. Whilst respondents with AN appeared to perform remarkably well in their professional and educational lives, the cognitive difficulties were described as significantly impacting their ability to engage in life, particularly in the context of leisure and socialising. Respondents recovered from AN noted the importance of developing a non-AN identity as well as better emotion-regulation skills as central to recovery. CONCLUSIONS Whilst people with AN may be able to adapt their lives to the demands of the illness, they report significant cognitive difficulties that interfere with their engagement in many aspects of daily life. This should be considered by professionals working in the clinical management of AN. Treatments focused on ameliorating cognitive difficulties, and promoting emotional regulation and identity in AN are warranted.
Collapse
Affiliation(s)
- Johanna Louise Keeler
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 103 Denmark Hill, Section of Eating Disorders, London, SE5 8AF, UK.
| | - Carol Yael Konyn
- Department of Social, Genetic & Developmental Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Janet Treasure
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 103 Denmark Hill, Section of Eating Disorders, London, SE5 8AF, UK.,Bethlem Royal Hospital, South London and Maudsley NHS Foundation Trust, Monks Orchard Road, Beckenham, Kent, BR3 3BX, UK
| | - Valentina Cardi
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 103 Denmark Hill, Section of Eating Disorders, London, SE5 8AF, UK.,Department of General Psychology, University of Padova, Padua, Italy
| | - Hubertus Himmerich
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 103 Denmark Hill, Section of Eating Disorders, London, SE5 8AF, UK.,Bethlem Royal Hospital, South London and Maudsley NHS Foundation Trust, Monks Orchard Road, Beckenham, Kent, BR3 3BX, UK
| | - Kate Tchanturia
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, 103 Denmark Hill, Section of Eating Disorders, London, SE5 8AF, UK.,Bethlem Royal Hospital, South London and Maudsley NHS Foundation Trust, Monks Orchard Road, Beckenham, Kent, BR3 3BX, UK.,Illia State University, Tbilisi, Georgia.,Psychological Set Research and Correction Center, Tbilisi State Medical University, Tbilisi, Georgia
| | - Hazel Mycroft
- School of Psychology, University of Exeter, Exeter, Devon, UK
| |
Collapse
|
4
|
Ng KP, Qian X, Ng KK, Ji F, Rosa-Neto P, Gauthier S, Kandiah N, Zhou JH. Stage-dependent differential influence of metabolic and structural networks on memory across Alzheimer's disease continuum. eLife 2022; 11:e77745. [PMID: 36053063 PMCID: PMC9477498 DOI: 10.7554/elife.77745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Background Large-scale neuronal network breakdown underlies memory impairment in Alzheimer's disease (AD). However, the differential trajectories of the relationships between network organisation and memory across pathology and cognitive stages in AD remain elusive. We determined whether and how the influences of individual-level structural and metabolic covariance network integrity on memory varied with amyloid pathology across clinical stages without assuming a constant relationship. Methods Seven hundred and eight participants from the Alzheimer's Disease Neuroimaging Initiative were studied. Individual-level structural and metabolic covariance scores in higher-level cognitive and hippocampal networks were derived from magnetic resonance imaging and [18F] fluorodeoxyglucose positron emission tomography using seed-based partial least square analyses. The non-linear associations between network scores and memory across cognitive stages in each pathology group were examined using sparse varying coefficient modelling. Results We showed that the associations of memory with structural and metabolic networks in the hippocampal and default mode regions exhibited pathology-dependent differential trajectories across cognitive stages using sparse varying coefficient modelling. In amyloid pathology group, there was an early influence of hippocampal structural network deterioration on memory impairment in the preclinical stage, and a biphasic influence of the angular gyrus-seeded default mode metabolic network on memory in both preclinical and dementia stages. In non-amyloid pathology groups, in contrast, the trajectory of the hippocampus-memory association was opposite and weaker overall, while no metabolism covariance networks were related to memory. Key findings were replicated in a larger cohort of 1280 participants. Conclusions Our findings highlight potential windows of early intervention targeting network breakdown at the preclinical AD stage. Funding Data collection and sharing for this project was funded by the Alzheimer's Disease Neuroimaging Initiative (ADNI) (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). We also acknowledge the funding support from the Duke NUS/Khoo Bridge Funding Award (KBrFA/2019-0020) and NMRC Open Fund Large Collaborative Grant (OFLCG09May0035).
Collapse
Affiliation(s)
- Kok Pin Ng
- Department of Neurology, National Neuroscience InstituteSingaporeSingapore
- Duke-NUS Medical SchoolSingaporeSingapore
- Lee Kong Chian School of Medicine, Nanyang Technological University SingaporeSingaporeSingapore
| | - Xing Qian
- Centre for Sleep and Cognition and Centre for Translational MR Research,Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Kwun Kei Ng
- Centre for Sleep and Cognition and Centre for Translational MR Research,Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Fang Ji
- Centre for Sleep and Cognition and Centre for Translational MR Research,Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l’Ouest-de-l’Île-de-Montréal, and Departments of Neurology, Neurosurgery, Psychiatry, Pharmacology and Therapeutics, McGill UniversityMontrealCanada
- Montreal Neurological Institute, McGill UniversityMontrealCanada
| | - Serge Gauthier
- Department of Neurology & Neurosurgery, McGill UniversityMontrealCanada
| | - Nagaendran Kandiah
- Lee Kong Chian School of Medicine, Nanyang Technological University SingaporeSingaporeSingapore
| | - Juan Helen Zhou
- Centre for Sleep and Cognition and Centre for Translational MR Research,Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Department of Electrical and Computer Engineering, National University of SingaporeSingaporeSingapore
- Integrative Sciences and Engineering Programme (ISEP), National University of SingaporeSingaporeSingapore
| | | |
Collapse
|
5
|
Fixemer S, Ameli C, Hammer G, Salamanca L, Uriarte Huarte O, Schwartz C, Gérardy JJ, Mechawar N, Skupin A, Mittelbronn M, Bouvier DS. Microglia phenotypes are associated with subregional patterns of concomitant tau, amyloid-β and α-synuclein pathologies in the hippocampus of patients with Alzheimer's disease and dementia with Lewy bodies. Acta Neuropathol Commun 2022; 10:36. [PMID: 35296366 PMCID: PMC8925098 DOI: 10.1186/s40478-022-01342-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/26/2022] Open
Abstract
The cellular alterations of the hippocampus lead to memory decline, a shared symptom between Alzheimer’s disease (AD) and dementia with Lewy Bodies (DLB) patients. However, the subregional deterioration pattern of the hippocampus differs between AD and DLB with the CA1 subfield being more severely affected in AD. The activation of microglia, the brain immune cells, could play a role in its selective volume loss. How subregional microglia populations vary within AD or DLB and across these conditions remains poorly understood. Furthermore, how the nature of the hippocampal local pathological imprint is associated with microglia responses needs to be elucidated. To this purpose, we employed an automated pipeline for analysis of 3D confocal microscopy images to assess CA1, CA3 and DG/CA4 subfields microglia responses in post-mortem hippocampal samples from late-onset AD (n = 10), DLB (n = 8) and age-matched control (CTL) (n = 11) individuals. In parallel, we performed volumetric analyses of hyperphosphorylated tau (pTau), amyloid-β (Aβ) and phosphorylated α-synuclein (pSyn) loads. For each of the 32,447 extracted microglia, 16 morphological features were measured to classify them into seven distinct morphological clusters. Our results show similar alterations of microglial morphological features and clusters in AD and DLB, but with more prominent changes in AD. We identified two distinct microglia clusters enriched in disease conditions and particularly increased in CA1 and DG/CA4 of AD and CA3 of DLB. Our study confirms frequent concomitance of pTau, Aβ and pSyn loads across AD and DLB but reveals a specific subregional pattern for each type of pathology, along with a generally increased severity in AD. Furthermore, pTau and pSyn loads were highly correlated across subregions and conditions. We uncovered tight associations between microglial changes and the subfield pathological imprint. Our findings suggest that combinations and severity of subregional pTau, Aβ and pSyn pathologies transform local microglia phenotypic composition in the hippocampus. The high burdens of pTau and pSyn associated with increased microglial alterations could be a factor in CA1 vulnerability in AD.
Collapse
|
6
|
Chao FL, Zhang Y, Zhang L, Jiang L, Zhou CN, Tang J, Liang X, Fan JH, Dou XY, Tang Y. Fluoxetine Promotes Hippocampal Oligodendrocyte Maturation and Delays Learning and Memory Decline in APP/PS1 Mice. Front Aging Neurosci 2021; 12:627362. [PMID: 33519426 PMCID: PMC7838348 DOI: 10.3389/fnagi.2020.627362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Oligodendrogenesis dysfunction impairs memory consolidation in adult mice, and an oligodendrocyte abnormality is an important change occurring in Alzheimer's disease (AD). While fluoxetine (FLX) is known to delay memory decline in AD models, its effects on hippocampal oligodendrogenesis are unclear. Here, we subjected 8-month-old male amyloid precursor protein (APP)/presenilin 1 (PS1) mice to the FLX intervention for 2 months. Their exploratory behaviors and general activities in a novel environment, spatial learning and memory and working and reference memory were assessed using the open-field test, Morris water maze, and Y maze. Furthermore, changes in hippocampal oligodendrogenesis were investigated using stereology, immunohistochemistry, immunofluorescence staining, and Western blotting techniques. FLX delayed declines in the spatial learning and memory, as well as the working and reference memory of APP/PS1 mice. In addition, APP/PS1 mice exhibited immature hippocampal oligodendrogenesis, and FLX increased the numbers of 2'3'cyclic nucleotide 3'-phosphodiesterase (CNPase)+ and newborn CNPase+ oligodendrocytes in the hippocampi of APP/PS1 mice. Moreover, FLX increased the density of SRY-related HMG-box 10 protein (SOX10)+ cells and reduced the percentage of oligodendrocyte lineage cells displaying the senescence phenotype (CDKN2A/p16INK4a) in the hippocampus of APP/PS1 mice. Moreover, FLX had no effect on the serotonin (5-HT) 1A receptor (5-HT1AR) content or number of 5-HT1AR+ oligodendrocytes, but it reduced the content and activity of glycogen synthase kinase 3β (GSK3β) in the hippocampus of APP/PS1 transgenic mice. Taken together, FLX delays the senescence of oligodendrocyte lineage cells and promotes oligodendrocyte maturation in the hippocampus of APP/PS1 mice. FLX may regulate GSK3β through a mechanism other than 5-HT1AR and then inhibit the negative effect of GSK3β on oligodendrocyte maturation in the hippocampus of an AD mouse model.
Collapse
Affiliation(s)
- Feng-lei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Zhang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Lin Jiang
- Experimental Teaching Management Center, Chongqing Medical University, Chongqing, China
| | - Chun-ni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xin Liang
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Jin-hua Fan
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Xiao-yun Dou
- Academy of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Slachevsky A, Zitko P, Martínez-Pernía D, Forno G, Court FA, Lillo P, Villagra R, Duran-Aniotz C, Parrao T, Assar R, Orellana P, Toledo C, Rivera R, Ibañez A, Parra MA, González-Billault C, Amieva H, Thumala D. GERO Cohort Protocol, Chile, 2017-2022: Community-based Cohort of Functional Decline in Subjective Cognitive Complaint elderly. BMC Geriatr 2020; 20:505. [PMID: 33238908 PMCID: PMC7690082 DOI: 10.1186/s12877-020-01866-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/03/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND With the global population aging and life expectancy increasing, dementia has turned a priority in the health care system. In Chile, dementia is one of the most important causes of disability in the elderly and the most rapidly growing cause of death in the last 20 years. Cognitive complaint is considered a predictor for cognitive and functional decline, incident mild cognitive impairment, and incident dementia. The GERO cohort is the Chilean core clinical project of the Geroscience Center for Brain Health and Metabolism (GERO). The objective of the GERO cohort is to analyze the rate of functional decline and progression to clinical dementia and their associated risk factors in a community-dwelling elderly with subjective cognitive complaint, through a population-based study. We also aim to undertake clinical research on brain ageing and dementia disorders, to create data and biobanks with the appropriate infrastructure to conduct other studies and facilitate to the national and international scientific community access to the data and samples for research. METHODS The GERO cohort aims the recruitment of 300 elderly subjects (> 70 years) from Santiago (Chile), following them up for at least 3 years. Eligible people are adults not diagnosed with dementia with subjective cognitive complaint, which are reported either by the participant, a proxy or both. Participants are identified through a household census. The protocol for evaluation is based on a multidimensional approach including socio-demographic, biomedical, psychosocial, neuropsychological, neuropsychiatric and motor assessments. Neuroimaging, blood and stool samples are also obtained. This multidimensional evaluation is carried out in a baseline and 2 follow-ups assessments, at 18 and 36 months. In addition, in months 6, 12, 24, and 30, a telephone interview is performed in order to keep contact with the participants and to assess general well-being. DISCUSSION Our work will allow us to determine multidimensional risks factors associated with functional decline and conversion to dementia in elderly with subjective cognitive complain. The aim of our GERO group is to establish the capacity to foster cutting edge and multidisciplinary research on aging in Chile including basic and clinical research. TRIAL REGISTRATION NCT04265482 in ClinicalTrials.gov. Registration Date: February 11, 2020. Retrospectively Registered.
Collapse
Affiliation(s)
- Andrea Slachevsky
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile.
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile.
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile.
- Department of Neurology and Psychiatry, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile.
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| | - Pedro Zitko
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Health Service & Population Research Department, IoPPN, King's College London, London, UK
- Escuela de Salud Pública, Universidad de Chile, Santiago, Chile
| | - David Martínez-Pernía
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Gonzalo Forno
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
| | - Felipe A Court
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, USA
| | - Patricia Lillo
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- South Neurology Department, Faculty of Medicine, University of Chile, Santiago, Chile
- Unidad de Neurología, Hospital San José, Santiago, Chile
| | - Roque Villagra
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- East Neurology Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudia Duran-Aniotz
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Teresa Parrao
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Facultad de Psicología, Universidad Alberto Hurtado, Santiago, Chile
| | - Rodrigo Assar
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paulina Orellana
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Carolina Toledo
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Rodrigo Rivera
- Neuroradiologic Department, Instituto de Neurocirugia Asenjo, SSMO, Santiago, Chile
| | - Agustín Ibañez
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Universidad Autónoma del Caribe, Barranquilla, Colombia
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), California, USA
| | - Mario A Parra
- Universidad Autónoma del Caribe, Barranquilla, Colombia
- Psychology Department, School of Psychological Sciences & Health, University of Strathclyde, Glasgow, UK
| | - Christian González-Billault
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- The Buck Institute for Research on Aging, Novato, USA
- Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile
| | - Helena Amieva
- INSERM, Bordeaux Population Health Research Center, UMR 1219, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Daniela Thumala
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Escuela de Psicologia, Facultad de Ciencias Sociales, University of Chile, Santiago, Chile
| |
Collapse
|
8
|
Olesen MA, Torres AK, Jara C, Murphy MP, Tapia-Rojas C. Premature synaptic mitochondrial dysfunction in the hippocampus during aging contributes to memory loss. Redox Biol 2020; 34:101558. [PMID: 32447261 PMCID: PMC7248293 DOI: 10.1016/j.redox.2020.101558] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Aging is a process characterized by cognitive impairment and mitochondrial dysfunction. In neurons, these organelles are classified as synaptic and non-synaptic mitochondria depending on their localization. Interestingly, synaptic mitochondria from the cerebral cortex accumulate more damage and are more sensitive to swelling than non-synaptic mitochondria. The hippocampus is fundamental for learning and memory, synaptic processes with high energy demand. However, it is unknown if functional differences are found in synaptic and non-synaptic hippocampal mitochondria; and whether this could contribute to memory loss during aging. In this study, we used 3, 6, 12 and 18 month-old (mo) mice to evaluate hippocampal memory and the function of both synaptic and non-synaptic mitochondria. Our results indicate that recognition memory is impaired from 12mo, whereas spatial memory is impaired at 18mo. This was accompanied by a differential function of synaptic and non-synaptic mitochondria. Interestingly, we observed premature dysfunction of synaptic mitochondria at 12mo, indicated by increased ROS generation, reduced ATP production and higher sensitivity to calcium overload, an effect that is not observed in non-synaptic mitochondria. In addition, at 18mo both mitochondrial populations showed bioenergetic defects, but synaptic mitochondria were prone to swelling than non-synaptic mitochondria. Finally, we treated 2, 11, and 17mo mice with MitoQ or Curcumin (Cc) for 5 weeks, to determine if the prevention of synaptic mitochondrial dysfunction could attenuate memory loss. Our results indicate that reducing synaptic mitochondrial dysfunction is sufficient to decrease age-associated cognitive impairment. In conclusion, our results indicate that age-related alterations in ATP produced by synaptic mitochondria are correlated with decreases in spatial and object recognition memory and propose that the maintenance of functional synaptic mitochondria is critical to prevent memory loss during aging. Hippocampus-dependent learning and memory are impaired with age, which correlated with synaptic mitochondrial dysfunction. Synaptic mitochondria fail before non-synaptic mitochondria, indicating premature synaptic mitochondrial damage in aging. Reducing synaptic mitochondrial dysfunction, with MitoQ or Curcumin, decrease age-associated hippocampal memory impairment. Age-related changes in ATP production of synaptic mitochondria correlated with decreased hippocampal memory. Maintenance of functional synaptic mitochondria is critical to prevent memory loss during aging.
Collapse
Affiliation(s)
- Margrethe A Olesen
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Chile
| | - Angie K Torres
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Chile
| | - Claudia Jara
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Chile
| | - Michael P Murphy
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Universidad San Sebastián, Chile.
| |
Collapse
|
9
|
Jiang L, Ma J, Zhang Y, Zhou CN, Zhang L, Chao FL, Chen LM, Jiang R, Wu H, Tang Y. Effect of running exercise on the number of the neurons in the hippocampus of young transgenic APP/PS1 mice. Brain Res 2018; 1692:56-65. [DOI: 10.1016/j.brainres.2018.04.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/08/2018] [Accepted: 04/25/2018] [Indexed: 12/31/2022]
|
10
|
Qian L, Liu R, Qin R, Zhao H, Xu Y. The associated volumes of sub-cortical structures and cognitive domain in patients of Mild Cognitive Impairment. J Clin Neurosci 2018; 56:56-62. [PMID: 30029954 DOI: 10.1016/j.jocn.2018.07.010] [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: 01/17/2018] [Revised: 05/21/2018] [Accepted: 07/08/2018] [Indexed: 10/28/2022]
Abstract
This study aimed to explore the relationship between sub-cortical structures alterations and the cognitive domains in Mild Cognitive Impairment (MCI) patients, expected to find identifying sub-cortical structure markers of MCI progression to dementia. A total of 67 MCI patients (8 subjects refused to follow up) were recruited, who were divided into 21 stable MCI (sMCI) and 38 progress MCI (pMCI), according to cognitive assays. FreeSurfer software was used to perform volumetric measurements of the sub-cortical structures from 3.0 T magnetic resonance scans. Data revealed that pMCI subjects had lower scores in memory, language, executive and visual spatial compared with sMCI subjects. Compared with the sMCI group, the volume of the left thalamus, bilateral hippocampus, corpus callosum posterior and corpus callosum central was smaller in pMCI subjects. Partial correlation and general linear regression analysis showed that the left hippocampus was predicted region for memory, left thalamus was predicted region for language, executive and visual spatial. These current results suggest that the volumes of sub-cortical structures in stable MCI and progress MCI patients were heterogeneous. Among these regions, the left hippocampus was predicted region for memory, left thalamus was predicted region for language, executive and visual spatial, suggesting that these structures might be important for detecting the subtle effects of MCI patients' cognitive domain or to assess the effectiveness of therapeutic intervention for MCI.
Collapse
Affiliation(s)
- Lai Qian
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, Jiangsu, China; The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China; Nanjing Clinic Medicine Center for Neurological and Psychiatric Diseases, Nanjing, China
| | - Renyuan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China; Nanjing Clinic Medicine Center for Neurological and Psychiatric Diseases, Nanjing, China; Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, Jiangsu, China
| | - Ruomeng Qin
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, Jiangsu, China; The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China; Nanjing Clinic Medicine Center for Neurological and Psychiatric Diseases, Nanjing, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, Jiangsu, China; The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China; Nanjing Clinic Medicine Center for Neurological and Psychiatric Diseases, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing 210008, Jiangsu, China; The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing, China; Nanjing Clinic Medicine Center for Neurological and Psychiatric Diseases, Nanjing, China.
| |
Collapse
|
11
|
The anteroposterior and primary-to-posterior limbic ratios as MRI-derived volumetric markers of Alzheimer's disease. J Neurol Sci 2017; 378:110-119. [PMID: 28566144 DOI: 10.1016/j.jns.2017.04.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/17/2017] [Accepted: 04/26/2017] [Indexed: 11/21/2022]
Abstract
BACKGROUND/AIMS Alzheimer's disease (AD) shows a characteristic pattern of brain atrophy, with predominant involvement of posterior limbic structures, and relative preservation of rostral limbic and primary cortical regions. We aimed to investigate the diagnostic utility of two gray matter volume ratios based on this pattern, and to develop a fully automated method to calculate them from unprocessed MRI files. PATIENTS AND METHODS Cross-sectional study of 118 subjects from the ADNI database, including normal controls and patients with mild cognitive impairment (MCI) and AD. Clinical variables and 3T T1-weighted MRI files were analyzed. Regional gray matter and total intracranial volumes were calculated with a shell script (gm_extractor) based on FSL. Anteroposterior and primary-to-posterior limbic ratios (APL and PPL) were calculated from these values. Diagnostic utility of variables was tested in logistic regression models using Bayesian model averaging for variable selection. External validity was evaluated with bootstrap sampling and a test set of 60 subjects. RESULTS gm_extractor showed high test-retest reliability and high concurrent validity with FSL's FIRST. Volumetric measurements agreed with the expected anatomical pattern associated with AD. APL and PPL ratios were significantly different between groups, and were selected instead of hippocampal and entorhinal volumes to differentiate normal from MCI or cognitively impaired (MCI plus AD) subjects. CONCLUSION APL and PPL ratios may be useful components of models aimed to differentiate normal subjects from patients with MCI or AD. These values, and other gray matter volumes, may be reliably calculated with gm_extractor.
Collapse
|
12
|
Schreurs A, Sabanov V, Balschun D. Distinct Properties of Long-Term Potentiation in the Dentate Gyrus along the Dorsoventral Axis: Influence of Age and Inhibition. Sci Rep 2017; 7:5157. [PMID: 28698637 PMCID: PMC5506024 DOI: 10.1038/s41598-017-05358-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
The hippocampus is important for spatial navigation, episodic memory and affective behaviour. Increasing evidence suggests that these multiple functions are accomplished by different segments along the dorsal-ventral (septal-temporal) axis. Long-term potentiation (LTP), the best-investigated cellular correlate of learning and memory, has distinct properties along this axis in the CA1 region, but so far, little is known about longitudinal differences in dentate gyrus (DG). Therefore, here we examined potential dorsoventral differences in DG-LTP using in vitro multi-electrode array recordings. In young mice, we found higher basal synaptic transmission in the dorsal DG, while the LTP magnitude markedly increased towards the ventral pole. Strikingly, these differences were greatly reduced in slices from middle-aged mice. Short-term plasticity, evaluated by paired-pulse ratios, was similar across groups. Recordings in the presence and absence of GABAA-receptor blocker picrotoxin suggested a higher inhibitory tone in the ventral DG of young mice, confirmed by an increased frequency of miniature inhibitory postsynaptic currents. Our findings support the view that the hippocampus contains discrete functional domains along its dorsoventral axis and demonstrate that these are subject to age-dependent changes. Since these characteristics are presumably conserved in the human hippocampus, our findings have important clinical implications for hippocampus- and age-related disorders.
Collapse
Affiliation(s)
- An Schreurs
- KU Leuven, Faculty of Psychology and Educational Sciences, Brain & Cognition, Laboratory of Biological Psychology, Leuven, Belgium
| | - Victor Sabanov
- KU Leuven, Faculty of Psychology and Educational Sciences, Brain & Cognition, Laboratory of Biological Psychology, Leuven, Belgium
| | - Detlef Balschun
- KU Leuven, Faculty of Psychology and Educational Sciences, Brain & Cognition, Laboratory of Biological Psychology, Leuven, Belgium.
| |
Collapse
|
13
|
Kälin AM, Park MTM, Chakravarty MM, Lerch JP, Michels L, Schroeder C, Broicher SD, Kollias S, Nitsch RM, Gietl AF, Unschuld PG, Hock C, Leh SE. Subcortical Shape Changes, Hippocampal Atrophy and Cortical Thinning in Future Alzheimer's Disease Patients. Front Aging Neurosci 2017; 9:38. [PMID: 28326033 PMCID: PMC5339600 DOI: 10.3389/fnagi.2017.00038] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 02/13/2017] [Indexed: 11/13/2022] Open
Abstract
Efficacy of future treatments depends on biomarkers identifying patients with mild cognitive impairment at highest risk for transitioning to Alzheimer's disease. Here, we applied recently developed analysis techniques to investigate cross-sectional differences in subcortical shape and volume alterations in patients with stable mild cognitive impairment (MCI) (n = 23, age range 59–82, 47.8% female), future converters at baseline (n = 10, age range 66–84, 90% female) and at time of conversion (age range 68–87) compared to group-wise age and gender matched healthy control subjects (n = 23, age range 61–81, 47.8% female; n = 10, age range 66–82, 80% female; n = 10, age range 68–82, 70% female). Additionally, we studied cortical thinning and global and local measures of hippocampal atrophy as known key imaging markers for Alzheimer's disease. Apart from bilateral striatal volume reductions, no morphometric alterations were found in cognitively stable patients. In contrast, we identified shape alterations in striatal and thalamic regions in future converters at baseline and at time of conversion. These shape alterations were paralleled by Alzheimer's disease like patterns of left hemispheric morphometric changes (cortical thinning in medial temporal regions, hippocampal total and subfield atrophy) in future converters at baseline with progression to similar right hemispheric alterations at time of conversion. Additionally, receiver operating characteristic curve analysis indicated that subcortical shape alterations may outperform hippocampal volume in identifying future converters at baseline. These results further confirm the key role of early cortical thinning and hippocampal atrophy in the early detection of Alzheimer's disease. But first and foremost, and by distinguishing future converters but not patients with stable cognitive abilities from cognitively normal subjects, our results support the value of early subcortical shape alterations and reduced hippocampal subfield volumes as potential markers for the early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Andrea M Kälin
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Min T M Park
- Cerebral Imaging Centre, Douglas Mental Health University InstituteMontreal, QC, Canada; Schulich School of Medicine and Dentistry, Western UniversityLondon, ON, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University InstituteMontreal, QC, Canada; Departments of Psychiatry and Biological and Biomedical Engineering, McGill UniversityMontreal, QC, Canada
| | - Jason P Lerch
- The Hospital for Sick ChildrenToronto, ON, Canada; Department of Medical Biophysics, The University of TorontoToronto, ON, Canada
| | - Lars Michels
- Clinic of Neuroradiology, University Hospital Zurich, University of ZurichZurich, Switzerland; Center for MR Research, University Children's Hospital ZurichZurich, Switzerland
| | - Clemens Schroeder
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Sarah D Broicher
- Neuropsychology Unit, Department of Neurology, University Hospital Zurich Zurich, Switzerland
| | - Spyros Kollias
- Clinic of Neuroradiology, University Hospital Zurich, University of Zurich Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Anton F Gietl
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Paul G Unschuld
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| | - Sandra E Leh
- Institute for Regenerative Medicine, University of Zurich Schlieren, Switzerland
| |
Collapse
|
14
|
Leh SE, Kälin AM, Schroeder C, Park MTM, Chakravarty MM, Freund P, Gietl AF, Riese F, Kollias S, Hock C, Michels L. Volumetric and shape analysis of the thalamus and striatum in amnestic mild cognitive impairment. J Alzheimers Dis 2016; 49:237-49. [PMID: 26444755 DOI: 10.3233/jad-150080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alterations in brain structures, including progressive neurodegeneration, are a hallmark in patients with Alzheimer's disease (AD). However, pathological mechanisms, such as the accumulation of amyloid and the proliferation of tau, are thought to begin years, even decades, before the initial clinical manifestations of AD. In this study, we compare the brain anatomy of amnestic mild cognitive impairment patients (aMCI, n = 16) to healthy subjects (CS, n = 22) using cortical thickness, subcortical volume, and shape analysis, which we believe to be complimentary to volumetric measures. We were able to replicate "classical" cortical thickness alterations in aMCI in the hippocampus, amygdala, putamen, insula, and inferior temporal regions. Additionally, aMCI showed significant thalamic and striatal shape differences. We observed higher global amyloid deposition in aMCI, a significant correlation between striatal displacement and global amyloid, and an inverse correlation between executive function and right-hemispheric thalamic displacement. In contrast, no volumetric differences were detected in thalamic, striatal, and hippocampal regions. Our results provide new evidence for early subcortical neuroanatomical changes in patients with aMCI, which are linked to cognitive abilities and amyloid deposition. Hence, shape analysis may aid in the identification of structural biomarkers for identifying individuals at highest risk of conversion to AD.
Collapse
Affiliation(s)
- Sandra E Leh
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Andrea M Kälin
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Clemens Schroeder
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Min Tae M Park
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Canada.,Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Canada.,Departments of Psychiatry and Biomedical Engineering, McGill University, Montreal, Canada
| | - Patrick Freund
- Spinal Cord Injury Center, University Hospital Balgrist, Switzerland.,Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, University College London, London, UK.,Wellcome Trust Centre for Neuroimaging, UCL Institute of Neurology, University College London, London, UK.,Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Anton F Gietl
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Florian Riese
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Spyros Kollias
- Institute of Neuroradiology, University of Zurich, Switzerland
| | - Christoph Hock
- Division of Psychiatry Research and Psychogeriatric Medicine, University of Zurich, Switzerland
| | - Lars Michels
- Institute of Neuroradiology, University of Zurich, Switzerland
| |
Collapse
|
15
|
Bruno D, Grothe MJ, Nierenberg J, Zetterberg H, Blennow K, Teipel SJ, Pomara N. A study on the specificity of the association between hippocampal volume and delayed primacy performance in cognitively intact elderly individuals. Neuropsychologia 2015; 69:1-8. [PMID: 25613646 DOI: 10.1016/j.neuropsychologia.2015.01.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/15/2015] [Accepted: 01/17/2015] [Indexed: 11/19/2022]
Abstract
Delayed recall at the primacy position (first few items on a list) has been shown to predict cognitive decline in cognitively intact elderly participants, with poorer delayed primacy performance associated with more pronounced generalized cognitive decline during follow-up. We have previously suggested that this association is due to delayed primacy performance indexing memory consolidation, which in turn is thought to depend upon hippocampal function. Here, we test the hypothesis that hippocampal size is associated with delayed primacy performance in cognitively intact elderly individuals. Data were analyzed from a group (N=81) of cognitively intact participants, aged 60 or above. Serial position performance was measured with the Buschke selective reminding test (BSRT). Hippocampal size was automatically measured via MRI, and unbiased voxel-based analyses were also conducted to explore further regional specificity of memory performance. We conducted regression analyses of hippocampus volumes on serial position performance; other predictors included age, family history of Alzheimer's disease (AD), APOE ε4 status, education, and total intracranial volume. Our results collectively suggest that there is a preferential association between hippocampal volume and delayed primacy performance. These findings are consistent with the hypothesis that delayed primacy consolidation is associated with hippocampal size, and shed light on the relationship between delayed primacy performance and generalized cognitive decline in cognitively intact individuals, suggesting that delayed primacy consolidation may serve as a sensitive marker of hippocampal health in these individuals.
Collapse
Affiliation(s)
- Davide Bruno
- Department of Psychology, Liverpool Hope University, Hope Park, Liverpool L16 9JD, UK.
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany
| | - Jay Nierenberg
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Psychiatry, School of Medicine, New York University, New York City, NY, USA
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - Nunzio Pomara
- Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA; Department of Psychiatry, School of Medicine, New York University, New York City, NY, USA
| |
Collapse
|
16
|
O'brien JT. Commentary on: Amnestic mild cognitive impairment and incident dementia and Alzheimer's disease in geriatric depression. Int Psychogeriatr 2014; 26:2027-8. [PMID: 25382200 DOI: 10.1017/s1041610214002087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- John T O'brien
- Department of Psychiatry,University of Cambridge,United Kingdom
| |
Collapse
|
17
|
Abstract
There is increasing interest in finding markers of Alzheimer's disease (AD) that are discriminative even at an early, pre-dementia stage. This interest is driven partly by a desire to improve clinical diagnosis in more mildly affected individuals, and also by the recent paradigm shift in thinking about clinical trials for AD. This shift is a result of concern that the recent failures of high-profile clinical trials conducted in patients with mild to moderate AD may have been because therapy was “too little, too late.” The implication being that if only treatments had been trialled earlier they would have had a greater chance of success. Certainly, lessons from other aspects of medicine have shown that treatments may be most, or in some cases only, effective if given early in disease. If we did have therapies that could slow disease progression at a very early stage that would increase the interest in early markers of disease. Ideally, such therapies would be given when the minimum of functional decline and irreversible neuronal loss had already occurred. From economic and public health standpoints, delaying symptom onset would be very important: a delay of five years has been estimated to reduce projections for prevalence of symptomatic AD by about 50% (Brookmeyer et al. , 1998).
Collapse
Affiliation(s)
- Josephine Barnes
- Dementia Research Centre,Department of Neurodegenerative Disease,UCL Institute of Neurology,Queen Square,London WC1N 3BGUnited Kingdom
| | - Nick C Fox
- Dementia Research Centre,Department of Neurodegenerative Disease,UCL Institute of Neurology,Queen Square,London WC1N 3BGUnited Kingdom
| |
Collapse
|
18
|
Adler DA, Ammanuel S, Lei J, Dada T, Borbiev T, Johnston MV, Kadam SD, Burd I. Circadian cycle-dependent EEG biomarkers of pathogenicity in adult mice following prenatal exposure to in utero inflammation. Neuroscience 2014; 275:305-13. [PMID: 24954445 DOI: 10.1016/j.neuroscience.2014.06.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/29/2014] [Accepted: 06/11/2014] [Indexed: 11/19/2022]
Abstract
Intrauterine infection or inflammation in preterm neonates is a known risk for adverse neurological outcomes, including cognitive, motor and behavioral disabilities. Our previous data suggest that there is acute fetal brain inflammation in a mouse model of intrauterine exposure to lipopolysaccharides (LPS). We hypothesized that the in utero inflammation induced by LPS produces long-term electroencephalogram (EEG) biomarkers of neurodegeneration in the exposed mice that could be determined by using continuous quantitative video/EEG/electromyogram (EMG) analyses. A single LPS injection at E17 was performed in pregnant CD1 dams. Control dams were injected with same volumes of saline (LPS n=10, Control n=8). At postnatal age of P90-100, 24-h synchronous video/EEG/EMG recordings were done using a tethered recording system and implanted subdural electrodes. Behavioral state scoring was performed blind to treatment group, on each 10s EEG epoch using synchronous video, EMG and EEG trace signatures to generate individual hypnograms. Automated EEG power spectrums were analyzed for delta and theta-beta power ratios during wake vs. sleep cycles. Both control and LPS hypnograms showed an ultradian wake/sleep cycling. Since rodents are nocturnal animals, control mice showed the expected diurnal variation with significantly longer time spent in wake states during the dark cycle phase. In contrast, the LPS-treated mice lost this circadian rhythm. Sleep microstructure also showed significant alteration in the LPS mice specifically during the dark cycle, caused by significantly longer average non-rapid eye movement (NREM) cycle durations. No significance was found between treatment groups for the delta power data; however, significant activity-dependent changes in theta-beta power ratios seen in controls were absent in the LPS-exposed mice. In conclusion, exposure to in utero inflammation in CD1 mice resulted in significantly altered sleep architecture as adults that were circadian cycle and activity state dependent.
Collapse
Affiliation(s)
- D A Adler
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - S Ammanuel
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - J Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - T Dada
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - T Borbiev
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - M V Johnston
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - S D Kadam
- Department of Neuroscience, Hugo Moser Research Institute at Kennedy Krieger, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - I Burd
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA; Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|