101
|
Eid A, Mhatre I, Richardson JR. Gene-environment interactions in Alzheimer's disease: A potential path to precision medicine. Pharmacol Ther 2019; 199:173-187. [PMID: 30877021 DOI: 10.1016/j.pharmthera.2019.03.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the United States and afflicts >5.7 million Americans in 2018. Therapeutic options remain extremely limited to those that are symptom targeting, while no drugs have been approved for the modification or reversal of the disease itself. Risk factors for AD including aging, the female sex, as well as carrying an APOE4 genotype. These risk factors have been extensively examined in the literature, while less attention has been paid to modifiable risk factors, including lifestyle, and environmental risk factors such as exposures to air pollution and pesticides. This review highlights the most recent data on risk factors in AD and identifies gene by environment interactions that have been investigated. It also provides a suggested framework for a personalized therapeutic approach to AD, by combining genetic, environmental and lifestyle risk factors. Understanding modifiable risk factors and their interaction with non-modifiable factors (age, susceptibility alleles, and sex) is paramount for designing personalized therapeutic interventions.
Collapse
Affiliation(s)
- Aseel Eid
- Department of Environmental Health, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, United States of America
| | - Isha Mhatre
- Department of Environmental Health, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, United States of America; Department of Neurosciences, School of Biomedical Sciences, Kent State University, Kent, OH
| | - Jason R Richardson
- Department of Environmental Health, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, FL, United States of America.
| |
Collapse
|
102
|
Rezzani R, Franco C, Rodella LF. Sex differences of brain and their implications for personalized therapy. Pharmacol Res 2019; 141:429-442. [PMID: 30659897 DOI: 10.1016/j.phrs.2019.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/06/2023]
Abstract
Nowadays, it is known that the sex differences regard many organs, e.g., liver, vessels, pancreas, lungs, bronchi and also the brain. Sex differences are not just a matter of ethical and moral principles, as they are central to explain many still unknown diseases and their understanding is a prerequisite to develop an effective therapy for each individual. This review reports on those sex differences that are not only macroscopic and morphological, but also involve molecular and functional dimorphism in the brain. It will recapitulate the main structural differences between male and female brain including the neurotransmission systems; in particular, the main objective is to identify a correlation, already known or to be investigated in the future, between the differences that characterize male and female brains from a morphological and biochemical point of view and neurological syndromes. This correlation could provide a starting point for future scientific research aimed to investigate and define a personalized therapy.
Collapse
Affiliation(s)
- Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Caterina Franco
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
103
|
Abstract
Engaging in targeted exercise interventions is a promising, non-pharmacological strategy to mitigate the deleterious effects of aging and disease on brain health. However, despite its therapeutic potential, a large amount of variation exists in exercise efficacy in older adults aged 55 and older. In this review, we present the argument that biological sex may be an important moderator of the relationship between physical activity and cognition. Sex differences exist in dementia as well as in several associated risk factors, including genetics, cardiovascular factors, inflammation, hormones and social and psychological factors. Different exercise interventions, such as aerobic training and resistance training, influence cognition and brain health in older adults and these effects may be sex-dependent. The biological mechanisms underlying the beneficial effects of exercise on the brain may be different in males and females. Specifically, we examine sex differences in neuroplasticity, neurotrophic factors and physiological effects of exercise to highlight the possible mediators of sex differences in exercise efficacy on cognition. Future studies should address the potential sex difference in exercise efficacy if we are to develop effective, evidence-based exercise interventions to promote healthy brain aging for all individuals.
Collapse
Affiliation(s)
- Cindy K Barha
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.,Department of Physical Therapy, University of British Columbia, Vancouver, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.,Department of Physical Therapy, University of British Columbia, Vancouver, Canada.,Centre for Hip Health and Mobility, Vancouver, Canada
| |
Collapse
|
104
|
Deming Y, Dumitrescu L, Barnes LL, Thambisetty M, Kunkle B, Gifford KA, Bush WS, Chibnik LB, Mukherjee S, De Jager PL, Kukull W, Huentelman M, Crane PK, Resnick SM, Keene CD, Montine TJ, Schellenberg GD, Haines JL, Zetterberg H, Blennow K, Larson EB, Johnson SC, Albert M, Moghekar A, Del Aguila JL, Fernandez MV, Budde J, Hassenstab J, Fagan AM, Riemenschneider M, Petersen RC, Minthon L, Chao MJ, Van Deerlin VM, Lee VMY, Shaw LM, Trojanowski JQ, Peskind ER, Li G, Davis LK, Sealock JM, Cox NJ, Goate AM, Bennett DA, Schneider JA, Jefferson AL, Cruchaga C, Hohman TJ. Sex-specific genetic predictors of Alzheimer's disease biomarkers. Acta Neuropathol 2018; 136:857-872. [PMID: 29967939 PMCID: PMC6280657 DOI: 10.1007/s00401-018-1881-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
Abstract
Cerebrospinal fluid (CSF) levels of amyloid-β 42 (Aβ42) and tau have been evaluated as endophenotypes in Alzheimer's disease (AD) genetic studies. Although there are sex differences in AD risk, sex differences have not been evaluated in genetic studies of AD endophenotypes. We performed sex-stratified and sex interaction genetic analyses of CSF biomarkers to identify sex-specific associations. Data came from a previous genome-wide association study (GWAS) of CSF Aβ42 and tau (1527 males, 1509 females). We evaluated sex interactions at previous loci, performed sex-stratified GWAS to identify sex-specific associations, and evaluated sex interactions at sex-specific GWAS loci. We then evaluated sex-specific associations between prefrontal cortex (PFC) gene expression at relevant loci and autopsy measures of plaques and tangles using data from the Religious Orders Study and Rush Memory and Aging Project. In Aβ42, we observed sex interactions at one previous and one novel locus: rs316341 within SERPINB1 (p = 0.04) and rs13115400 near LINC00290 (p = 0.002). These loci showed stronger associations among females (β = - 0.03, p = 4.25 × 10-8; β = 0.03, p = 3.97 × 10-8) than males (β = - 0.02, p = 0.009; β = 0.01, p = 0.20). Higher levels of expression of SERPINB1, SERPINB6, and SERPINB9 in PFC was associated with higher levels of amyloidosis among females (corrected p values < 0.02) but not males (p > 0.38). In total tau, we observed a sex interaction at a previous locus, rs1393060 proximal to GMNC (p = 0.004), driven by a stronger association among females (β = 0.05, p = 4.57 × 10-10) compared to males (β = 0.02, p = 0.03). There was also a sex-specific association between rs1393060 and tangle density at autopsy (pfemale = 0.047; pmale = 0.96), and higher levels of expression of two genes within this locus were associated with lower tangle density among females (OSTN p = 0.006; CLDN16 p = 0.002) but not males (p ≥ 0.32). Results suggest a female-specific role for SERPINB1 in amyloidosis and for OSTN and CLDN16 in tau pathology. Sex-specific genetic analyses may improve understanding of AD's genetic architecture.
Collapse
Affiliation(s)
- Yuetiva Deming
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, 1207 17th Avenue S, Nashville, TN, 37212, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Madhav Thambisetty
- Unit of Clinical and Translational Neuroscience, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Brian Kunkle
- Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, 1207 17th Avenue S, Nashville, TN, 37212, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Lori B Chibnik
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Philip L De Jager
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, USA
- Cell Circuits Program, Broad Institute, Cambridge, MA, USA
| | - Walter Kukull
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Matt Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Susan M Resnick
- Unit of Clinical and Translational Neuroscience, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan L Haines
- Department of Population and Quantitative Health Sciences, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Eric B Larson
- Department of Medicine, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Sterling C Johnson
- Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center of the Wm. S. Middleton Memorial VA Hospital, Madison, WI, USA
| | - Marilyn Albert
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jorge L Del Aguila
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - Maria Victoria Fernandez
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - Jason Hassenstab
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - Anne M Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Michael J Chao
- Ronald M Loeb Center for Alzheimer's Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vivianna M Van Deerlin
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Peskind
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
| | - Gail Li
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Lea K Davis
- Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julia M Sealock
- Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy J Cox
- Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alison M Goate
- Ronald M Loeb Center for Alzheimer's Disease, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Angela L Jefferson
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, 1207 17th Avenue S, Nashville, TN, 37212, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660 S. Euclid Ave. B8134, St. Louis, MO, 63110, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, 1207 17th Avenue S, Nashville, TN, 37212, USA.
| |
Collapse
|
105
|
Sapkota S, Ramirez J, Stuss DT, Masellis M, Black SE. Clinical dementia severity associated with ventricular size is differentially moderated by cognitive reserve in men and women. ALZHEIMERS RESEARCH & THERAPY 2018; 10:89. [PMID: 30185213 PMCID: PMC6123907 DOI: 10.1186/s13195-018-0419-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/07/2018] [Indexed: 01/08/2023]
Abstract
Background Interindividual differences in cognitive reserve (CR) are associated with complex and dynamic clinical phenotypes observed in cognitive impairment and dementia. We tested whether (1) CR early in life (E-CR; measured by education and IQ), (2) CR later in life (L-CR; measured by occupation), and (3) CR panel (CR-P) with the additive effects of E-CR and L-CR, act as moderating factors between baseline ventricular size and clinical dementia severity at baseline and across 2 years. We further examined whether this moderation is differentially represented by sex. Methods We examined a longitudinal model using patients (N = 723; mean age = 70.8 ± 9.4 years; age range = 38–90 years; females = 374) from the Sunnybrook Dementia Study. The patients represented Alzheimer’s disease (n = 439), mild cognitive impairment (n = 77), vascular cognitive impairment (n = 52), Lewy body disease (n = 30), and frontotemporal dementia (n = 125). Statistical analyses included (1) latent growth modeling to determine how clinical dementia severity changes over 2 years (measured by performance on the Dementia Rating Scale), (2) confirmatory factor analysis to establish a baseline E-CR factor, and (3) path analysis to predict dementia severity. Baseline age (continuous) and Apolipoprotein E status (ɛ4−/ɛ4+) were included as covariates. Results The association between higher baseline ventricular size and dementia severity was moderated by (1) E-CR and L-CR and (2) CR-P. This association was differentially represented in men and women. Specifically, men in only the low CR-P had higher baseline clinical dementia severity with larger baseline ventricular size. However, women in the low CR-P showed the (1) highest baseline dementia severity and (2) fastest 2-year decline with larger baseline ventricular size. Conclusions Clinical dementia severity associated with ventricular size may be (1) selectively moderated by complex and additive CR networks and (2) differentially represented by sex. Trials registration ClinicalTrials.gov, NCT01800214. Registered on 27 February 2013. Electronic supplementary material The online version of this article (10.1186/s13195-018-0419-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, M6-192, Toronto, ON, M4N 3M5, Canada.
| | - Joel Ramirez
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, M6-192, Toronto, ON, M4N 3M5, Canada
| | - Donald T Stuss
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, M6-192, Toronto, ON, M4N 3M5, Canada.,Departments of Medicine, University of Toronto, 190 Elizabeth Street, R. Fraser Elliot Building, 3-805, Toronto, ON, M5G 2C4, Canada.,Department of Psychology, University of Toronto, 100 St. George Street, 4th Floor, Sidney Smith Hall, Toronto, ON, M5S 3G3, Canada.,Rotman Research Institute of Baycrest Centre, 3560 Bathurst Street, Toronto, ON, M6H 4A6, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, M6-192, Toronto, ON, M4N 3M5, Canada.,Department of Medicine (Neurology), University of Toronto, 190 Elizabeth Street, R. Fraser Elliot Building, 3-805, Toronto, ON, M5G 2C4, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, M6-192, Toronto, ON, M4N 3M5, Canada.,Department of Medicine (Neurology), University of Toronto, 190 Elizabeth Street, R. Fraser Elliot Building, 3-805, Toronto, ON, M5G 2C4, Canada
| |
Collapse
|
106
|
Nebel RA, Aggarwal NT, Barnes LL, Gallagher A, Goldstein JM, Kantarci K, Mallampalli MP, Mormino EC, Scott L, Yu WH, Maki PM, Mielke MM. Understanding the impact of sex and gender in Alzheimer's disease: A call to action. Alzheimers Dement 2018; 14:1171-1183. [PMID: 29907423 PMCID: PMC6400070 DOI: 10.1016/j.jalz.2018.04.008] [Citation(s) in RCA: 519] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Precision medicine methodologies and approaches have advanced our understanding of the clinical presentation, development, progression, and management of Alzheimer's disease (AD) dementia. However, sex and gender have not yet been adequately integrated into many of these approaches. METHODS The Society for Women's Health Research Interdisciplinary Network on AD, comprised of an expert panel of scientists and clinicians, reviewed ongoing and published research related to sex and gender differences in AD. RESULTS The current review is a result of this Network's efforts and aims to: (1) highlight the current state-of-the-science in the AD field on sex and gender differences; (2) address knowledge gaps in assessing sex and gender differences; and (3) discuss 12 priority areas that merit further research. DISCUSSION The exclusion of sex and gender has impeded faster advancement in the detection, treatment, and care of AD across the clinical spectrum. Greater attention to these differences will improve outcomes for both sexes.
Collapse
Affiliation(s)
- Rebecca A Nebel
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA.
| | - Neelum T Aggarwal
- Department of Neurological Sciences and the Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Department of Neurological Sciences and the Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Aimee Gallagher
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA
| | - Jill M Goldstein
- Department of Psychiatry, Harvard Medical School, and Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, and Massachusetts General Hospital, Boston, MA, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Monica P Mallampalli
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Laura Scott
- Cellular and Molecular Medicine Program, Johns Hopkins University, Baltimore, MD, USA
| | - Wai Haung Yu
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Pauline M Maki
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Michelle M Mielke
- Department of Epidemiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
107
|
Zhang T, Shaw ME, Walsh EI, Sachdev PS, Anstey KJ, Cherbuin N. Higher fasting plasma glucose is associated with smaller striatal volume and poorer fine motor skills in a longitudinal cohort. Psychiatry Res Neuroimaging 2018; 278:1-6. [PMID: 29935440 DOI: 10.1016/j.pscychresns.2018.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/05/2023]
Abstract
Previous studies have demonstrated associations between higher blood glucose and brain atrophy and functional deficits, however, little is known about the association between blood glucose, striatal volume and striatal function despite sensori-motor deficits being reported in diabetes. This study investigated the relationship between blood glucose levels, striatal volume and fine motor skills in a longitudinal cohort of cognitively healthy individuals living in the community with normal or impaired fasting glucose or type 2 diabetes. Participants were 271 cognitively healthy individuals (mean age 63 years at inclusion) with normal fasting glucose levels (<5.6 mmol/L) (n=173), impaired fasting glucose (5.6-6.9 mmol/L) (n=57), or with type 2 diabetes (≥7.0 mmol/L) (n=41). Fasting glucose, Purdue Pegboard scores as measurement of fine motor skills, and brain scans were collected at wave 1, 2 and 4, over a total follow-up of twelve years. Striatal volumes were measured using FreeSurfer after controlling for age, sex and intracranial volume. Results showed that type 2 diabetes was associated with smaller right putamen volume and lower Purdue Pegboard scores after controlling for age, sex and intracranial volume. These findings add to the evidence suggesting that higher blood glucose levels, especially type 2 diabetes, may impair brain structure and function.
Collapse
Affiliation(s)
- Tianqi Zhang
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Marnie E Shaw
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Erin I Walsh
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | | | - Kaarin J Anstey
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, Australian National University, Canberra, Australia.
| |
Collapse
|
108
|
Gao L, Chen J, Gu L, Shu H, Wang Z, Liu D, Yan Y, Zhang Z. Effects of Gender and Apolipoprotein E on Novelty MMN and P3a in Healthy Elderly and Amnestic Mild Cognitive Impairment. Front Aging Neurosci 2018; 10:256. [PMID: 30186155 PMCID: PMC6110901 DOI: 10.3389/fnagi.2018.00256] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 08/03/2018] [Indexed: 01/27/2023] Open
Abstract
Background: The apolipoprotein E epsilon4 (ApoE ε4) allele and female gender may be important risk factors for the development of Alzheimer’s disease and amnestic mild cognitive impairment (aMCI). Novelty mismatch negativity (MMN) represents the pre-attentive index of deviance detection and P3a represents the attention orienting response. Furthermore, MMN and P3a components have been reported to be potential markers in aMCI. Therefore, this study will investigate the effects of gender and ApoE on auditory novelty MMN and P3a and their relationship to neuropsychological performance in aMCI. Methods: Thirty nine aMCI subjects and 44 controls underwent neuropsychological assessment and ApoE genotyping. Novelty MMN and P3a components were investigated during an auditory novelty oddball task. Results: Firstly, novelty MMN latency was significantly shorter in aMCI than in healthy control (HC) group. Secondly, novelty MMN latency was negatively correlated with episodic memory in aMCI, but not in HC. Novelty P3a latency was negatively correlated with information processing speed in all subjects. For gender effect, novelty MMN latency was shorter in aMCI females than in HC females. Moreover, novelty P3a amplitudes were lower in males than in females in both aMCI and HC. For the effect of ApoE status, novelty MMN latency was shorter in aMCI ApoE ε4- than HC ApoE ε4-. Conclusion: aMCI presents altered pre-attentive processing indexed by novelty MMN components. Furthermore, there may be a compensatory mechanism on the impaired processing in aMCI. It further suggests that aMCI female and ApoE ε4- recruited the compensatory mechanism.
Collapse
Affiliation(s)
- Lijuan Gao
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jiu Chen
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Lihua Gu
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hao Shu
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zan Wang
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Duan Liu
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yanna Yan
- Department of Psychology, Xinxiang Medical University, Xinxiang, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Department of Psychology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
109
|
K Barha C, Liu-Ambrose T. Exercise and the Aging Brain: Considerations for Sex Differences. Brain Plast 2018. [DOI: 10.3233/bpl-1867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Cindy K Barha
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
| | - Teresa Liu-Ambrose
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
- Department of Physical Therapy, University of British Columbia, Vancouver, Canada
- Centre for Hip Health and Mobility, Vancouver, Canada
| |
Collapse
|
110
|
Precision medicine and drug development in Alzheimer's disease: the importance of sexual dimorphism and patient stratification. Front Neuroendocrinol 2018; 50:31-51. [PMID: 29902481 DOI: 10.1016/j.yfrne.2018.06.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (ND) are among the leading causes of disability and mortality. Considerable sex differences exist in the occurrence of the various manifestations leading to cognitive decline. Alzheimer's disease (AD) exhibits substantial sexual dimorphisms and disproportionately affects women. Women have a higher life expectancy compared to men and, consequently, have more lifespan to develop AD. The emerging precision medicine and pharmacology concepts - taking into account the individual genetic and biological variability relevant for disease risk, prevention, detection, diagnosis, and treatment - are expected to substantially enhance our knowledge and management of AD. Stratifying the affected individuals by sex and gender is an important basic step towards personalization of scientific research, drug development, and care. We hypothesize that sex and gender differences, extending from genetic to psychosocial domains, are highly relevant for the understanding of AD pathophysiology, and for the conceptualization of basic/translational research and for clinical therapy trial design.
Collapse
|
111
|
Congdon EE. Sex Differences in Autophagy Contribute to Female Vulnerability in Alzheimer's Disease. Front Neurosci 2018; 12:372. [PMID: 29988365 PMCID: PMC6023994 DOI: 10.3389/fnins.2018.00372] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with over 5. 4 million cases in the US alone (Alzheimer's Association, 2016). Clinically, AD is defined by the presence of plaques composed of Aβ and neurofibrillary pathology composed of the microtubule associated protein tau. Another key feature is the dysregulation of autophagy at key steps in the pathway. In AD, disrupted autophagy contributes to disease progression through the failure to clear pathological protein aggregates, insulin resistance, and its role in the synthesis of Aβ. Like many psychiatric and neurodegenerative diseases, the risk of developing AD, and disease course are dependent on the sex of the patient. One potential mechanism through which these differences occur, is the effects of sex hormones on autophagy. In women, the loss of hormones with menopause presents both a risk factor for developing AD, and an obvious example of where sex differences in AD can stem from. However, because AD pathology can begin decades before menopause, this does not provide the full answer. We propose that sex-based differences in autophagy regulation during the lifespan contribute to the increased risk of AD, and greater severity of pathology seen in women.
Collapse
Affiliation(s)
- Erin E Congdon
- Neuroscience and Physiology, School of Medicine, New York University, New York City, NY, United States
| |
Collapse
|
112
|
Loprinzi PD, Frith E. The Role of Sex in Memory Function: Considerations and Recommendations in the Context of Exercise. J Clin Med 2018; 7:jcm7060132. [PMID: 29857518 PMCID: PMC6028920 DOI: 10.3390/jcm7060132] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/31/2018] [Accepted: 05/31/2018] [Indexed: 01/16/2023] Open
Abstract
There is evidence to suggest that biological sex plays a critical role in memory function, with sex differentially influencing memory type. In this review, we detail the current evidence evaluating sex-specific effects on various memory types. We also discuss potential mechanisms that explain these sex-specific effects, which include sex differences in neuroanatomy, neurochemical differences, biological differences, and cognitive and affect-related differences. Central to this review, we also highlight that, despite the established sex differences in memory, there is little work directly comparing whether males and females have a differential exercise-induced effect on memory function. As discussed herein, such a differential effect is plausible given the clear sex-specific effects on memory, exercise response, and molecular mediators of memory. We emphasize that future work should be carefully powered to detect sex differences. Future research should also examine these potential exercise-related sex-specific effects for various memory types and exercise intensities and modalities. This will help enhance our understanding of whether sex indeed moderates the effects of exercise and memory function, and as such, will improve our understanding of whether sex-specific, memory-enhancing interventions should be developed, implemented, and evaluated.
Collapse
Affiliation(s)
- Paul D Loprinzi
- Exercise Psychology Laboratory, Physical Activity Epidemiology Laboratory, School of Applied Sciences, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, MS 38677, USA.
| | - Emily Frith
- Exercise Psychology Laboratory, Physical Activity Epidemiology Laboratory, School of Applied Sciences, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, MS 38677, USA.
| |
Collapse
|
113
|
Sohn D, Shpanskaya K, Lucas JE, Petrella JR, Saykin AJ, Tanzi RE, Samatova NF, Doraiswamy PM. Sex Differences in Cognitive Decline in Subjects with High Likelihood of Mild Cognitive Impairment due to Alzheimer's disease. Sci Rep 2018; 8:7490. [PMID: 29748598 PMCID: PMC5945611 DOI: 10.1038/s41598-018-25377-w] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 04/10/2018] [Indexed: 01/29/2023] Open
Abstract
Sex differences in Alzheimer’s disease (AD) biology and progression are not yet fully characterized. The goal of this study is to examine the effect of sex on cognitive progression in subjects with high likelihood of mild cognitive impairment (MCI) due to Alzheimer’s and followed up to 10 years in the Alzheimer’s Disease Neuroimaging Initiative (ADNI). Cerebrospinal fluid total-tau and amyloid-beta (Aβ42) ratio values were used to sub-classify 559 MCI subjects (216 females, 343 males) as having “high” or “low” likelihood for MCI due to Alzheimer’s. Data were analyzed using mixed-effects models incorporating all follow-ups. The worsening from baseline in Alzheimer’s Disease Assessment Scale-Cognitive score (mean, SD) (9 ± 12) in subjects with high likelihood of MCI due to Alzheimer’s was markedly greater than that in subjects with low likelihood (1 ± 6, p < 0.0001). Among MCI due to AD subjects, the mean worsening in cognitive score was significantly greater in females (11.58 ± 14) than in males (6.87 ± 11, p = 0.006). Our findings highlight the need to further investigate these findings in other populations and develop sex specific timelines for Alzheimer’s disease progression.
Collapse
Affiliation(s)
- Dongwha Sohn
- North Carolina State University, Department of Computer Science, Raleigh, NC, 27695, USA.,Oak Ridge National Laboratory, Computer Science and Mathematics Division, Oak Ridge, TN, 37831, USA
| | - Katie Shpanskaya
- Stanford University School of Medicine, Department of Radiology, Stanford, CA, 94025, USA
| | - Joseph E Lucas
- Duke University, Department of Statistical Science, Durham, NC, 27708, USA
| | - Jeffrey R Petrella
- Duke University Medical Center, Department of Radiology, Durham, NC, 27710, USA
| | - Andrew J Saykin
- Indiana University School of Medicine, Indiana Alzheimer Disease Center and the Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indianapolis, IN, 46202, USA
| | - Rudolph E Tanzi
- Massachusetts General Hospital and Harvard Medical School, Genetics and Aging Research Unit and Department of Neurology, Stanford, CA, 02129, USA
| | - Nagiza F Samatova
- North Carolina State University, Department of Computer Science, Raleigh, NC, 27695, USA.,Oak Ridge National Laboratory, Computer Science and Mathematics Division, Oak Ridge, TN, 37831, USA
| | - P Murali Doraiswamy
- Duke University Health System, Neurocognitive Disorders Program, Department of Psychiatry and the Duke Institute for Brain Sciences, Durham, NC, 27710, USA.
| |
Collapse
|
114
|
|
115
|
Fisher DW, Bennett DA, Dong H. Sexual dimorphism in predisposition to Alzheimer's disease. Neurobiol Aging 2018; 70:308-324. [PMID: 29754747 DOI: 10.1016/j.neurobiolaging.2018.04.004] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and rate of symptom progression, but the mechanisms underlying this sexual divergence are unknown. Although some have suggested this difference in risk is a reflection of the known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities toward the disease. Although a number of potential risk factors have been hypothesized to mediate these differences, none have been definitively verified. In this review, we first summarize the epidemiologic studies of prevalence and incidence of AD among the sexes. Next, we discuss the most likely risk factors to date that interact with biological sex, including (1) genetic factors, (2) sex hormones (3) deviations in brain structure, (4) inflammation and microglia, and (5) and psychosocial stress responses. Overall, though differences in life span are likely to account for part of the divide between the sexes in AD prevalence, the abundance of preclinical and clinical evidence presented here suggests an increase in intrinsic AD risk for women. Therefore, future studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD pathogenesis in both sexes, with the eventual goal of sex-specific prevention and treatment strategies.
Collapse
Affiliation(s)
- Daniel W Fisher
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Hongxin Dong
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
116
|
Longitudinal structural cerebral changes related to core CSF biomarkers in preclinical Alzheimer's disease: A study of two independent datasets. NEUROIMAGE-CLINICAL 2018; 19:190-201. [PMID: 30023169 PMCID: PMC6050455 DOI: 10.1016/j.nicl.2018.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/08/2018] [Accepted: 04/14/2018] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is characterized by an accumulation of β-amyloid (Aβ42) accompanied by brain atrophy and cognitive decline. Several recent studies have shown that Aβ42 accumulation is associated with gray matter (GM) changes prior to the development of cognitive impairment, in the so-called preclinical stage of the AD (pre-AD). It also has been proved that the GM atrophy profile is not linear, both in normal ageing but, especially, on AD. However, several other factors may influence this association and may have an impact on the generalization of results from different samples. In this work, we estimate differences in rates of GM volume change in cognitively healthy elders in association with baseline core cerebrospinal fluid (CSF) AD biomarkers, and assess to what these differences are sample dependent. We report the dependence of atrophy rates, measured in a two-year interval, on Aβ42, computed both over continuous and categorical values of Aβ42, at voxel-level (p < 0.001; k < 100) and corrected for sex, age and education. Analyses were performed jointly and separately, on two samples. The first sample was formed of 31 individuals (22 Ctrl and 9 pre-AD), aged 60–80 and recruited at the Hospital Clinic of Barcelona. The second sample was a replica of the first one with subjects selected from the ADNI dataset. We also investigated the dependence of the GM atrophy rate on the basal levels of continuous p-tau and on the p-tau/Aβ42 ratio. Correlation analyses on the whole sample showed a dependence of GM atrophy rates on Aβ42 in medial and orbital frontal, precuneus, cingulate, medial temporal regions and cerebellum. Correlations with p-tau were located in the left hippocampus, parahippocampus and striatal nuclei whereas correlation with p-tau/Aβ42 was mainly found in ventral and medial temporal areas. Regarding analyses performed separately, we found a substantial discrepancy of results between samples, illustrating the complexities of comparing two independent datasets even when using the same inclusion criteria. Such discrepancies may lead to significant differences in the sample size needed to detect a particular reduction on cerebral atrophy rates in prevention trials. Higher cognitive reserve and more advanced pathological progression in the ADNI sample could partially account for the observed discrepancies. Taken together, our findings in these two samples highlight the importance of comparing and merging independent datasets to draw more robust and generalizable conclusions on the structural changes in the preclinical stages of AD. GM atrophy rates depends differently on values of CSF Aβ42 than on CSF p-tau in the preclinical stage of AD. Discrepant results were obtained. Although nominally equivalent, samples might reflect different time-windows in the AD continuum. It is necessary a further effort to standardize CSF-biomarkers measures and thresholds to make different samples to be directly comparable.
Collapse
Key Words
- AD, Alzheimer's disease
- ADNI, Alzheimer's Disease Neuroimaging Initiative
- Alzheimer's disease
- Aβ42, amyloid beta
- CDR, Clinical Dementia Rating
- CSF biomarkers
- CSF, Cerebro-Spinal Fluid
- Ctrl, control
- DI, divergences of the longitudinal deformations
- ELISA, Enzyme-Linked ImmunoSorbent Assay
- FWE, Family Wise Error
- GM, gray matter
- HCB, Hospital Clinic Barcelona
- L, left
- Longitudinal VBM
- MMSE, Mini Mental State examination
- PLR, pairwise longitudinal registration
- Preclinical Alzheimer's disease
- R, right
- ROI, region of interest
- TIV, total intracranial volume
- VBM, voxel-based morphometry
- WM, white matter
- p-tau, phosphorylated tau
- preAD, preclinical Alzheimer's disease
- t-tau, total tau
Collapse
|
117
|
Lawrence E, Vegvari C, Ower A, Hadjichrysanthou C, De Wolf F, Anderson RM. A Systematic Review of Longitudinal Studies Which Measure Alzheimer's Disease Biomarkers. J Alzheimers Dis 2018; 59:1359-1379. [PMID: 28759968 PMCID: PMC5611893 DOI: 10.3233/jad-170261] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Alzheimer’s disease (AD) is a progressive and fatal neurodegenerative disease, with no effective treatment or cure. A gold standard therapy would be treatment to slow or halt disease progression; however, knowledge of causation in the early stages of AD is very limited. In order to determine effective endpoints for possible therapies, a number of quantitative surrogate markers of disease progression have been suggested, including biochemical and imaging biomarkers. The dynamics of these various surrogate markers over time, particularly in relation to disease development, are, however, not well characterized. We reviewed the literature for studies that measured cerebrospinal fluid or plasma amyloid-β and tau, or took magnetic resonance image or fluorodeoxyglucose/Pittsburgh compound B-positron electron tomography scans, in longitudinal cohort studies. We summarized the properties of the major cohort studies in various countries, commonly used diagnosis methods and study designs. We have concluded that additional studies with repeat measures over time in a representative population cohort are needed to address the gap in knowledge of AD progression. Based on our analysis, we suggest directions in which research could move in order to advance our understanding of this complex disease, including repeat biomarker measurements, standardization and increased sample sizes.
Collapse
Affiliation(s)
- Emma Lawrence
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Carolin Vegvari
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Alison Ower
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | | | - Frank De Wolf
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK.,Janssen Prevention Center, Leiden, The Netherlands
| | - Roy M Anderson
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
118
|
Fiford CM, Ridgway GR, Cash DM, Modat M, Nicholas J, Manning EN, Malone IB, Biessels GJ, Ourselin S, Carmichael OT, Cardoso MJ, Barnes J. Patterns of progressive atrophy vary with age in Alzheimer's disease patients. Neurobiol Aging 2018; 63:22-32. [PMID: 29220823 PMCID: PMC5805840 DOI: 10.1016/j.neurobiolaging.2017.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/14/2017] [Accepted: 11/06/2017] [Indexed: 01/18/2023]
Abstract
Age is not only the greatest risk factor for Alzheimer's disease (AD) but also a key modifier of disease presentation and progression. Here, we investigate how longitudinal atrophy patterns vary with age in mild cognitive impairment (MCI) and AD. Data comprised serial longitudinal 1.5-T magnetic resonance imaging scans from 153 AD, 339 MCI, and 191 control subjects. Voxel-wise maps of longitudinal volume change were obtained and aligned across subjects. Local volume change was then modeled in terms of diagnostic group and an interaction between group and age, adjusted for total intracranial volume, white-matter hyperintensity volume, and apolipoprotein E genotype. Results were significant at p < 0.05 with family-wise error correction for multiple comparisons. An age-by-group interaction revealed that younger AD patients had significantly faster atrophy rates in the bilateral precuneus, parietal, and superior temporal lobes. These results suggest younger AD patients have predominantly posterior progressive atrophy, unexplained by white-matter hyperintensity, apolipoprotein E, or total intracranial volume. Clinical trials may benefit from adapting outcome measures for patient groups with lower average ages, to capture progressive atrophy in posterior cortices.
Collapse
Affiliation(s)
- Cassidy M Fiford
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.
| | - Gerard R Ridgway
- FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Wellcome Trust Centre for Neuroimaging, 12 Queen Square, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Marc Modat
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - Emily N Manning
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Ian B Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sebastien Ourselin
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - M Jorge Cardoso
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Josephine Barnes
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| |
Collapse
|
119
|
Ardekani BA, Bermudez E, Mubeen AM, Bachman AH. Prediction of Incipient Alzheimer's Disease Dementia in Patients with Mild Cognitive Impairment. J Alzheimers Dis 2018; 55:269-281. [PMID: 27662309 DOI: 10.3233/jad-160594] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a transitional stage from normal aging to Alzheimer's disease (AD) dementia. It is extremely important to develop criteria that can be used to separate the MCI subjects at imminent risk of conversion to Alzheimer-type dementia from those who would remain stable. We have developed an automatic algorithm for computing a novel measure of hippocampal volumetric integrity (HVI) from structural MRI scans that may be useful for this purpose. OBJECTIVE To determine the utility of HVI in classification between stable and progressive MCI patients using the Random Forest classification algorithm. METHODS We used a 16-dimensional feature space including bilateral HVI obtained from baseline and one-year follow-up structural MRI, cognitive tests, and genetic and demographic information to train a Random Forest classifier in a sample of 164 MCI subjects categorized into two groups [progressive (n = 86) or stable (n = 78)] based on future conversion (or lack thereof) of their diagnosis to probable AD. RESULTS The overall accuracy of classification was estimated to be 82.3% (86.0% sensitivity, 78.2% specificity). The accuracy in women (89.1%) was considerably higher than that in men (78.9%). The prediction accuracy achieved in women is the highest reported in any previous application of machine learning to AD diagnosis in MCI. CONCLUSION The method presented in this paper can be used to separate stable MCI patients from those who are at early stages of AD dementia with high accuracy. There may be stronger indicators of imminent AD dementia in women with MCI as compared to men.
Collapse
Affiliation(s)
- Babak A Ardekani
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Elaine Bermudez
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Asim M Mubeen
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Alvin H Bachman
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | | |
Collapse
|
120
|
Koran MEI, Wagener M, Hohman TJ. Sex differences in the association between AD biomarkers and cognitive decline. Brain Imaging Behav 2018; 11:205-213. [PMID: 26843008 DOI: 10.1007/s11682-016-9523-8] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Women are disproportionately affected by Alzheimer's disease (AD) in terms of both disease prevalence and severity. Previous autopsy work has suggested that, in the presence of AD neuropathology, females are more susceptible to the clinical manifestation of AD. This manuscript extends that work by evaluating whether sex alters the established associations between cerebrospinal fluid (CSF) biomarker levels and brain aging outcomes (hippocampal volume, cognition). Participants were drawn from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and included individuals with normal cognition (n = 348), mild cognitive impairment (n = 565), and AD (n = 185). We leveraged mixed effects regression models to assess the interaction between sex and baseline cerebrospinal fluid biomarker levels of amyloid-β42 (Aβ-42) and total tau on cross-sectional and longitudinal brain aging outcomes. We found a significant interaction between sex and Aβ-42 on longitudinal hippocampal atrophy (p = 0.002), and longitudinal decline in memory (p = 0.017) and executive function (p = 0.025). Similarly, we observed an interaction between sex and total tau level on longitudinal hippocampal atrophy (p = 0.008), and longitudinal decline in executive function (p = 0.034). Women with Aβ-42 and total tau levels indicative of worse pathological changes showed more rapid hippocampal atrophy and cognitive decline. The sex difference was particularly pronounced among individuals with MCI, with lower education, and varied by APOE ε4 allele. These results suggest females may be more susceptible to the clinical manifestation of AD.
Collapse
Affiliation(s)
- Mary Ellen I Koran
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University School of Medicine, 1207 17th Ave S, Suite 204F, Nashville, TN, 37212, USA
| | - Madison Wagener
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University School of Medicine, 1207 17th Ave S, Suite 204F, Nashville, TN, 37212, USA
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University School of Medicine, 1207 17th Ave S, Suite 204F, Nashville, TN, 37212, USA.
| |
Collapse
|
121
|
Al-Janabi OM, Panuganti P, Abner EL, Bahrani AA, Murphy R, Bardach SH, Caban-Holt A, Nelson PT, Gold BT, Smith CD, Wilcock DM, Jicha GA. Global Cerebral Atrophy Detected by Routine Imaging: Relationship with Age, Hippocampal Atrophy, and White Matter Hyperintensities. J Neuroimaging 2018; 28:301-306. [PMID: 29314393 DOI: 10.1111/jon.12494] [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] [Received: 10/12/2017] [Revised: 11/29/2017] [Accepted: 12/10/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Interpreting the clinical significance of moderate-to-severe global cerebral atrophy (GCA) is a conundrum for many clinicians, who visually interpret brain imaging studies in routine clinical practice. GCA may be attributed to normal aging, Alzheimer's disease (AD), or cerebrovascular disease (CVD). Understanding the relationships of GCA with aging, AD, and CVD is important for accurate diagnosis and treatment decisions for cognitive complaints. METHODS To elucidate the relative associations of age, moderate-to-severe white matter hyperintensities (WMHs), and moderate-to-severe medial temporal lobe atrophy (MTA), with moderate-to-severe GCA, we visually rated clinical brain imaging studies of 325 participants from a community based sample. Logistic regression analysis was conducted to assess the relations of GCA with age, WMH, and MTA. RESULTS The mean age was 76.2 (±9.6) years, 40.6% were male, and the mean educational attainment was 15.1 (±3.7) years. Logistic regression results demonstrated that while a 1-year increase in age was associated with GCA (OR = 1.04; P = .04), MTA (OR = 3.7; P < .001), and WMH (OR = 8.80; P < .001) were strongly associated with GCA in our study population. Partial correlation analysis showed that the variance of GCA explained by age is less than the variance attributed to MTA and WMH (r = .13, .21, and .43, respectively). CONCLUSIONS Moderate-to-severe GCA is most likely to occur in the presence of AD or CVD and should not be solely attributed to age when evaluating clinical imaging findings in the workup of cognitive complaints. Developing optimal diagnostic and treatment strategies for cognitive decline in the setting of GCA requires an understanding of its risk factors in the aging population.
Collapse
Affiliation(s)
- Omar M Al-Janabi
- Sanders-Brown Center on Aging, Lexington, KY.,Departments of Behavioral Science, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Pradeep Panuganti
- Neurology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Erin L Abner
- Sanders-Brown Center on Aging, Lexington, KY.,Epidemiology and Biostatistics, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Ahmed A Bahrani
- Sanders-Brown Center on Aging, Lexington, KY.,Biomedical Engineering, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Ronan Murphy
- Sanders-Brown Center on Aging, Lexington, KY.,Neurology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Shoshana H Bardach
- Sanders-Brown Center on Aging, Lexington, KY.,Gerontology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Allison Caban-Holt
- Sanders-Brown Center on Aging, Lexington, KY.,Departments of Behavioral Science, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Lexington, KY.,Pathology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Brian T Gold
- Sanders-Brown Center on Aging, Lexington, KY.,Neuroscience, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Charles D Smith
- Sanders-Brown Center on Aging, Lexington, KY.,Neurology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Lexington, KY.,Physiology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Lexington, KY.,Departments of Behavioral Science, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY.,Neurology, University of Kentucky Colleges of Medicine, Public Health and Engineering, Lexington, KY
| |
Collapse
|
122
|
Hampel H, Toschi N, Babiloni C, Baldacci F, Black KL, Bokde AL, Bun RS, Cacciola F, Cavedo E, Chiesa PA, Colliot O, Coman CM, Dubois B, Duggento A, Durrleman S, Ferretti MT, George N, Genthon R, Habert MO, Herholz K, Koronyo Y, Koronyo-Hamaoui M, Lamari F, Langevin T, Lehéricy S, Lorenceau J, Neri C, Nisticò R, Nyasse-Messene F, Ritchie C, Rossi S, Santarnecchi E, Sporns O, Verdooner SR, Vergallo A, Villain N, Younesi E, Garaci F, Lista S. Revolution of Alzheimer Precision Neurology. Passageway of Systems Biology and Neurophysiology. J Alzheimers Dis 2018; 64:S47-S105. [PMID: 29562524 PMCID: PMC6008221 DOI: 10.3233/jad-179932] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Precision Neurology development process implements systems theory with system biology and neurophysiology in a parallel, bidirectional research path: a combined hypothesis-driven investigation of systems dysfunction within distinct molecular, cellular, and large-scale neural network systems in both animal models as well as through tests for the usefulness of these candidate dynamic systems biomarkers in different diseases and subgroups at different stages of pathophysiological progression. This translational research path is paralleled by an "omics"-based, hypothesis-free, exploratory research pathway, which will collect multimodal data from progressing asymptomatic, preclinical, and clinical neurodegenerative disease (ND) populations, within the wide continuous biological and clinical spectrum of ND, applying high-throughput and high-content technologies combined with powerful computational and statistical modeling tools, aimed at identifying novel dysfunctional systems and predictive marker signatures associated with ND. The goals are to identify common biological denominators or differentiating classifiers across the continuum of ND during detectable stages of pathophysiological progression, characterize systems-based intermediate endophenotypes, validate multi-modal novel diagnostic systems biomarkers, and advance clinical intervention trial designs by utilizing systems-based intermediate endophenotypes and candidate surrogate markers. Achieving these goals is key to the ultimate development of early and effective individualized treatment of ND, such as Alzheimer's disease. The Alzheimer Precision Medicine Initiative (APMI) and cohort program (APMI-CP), as well as the Paris based core of the Sorbonne University Clinical Research Group "Alzheimer Precision Medicine" (GRC-APM) were recently launched to facilitate the passageway from conventional clinical diagnostic and drug development toward breakthrough innovation based on the investigation of the comprehensive biological nature of aging individuals. The APMI movement is gaining momentum to systematically apply both systems neurophysiology and systems biology in exploratory translational neuroscience research on ND.
Collapse
Affiliation(s)
- Harald Hampel
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Department of Radiology, “Athinoula A. Martinos” Center for Biomedical Imaging, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Claudio Babiloni
- Department of Physiology and Pharmacology “Vittorio Erspamer”, University of Rome “La Sapienza”, Rome, Italy
- Institute for Research and Medical Care, IRCCS “San Raffaele Pisana”, Rome, Italy
| | - Filippo Baldacci
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arun L.W. Bokde
- Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - René S. Bun
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Francesco Cacciola
- Unit of Neurosurgery, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Enrica Cavedo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
- IRCCS “San Giovanni di Dio-Fatebenefratelli”, Brescia, Italy
| | - Patrizia A. Chiesa
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Olivier Colliot
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France; Department of Neuroradiology, AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France; Department of Neurology, AP-HP, Hôpital de la Pitié-Salpêtrière, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Paris, France
| | - Cristina-Maria Coman
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Bruno Dubois
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Andrea Duggento
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Stanley Durrleman
- Inserm, U1127, Paris, France; CNRS, UMR 7225 ICM, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, Paris, France; Institut du Cerveau et de la Moelle Épinière (ICM) Paris, France; Inria, Aramis project-team, Centre de Recherche de Paris, France
| | - Maria-Teresa Ferretti
- IREM, Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
- ZNZ Neuroscience Center Zurich, Zürich, Switzerland
| | - Nathalie George
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle Épinière, ICM, Ecole Normale Supérieure, ENS, Centre MEG-EEG, F-75013, Paris, France
| | - Remy Genthon
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Marie-Odile Habert
- Département de Médecine Nucléaire, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
- Laboratoire d’Imagerie Biomédicale, Sorbonne Universités, UPMC Univ Paris 06, Inserm U 1146, CNRS UMR 7371, Paris, France
| | - Karl Herholz
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, UK
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Foudil Lamari
- AP-HP, UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Stéphane Lehéricy
- Centre de NeuroImagerie de Recherche - CENIR, Institut du Cerveau et de la Moelle Épinière - ICM, F-75013, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM, F-75013, Paris, France
| | - Jean Lorenceau
- Institut de la Vision, INSERM, Sorbonne Universités, UPMC Univ Paris 06, UMR_S968, CNRS UMR7210, Paris, France
| | - Christian Neri
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, CNRS UMR 8256, Institut de Biologie Paris-Seine (IBPS), Place Jussieu, F-75005, Paris, France
| | - Robert Nisticò
- Department of Biology, University of Rome “Tor Vergata” & Pharmacology of Synaptic Disease Lab, European Brain Research Institute (E.B.R.I.), Rome, Italy
| | - Francis Nyasse-Messene
- Sorbonne Université, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
| | - Craig Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Simone Rossi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Department of Medicine, Surgery and Neurosciences, Section of Human Physiology University of Siena, Siena, Italy
| | - Emiliano Santarnecchi
- Department of Medicine, Surgery and Neurosciences, Unit of Neurology and Clinical Neurophysiology, Brain Investigation & Neuromodulation Lab. (Si-BIN Lab.), University of Siena, Siena, Italy
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Olaf Sporns
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
- IU Network Science Institute, Indiana University, Bloomington, IN, USA
| | | | - Andrea Vergallo
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | - Nicolas Villain
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| | | | - Francesco Garaci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
- Casa di Cura “San Raffaele Cassino”, Cassino, Italy
| | - Simone Lista
- AXA Research Fund & Sorbonne Université Chair, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l’hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l’hôpital, F-75013, Paris, France
| |
Collapse
|
123
|
Nordin K, Persson J, Stening E, Herlitz A, Larsson EM, Söderlund H. Structural whole-brain covariance of the anterior and posterior hippocampus: Associations with age and memory. Hippocampus 2017; 28:151-163. [DOI: 10.1002/hipo.22817] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 11/16/2017] [Accepted: 11/22/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Kristin Nordin
- Department of Psychology; Uppsala University; Uppsala Sweden
| | - Jonas Persson
- Department of Psychology; Uppsala University; Uppsala Sweden
| | - Eva Stening
- Department of Psychology; Uppsala University; Uppsala Sweden
| | - Agneta Herlitz
- Department of Clinical Neuroscience; Karolinska Institutet; Stockholm Sweden
| | - Elna-Marie Larsson
- Department of Surgical Sciences, Radiology; Uppsala University; Uppsala Sweden
| | | |
Collapse
|
124
|
Jahanshad N, Thompson PM. Multimodal neuroimaging of male and female brain structure in health and disease across the life span. J Neurosci Res 2017; 95:371-379. [PMID: 27870421 PMCID: PMC5119539 DOI: 10.1002/jnr.23919] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/13/2016] [Accepted: 08/22/2016] [Indexed: 12/27/2022]
Abstract
Sex differences in brain development and aging are important to identify, as they may help to understand risk factors and outcomes in brain disorders that are more prevalent in one sex compared with the other. Brain imaging techniques have advanced rapidly in recent years, yielding detailed structural and functional maps of the living brain. Even so, studies are often limited in sample size, and inconsistent findings emerge, one example being varying findings regarding sex differences in the size of the corpus callosum. More recently, large‐scale neuroimaging consortia such as the Enhancing Neuro Imaging Genetics through Meta Analysis Consortium have formed, pooling together expertise, data, and resources from hundreds of institutions around the world to ensure adequate power and reproducibility. These initiatives are helping us to better understand how brain structure is affected by development, disease, and potential modulators of these effects, including sex. This review highlights some established and disputed sex differences in brain structure across the life span, as well as pitfalls related to interpreting sex differences in health and disease. We also describe sex‐related findings from the ENIGMA consortium, and ongoing efforts to better understand sex differences in brain circuitry. © 2016 The Authors. Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California
| |
Collapse
|
125
|
McKenzie AT, Moyon S, Wang M, Katsyv I, Song WM, Zhou X, Dammer EB, Duong DM, Aaker J, Zhao Y, Beckmann N, Wang P, Zhu J, Lah JJ, Seyfried NT, Levey AI, Katsel P, Haroutunian V, Schadt EE, Popko B, Casaccia P, Zhang B. Multiscale network modeling of oligodendrocytes reveals molecular components of myelin dysregulation in Alzheimer's disease. Mol Neurodegener 2017; 12:82. [PMID: 29110684 PMCID: PMC5674813 DOI: 10.1186/s13024-017-0219-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/17/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Oligodendrocytes (OLs) and myelin are critical for normal brain function and have been implicated in neurodegeneration. Several lines of evidence including neuroimaging and neuropathological data suggest that Alzheimer's disease (AD) may be associated with dysmyelination and a breakdown of OL-axon communication. METHODS In order to understand this phenomenon on a molecular level, we systematically interrogated OL-enriched gene networks constructed from large-scale genomic, transcriptomic and proteomic data obtained from human AD postmortem brain samples. We then validated these networks using gene expression datasets generated from mice with ablation of major gene expression nodes identified in our AD-dysregulated networks. RESULTS The robust OL gene coexpression networks that we identified were highly enriched for genes associated with AD risk variants, such as BIN1 and demonstrated strong dysregulation in AD. We further corroborated the structure of the corresponding gene causal networks using datasets generated from the brain of mice with ablation of key network drivers, such as UGT8, CNP and PLP1, which were identified from human AD brain data. Further, we found that mice with genetic ablations of Cnp mimicked aspects of myelin and mitochondrial gene expression dysregulation seen in brain samples from patients with AD, including decreased protein expression of BIN1 and GOT2. CONCLUSIONS This study provides a molecular blueprint of the dysregulation of gene expression networks of OL in AD and identifies key OL- and myelination-related genes and networks that are highly associated with AD.
Collapse
Affiliation(s)
- Andrew T. McKenzie
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Sarah Moyon
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Neuroscience Initiative, The City University of New York, Advanced Science Research Center, 85 St. Nicholas Terrace, New York, NY 10031 USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Igor Katsyv
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Eric B. Dammer
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Duc M. Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322 USA
- Integrated Proteomics Core Facility, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Joshua Aaker
- Department of Neurology, The University of Chicago Pritzker School of Medicine, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Yongzhong Zhao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Noam Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - James J. Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322 USA
- Integrated Proteomics Core Facility, Emory University School of Medicine, Atlanta, GA 30322 USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Pavel Katsel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Vahram Haroutunian
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Mental Illness Research, Education, and Clinical Center (VISN 3), James J. Peters VA Medical Center, Bronx, NY 10468 USA
| | - Eric E. Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Brian Popko
- Department of Neurology, The University of Chicago Pritzker School of Medicine, 5841 S. Maryland Avenue, Chicago, IL 60637 USA
| | - Patrizia Casaccia
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
- Neuroscience Initiative, The City University of New York, Advanced Science Research Center, 85 St. Nicholas Terrace, New York, NY 10031 USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| |
Collapse
|
126
|
Gamberger D, Lavrač N, Srivatsa S, Tanzi RE, Doraiswamy PM. Identification of clusters of rapid and slow decliners among subjects at risk for Alzheimer's disease. Sci Rep 2017; 7:6763. [PMID: 28755001 PMCID: PMC5533731 DOI: 10.1038/s41598-017-06624-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/16/2017] [Indexed: 12/28/2022] Open
Abstract
The heterogeneity of Alzheimer's disease contributes to the high failure rate of prior clinical trials. We analyzed 5-year longitudinal outcomes and biomarker data from 562 subjects with mild cognitive impairment (MCI) from two national studies (ADNI) using a novel multilayer clustering algorithm. The algorithm identified homogenous clusters of MCI subjects with markedly different prognostic cognitive trajectories. A cluster of 240 rapid decliners had 2-fold greater atrophy and progressed to dementia at almost 5 times the rate of a cluster of 184 slow decliners. A classifier for identifying rapid decliners in one study showed high sensitivity and specificity in the second study. Characterizing subgroups of at risk subjects, with diverse prognostic outcomes, may provide novel mechanistic insights and facilitate clinical trials of drugs to delay the onset of AD.
Collapse
Affiliation(s)
| | | | - Shantanu Srivatsa
- Duke Institute for Brain Sciences, Duke University Health System, Durham, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit and Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - P Murali Doraiswamy
- Duke Institute for Brain Sciences, Duke University Health System, Durham, USA.
- Neurocognitive Disorders Program, Division of Translational Neuroscience, Department of Psychiatry, Duke University Health System, Durham, USA.
| |
Collapse
|
127
|
Cavedo E, Lista S, Rojkova K, Chiesa PA, Houot M, Brueggen K, Blautzik J, Bokde ALW, Dubois B, Barkhof F, Pouwels PJW, Teipel S, Hampel H. Disrupted white matter structural networks in healthy older adult APOE ε4 carriers - An international multicenter DTI study. Neuroscience 2017; 357:119-133. [PMID: 28596117 DOI: 10.1016/j.neuroscience.2017.05.048] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 05/24/2017] [Accepted: 05/29/2017] [Indexed: 12/20/2022]
Abstract
The ε4 allelic variant of the Apolipoprotein E gene (APOE ε4) is the best-established genetic risk factor for late-onset Alzheimer's disease (AD). White matter (WM) microstructural damages measured with Diffusion Tensor Imaging (DTI) represent an early sign of fiber tract disconnection in AD. We examined the impact of APOE ε4 on WM microstructure in elderly individuals from the multicenter European DTI Study on Dementia. Voxelwise statistical analysis of fractional anisotropy (FA), mean diffusivity, radial and axial diffusivity (MD, radD and axD respectively) was carried out using Tract-Based Spatial Statistics. Seventy-four healthy elderly individuals - 31 APOE ε4 carriers (APOE ε4+) and 43 APOE ε4 non-carriers (APOE ε4-) -were considered for data analysis. All the results were corrected for scanner acquisition protocols, age, gender and for multiple comparisons. APOE ε4+ and APOE ε4- subjects were comparable regarding sociodemographic features and global cognition. A significant reduction of FA and increased radD was found in the APOE ε4+ compared to the APOE ε4- in the cingulum, in the corpus callosum, in the inferior fronto-occipital and in the inferior longitudinal fasciculi, internal and external capsule. APOE ε4+, compared to APOE ε4- showed higher MD in the genu, right internal capsule, superior longitudinal fasciculus and corona radiate. Comparisons stratified by center supported the results obtained on the whole sample. These findings support previous evidence in monocentric studies indicating a modulatory role of APOE ɛ4 allele on WM microstructure in elderly individuals at risk for AD suggesting early vulnerability and/or reduced resilience of WM tracts involved in AD.
Collapse
Affiliation(s)
- Enrica Cavedo
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013 Paris, France; Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | - Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013 Paris, France
| | - Katrine Rojkova
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013 Paris, France
| | - Patrizia A Chiesa
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013 Paris, France
| | - Marion Houot
- Institute of Memory and Alzheimer's Disease (IM2A), Centre of Excellence of Neurodegenerative Disease (CoEN), ICM, APHP Department of Neurology, Hopital Pitié-Salpêtrière, University Paris 6, Paris, France
| | | | - Janusch Blautzik
- Institute for Clinical Radiology, Department of MRI, Ludwig Maximilian University Munich, Germany
| | - Arun L W Bokde
- Cognitive Systems Group, Discipline of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland; and Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Bruno Dubois
- Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Institute of Memory and Alzheimer's Disease (IM2A) & Brain and Spine Institute (ICM) UMR S 1127, Departament of Neurology, Hopital Pitié-Salpêtrière, Paris, France
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Centre, The Netherlands
| | - Petra J W Pouwels
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Centre, The Netherlands
| | - Stefan Teipel
- DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany; Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013 Paris, France.
| | | |
Collapse
|
128
|
Knyazev GG, Savostyanov AN, Bocharov AV, Slobodskaya HR, Bairova NB, Tamozhnikov SS, Stepanova VV. Effortful control and resting state networks: A longitudinal EEG study. Neuroscience 2017; 346:365-381. [DOI: 10.1016/j.neuroscience.2017.01.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 10/20/2022]
|
129
|
Nordin K, Herlitz A, Larsson EM, Söderlund H. Overlapping effects of age on associative memory and the anterior hippocampus from middle to older age. Behav Brain Res 2017; 317:350-359. [DOI: 10.1016/j.bbr.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/28/2016] [Accepted: 10/01/2016] [Indexed: 10/20/2022]
|
130
|
Li W, Qiu Q, Sun L, Yue L, Wang T, Li X, Xiao S. Sex differences in obesity and cognitive function in a cognitively normal aging Chinese Han population. Neuropsychiatr Dis Treat 2017; 13:2405-2410. [PMID: 29066899 PMCID: PMC5604567 DOI: 10.2147/ndt.s145245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Sex differences in Alzheimer's disease and mild cognitive impairment have been well recognized. However, sex differences in cognitive function and obesity in cognitively normal aging Chinese Han population have not attracted much attention. OBJECTIVE The aim of this study was to investigate the relationship between sex, obesity, and cognitive function in an elderly Chinese population with normal cognitive function. SUBJECTS AND METHODS A total of 228 cognitively normal aging participants (males/females =93/135) entered this study. Their general demographic information (sex, age, and education) was collected by standardized questionnaire. Apolipoprotein E (APOE) genotype and serum lipid levels were measured. The Montreal Cognitive Assessment (MoCA) was used to assess participants' cognitive function. RESULTS The prevalence of obesity in elderly women (18/133, 13.5%) was significantly higher than that in men (5/92, 5.4%, P=0.009). Regression analyses showed that obesity was associated with drinking alcohol (OR =13.695, P=0.045) and triglyceride (OR =1.436, P=0.048) in women and limited to low-density lipoprotein (OR =11.829, P=0.023) in men. Women performed worse on the naming score for MoCA than men (P<0.01). Stepwise linear regression analysis showed that education (t=3.689, P<0.001) and smoking (t=2.031, P=0.045) were related to the score of naming in female, while high-density lipoprotein (t=-2.077, P=0.041) was related to the score of naming in male; however, no correlation was found between body mass index and cognitive function in both male and female (P>0.05). CONCLUSION Our finding suggests that there are significant sex differences in obesity and specific cognitive domains in aging Chinese Han population with normal cognitive function.
Collapse
Affiliation(s)
- Wei Li
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qi Qiu
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lin Sun
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ling Yue
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Tao Wang
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xia Li
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shifu Xiao
- Alzheimer's Disease and Related Disorders Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
131
|
Lin KA, Rundel C, Doraiswamy PM. Serum SHBG Levels are not Associated with Longitudinal Cognitive Decline in Mild Cognitive Impairment. J Alzheimers Dis 2016; 55:1123-1130. [DOI: 10.3233/jad-160513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Katherine Amy Lin
- Department of Psychiatry, Duke University Medical Center, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Colin Rundel
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - P. Murali Doraiswamy
- Department of Psychiatry, Duke University Medical Center, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | | |
Collapse
|
132
|
Tarai S, Bit A, dos Reis HJ, Palotás A, Rizvanov A, Bissoyi A. Stratifying Heterogeneous Dimension of Neurodegenerative Diseases: Intervention for Stipulating Epigenetic Factors to Combat Oxidative Stress in Human Brain. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0240-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
133
|
Gender-Specific Expression of Ubiquitin-Specific Peptidase 9 Modulates Tau Expression and Phosphorylation: Possible Implications for Tauopathies. Mol Neurobiol 2016; 54:7979-7993. [PMID: 27878758 PMCID: PMC5684262 DOI: 10.1007/s12035-016-0299-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/14/2016] [Indexed: 01/05/2023]
Abstract
Public transcriptomic studies have shown that several genes display pronounced gender differences in their expression in the human brain, which may influence the manifestations and risk for neuronal disorders. Here, we apply a transcriptome-wide analysis to discover genes with gender-specific expression and significant alterations in public postmortem brain tissue from Alzheimer’s disease (AD) patients compared to controls. We identify the sex-linked ubiquitin-specific peptidase 9 (USP9) as an outstanding candidate gene with highly significant expression differences between the genders and male-specific underexpression in AD. Since previous studies have shown that USP9 can modulate the phosphorylation of the AD-associated protein MAPT, we investigate functional associations between USP9 and MAPT in further detail. After observing a high positive correlation between the expression of USP9 and MAPT in the public transcriptomics data, we show that USP9 knockdown results in significantly decreased MAPT expression in a DU145 cell culture model and a concentration-dependent decrease for the MAPT orthologs mapta and maptb in a zebrafish model. From the analysis of microarray and qRT-PCR experiments for the knockdown in DU145 cells and prior knowledge from the literature, we derive a data-congruent model for a USP9-dependent regulatory mechanism modulating MAPT expression via BACH1 and SMAD4. Overall, the analyses suggest USP9 may contribute to molecular gender differences observed in tauopathies and provide a new target for intervention strategies to modulate MAPT expression.
Collapse
|
134
|
Gender, apolipoprotein E genotype, and mesial temporal atrophy: 2-year follow-up in patients with stable mild cognitive impairment and with progression from mild cognitive impairment to Alzheimer’s disease. Neuroradiology 2016; 58:1143-1151. [DOI: 10.1007/s00234-016-1740-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/10/2016] [Indexed: 11/27/2022]
|
135
|
Melnikova T, Park D, Becker L, Lee D, Cho E, Sayyida N, Tian J, Bandeen-Roche K, Borchelt DR, Savonenko AV. Sex-related dimorphism in dentate gyrus atrophy and behavioral phenotypes in an inducible tTa:APPsi transgenic model of Alzheimer's disease. Neurobiol Dis 2016; 96:171-185. [PMID: 27569580 DOI: 10.1016/j.nbd.2016.08.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 07/07/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
Sex differences are a well-known phenomenon in Alzheimer's disease (AD), with women having a higher risk for AD than men. Many AD mouse models display a similar sex-dependent pattern, with females showing earlier cognitive deficits and more severe neuropathology than males. However, whether those differences are relevant to human disease is unclear. Here we show that in AD mouse models that overexpress amyloid precursor protein (APP) under control of the prion protein promoter (PrP), female transgenic mice have higher APP expression than males, complicating interpretations of the role of sex-related factors in such models. By contrast, in a tTa:APPsi model, in which APP expression is driven by the tetracycline transactivator (tTa) from the CaMKIIα promoter, there are no sex-related differences in expression or processing of APP. In addition, the levels of Aβ dimers and tetramers, as well as Aβ peptide accumulation, are similar between sexes. Behavioral testing demonstrated that both male and female tTa:APPsi mice develop age-dependent deficits in spatial recognition memory and conditional freezing to context. These cognitive deficits were accompanied by habituation-associated hyperlocomotion and startle hyper-reactivity. Significant sex-related dimorphisms were observed, due to females showing earlier onsets of the deficits in conditioned freezing and hyperlocomotion. In addition, tTa:APPsi males but not females demonstrated a lack of novelty-induced activation. Both males and females showed atrophy of the dentate gyrus (DG) of the dorsal hippocampus, associated with widening of the pyramidal layer of the CA1 area in both sexes. Ventral DG was preserved. Sex-related differences were limited to the DG, with females showing more advanced degeneration than males. Collectively, our data show that the tTa:APPsi model is characterized by a lack of sex-related differences in APP expression, making this model useful in deciphering the mechanisms of sex differences in AD pathogenesis. Sex-related dimorphisms observed in this model under conditions of equal APP expression between sexes suggest a higher sensitivity of females to the effects of APP and/or Aβ production.
Collapse
Affiliation(s)
- Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - DaMin Park
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Lauren Becker
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Deidre Lee
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Eugenia Cho
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Nuzhat Sayyida
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Jing Tian
- Department of Biostatistics, School of Public Health, The Johns Hopkins University, 615 N Wolfe St E3527, Baltimore, MD 21205, USA.
| | - Karen Bandeen-Roche
- Department of Biostatistics, School of Public Health, The Johns Hopkins University, 615 N Wolfe St E3527, Baltimore, MD 21205, USA.
| | - David R Borchelt
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, 100 Newell Drive, Gainesville, FL 32610, USA.
| | - Alena V Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| |
Collapse
|
136
|
|
137
|
Granger MW, Franko B, Taylor MW, Messier C, George-Hyslop PS, Bennett SA. A TgCRND8 Mouse Model of Alzheimer’s Disease Exhibits Sexual Dimorphisms in Behavioral Indices of Cognitive Reserve. J Alzheimers Dis 2016; 51:757-73. [DOI: 10.3233/jad-150587] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Matthew W. Granger
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Bettina Franko
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew W. Taylor
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Claude Messier
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Peter St. George-Hyslop
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK and Tanz Centre for Research in Neurodegenerative Diseases University of Toronto, Toronto, ON, Canada
| | - Steffany A.L. Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
138
|
Ardekani BA, Convit A, Bachman AH. Analysis of the MIRIAD Data Shows Sex Differences in Hippocampal Atrophy Progression. J Alzheimers Dis 2016; 50:847-57. [DOI: 10.3233/jad-150780] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Babak A. Ardekani
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Antonio Convit
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
- Departments of Psychiatry, Medicine and Radiology, New York University School of Medicine, New York, NY, USA
| | - Alvin H. Bachman
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
139
|
Gale SD, Baxter L, Thompson J. Greater memory impairment in dementing females than males relative to sex-matched healthy controls. J Clin Exp Neuropsychol 2016; 38:527-33. [PMID: 26735615 DOI: 10.1080/13803395.2015.1132298] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Previously we demonstrated sex differences in episodic memory in healthy elderly and suggested that normative data be separated by sex. The present study extended the exploration of sex differences on memory measures into two clinical populations, mild cognitive impairment (MCI) and Alzheimer's disease (AD). Seventy-six subjects with MCI and 101 subjects with AD diagnosed by a multidisciplinary team were included. These two groups were also compared to a group of 177 healthy elderly control participants. Sex differences on the Rey Auditory Verbal Learning Test (RAVLT; total and delayed recall) raw scores and Brief Visuospatial Memory Test-Revised (BVMT-R) were demonstrated within the healthy but not the MCI or AD groups. Calculating z scores by sex for both dementing groups based on the healthy controls suggested a larger performance gap between healthy and dementing women than between healthy and dementing men. MCI females were on average 0.48 standard deviations lower for total verbal learning compared to healthy female controls than were MCI males when compared to healthy male controls. For verbal delayed recall the gap was even larger (SD = 1.09). Similarly, on the BVMT-R, a measure of visual memory, the difference was 0.60 standard deviations for total visual learning and 0.99 standard deviations for delayed recall. This same sex difference, with females showing greater impairment compared to the controls group than did the males, was also present within the AD group. The greater memory impairment in dementing females rather than males when compared to sex-matched healthy controls was unlikely to be due to more severe illness since females performed equivalently to males on the Clinical Dementia Rating Scale, Mini-Mental Status Examination, and Dementia Rating Scale, and were also similar for age, education, and apolipoprotein status. The present study suggested relatively greater memory impairment in females with MCI or AD than in controls.
Collapse
Affiliation(s)
- Shawn D Gale
- a Department of Psychology , Brigham Young University , Provo , UT , USA.,b Neuroscience Center , Brigham Young University , Provo , UT , USA
| | - Leslie Baxter
- c Department of Neuroimaging , Barrow Neurological Institute , Phoenix , AZ , USA
| | - Juliann Thompson
- a Department of Psychology , Brigham Young University , Provo , UT , USA
| |
Collapse
|
140
|
Deak F, Freeman WM, Ungvari Z, Csiszar A, Sonntag WE. Recent Developments in Understanding Brain Aging: Implications for Alzheimer's Disease and Vascular Cognitive Impairment. J Gerontol A Biol Sci Med Sci 2016; 71:13-20. [PMID: 26590911 PMCID: PMC4851715 DOI: 10.1093/gerona/glv206] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/22/2015] [Indexed: 01/18/2023] Open
Abstract
As the population of the Western world is aging, there is increasing awareness of age-related impairments in cognitive function and a rising interest in finding novel approaches to preserve cerebral health. A special collection of articles in The Journals of Gerontology: Biological Sciences and Medical Sciences brings together information of different aspects of brain aging, from latest developments in the field of neurodegenerative disorders to cerebral microvascular mechanisms of cognitive decline. It is emphasized that although the cellular changes that occur within aging neurons have been widely studied, more research is required as new signaling pathways are discovered that can potentially protect cells. New avenues for research targeting cellular senescence, epigenetics, and endocrine mechanisms of brain aging are also discussed. Based on the current literature it is clear that understanding brain aging and reducing risk for neurological disease with age requires searching for mechanisms and treatment options beyond the age-related changes in neuronal function. Thus, comprehensive approaches need to be developed that address the multiple, interrelated mechanisms of brain aging. Attention is brought to the importance of maintenance of cerebromicrovascular health, restoring neuroendocrine balance, and the pressing need for funding more innovative research into the interactions of neuronal, neuroendocrine, inflammatory and microvascular mechanisms of cognitive impairment, and Alzheimer's disease.
Collapse
Affiliation(s)
- Ferenc Deak
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Willard M Freeman
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - Anna Csiszar
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center.
| |
Collapse
|
141
|
Cacciottolo M, Christensen A, Moser A, Liu J, Pike CJ, Smith C, LaDu MJ, Sullivan PM, Morgan TE, Dolzhenko E, Charidimou A, Wahlund LO, Wiberg MK, Shams S, Chiang GCY, Finch CE. The APOE4 allele shows opposite sex bias in microbleeds and Alzheimer's disease of humans and mice. Neurobiol Aging 2016; 37:47-57. [PMID: 26686669 PMCID: PMC4687024 DOI: 10.1016/j.neurobiolaging.2015.10.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/06/2015] [Accepted: 10/11/2015] [Indexed: 12/21/2022]
Abstract
The apolipoprotein APOE4 allele confers greater risk of Alzheimer's disease (AD) for women than men, in conjunction with greater clinical deficits per unit of AD neuropathology (plaques, tangles). Cerebral microbleeds, which contribute to cognitive dysfunctions during AD, also show APOE4 excess, but sex-APOE allele interactions are not described. We report that elderly men diagnosed for mild cognitive impairment and AD showed a higher risk of cerebral cortex microbleeds with APOE4 allele dose effect in 2 clinical cohorts (ADNI and KIDS). Sex-APOE interactions were further analyzed in EFAD mice carrying human APOE alleles and familial AD genes (5XFAD (+/-) /human APOE(+/+)). At 7 months, E4FAD mice had cerebral cortex microbleeds with female excess, in contrast to humans. Cerebral amyloid angiopathy, plaques, and soluble Aβ also showed female excess. Both the cerebral microbleeds and cerebral amyloid angiopathy increased in proportion to individual Aβ load. In humans, the opposite sex bias of APOE4 allele for microbleeds versus the plaques and tangles is the first example of organ-specific, sex-linked APOE allele effects, and further shows AD as a uniquely human condition.
Collapse
Affiliation(s)
- Mafalda Cacciottolo
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Christensen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Alexandra Moser
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jiahui Liu
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Conor Smith
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University, Durham VA Medical Center and GRECC, Durham, NC, USA
| | - Todd E Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Egor Dolzhenko
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Andreas Charidimou
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden; Division of Clinical Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Kristofferson Wiberg
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Shams
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention, and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Caleb E Finch
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA; Department of Biological Sciences, Dornsife College, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
142
|
Cerebral atrophy in mild cognitive impairment: A systematic review with meta-analysis. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2015; 1:487-504. [PMID: 27239527 PMCID: PMC4879488 DOI: 10.1016/j.dadm.2015.11.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Although mild cognitive impairment (MCI) diagnosis is mainly based on cognitive assessment, reliable estimates of structural changes in specific brain regions, that could be contrasted against normal brain aging and inform diagnosis, are lacking. This study aimed to systematically review the literature reporting on MCI-related brain changes. METHODS The MEDLINE database was searched for studies investigating longitudinal structural changes in MCI. Studies with compatible data were included in the meta-analyses. A qualitative review was conducted for studies excluded from meta-analyses. RESULTS The analyses revealed a 2.2-fold higher volume loss in the hippocampus, 1.8-fold in the whole brain, and 1.5-fold in the entorhinal cortex in MCI participants. DISCUSSION Although the medial temporal lobe is likely to be more vulnerable to MCI pathology, atrophy in this brain area represents a relatively small proportion of whole brain loss, suggesting that future investigations are needed to identify the source of unaccounted volume loss in MCI.
Collapse
|
143
|
Hua X, Ching CRK, Mezher A, Gutman BA, Hibar DP, Bhatt P, Leow AD, Jack CR, Bernstein MA, Weiner MW, Thompson PM. MRI-based brain atrophy rates in ADNI phase 2: acceleration and enrichment considerations for clinical trials. Neurobiol Aging 2015; 37:26-37. [PMID: 26545631 PMCID: PMC4827255 DOI: 10.1016/j.neurobiolaging.2015.09.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 08/30/2015] [Accepted: 09/22/2015] [Indexed: 01/31/2023]
Abstract
The goal of this work was to assess statistical power to detect treatment effects in Alzheimer’s disease (AD) clinical trials using magnetic resonance imaging (MRI)–derived brain biomarkers. We used unbiased tensor-based morphometry (TBM) to analyze n = 5,738 scans, from Alzheimer’s Disease Neuroimaging Initiative 2 participants scanned with both accelerated and nonaccelerated T1-weighted MRI at 3T. The study cohort included 198 healthy controls, 111 participants with significant memory complaint, 182 with early mild cognitive impairment (EMCI) and 177 late mild cognitive impairment (LMCI), and 155 AD patients, scanned at screening and 3, 6, 12, and 24 months. The statistical power to track brain change in TBM-based imaging biomarkers depends on the interscan interval, disease stage, and methods used to extract numerical summaries. To achieve reasonable sample size estimates for potential clinical trials, the minimal scan interval was 6 months for LMCI and AD and 12 months for EMCI. TBM-based imaging biomarkers were not sensitive to MRI scan acceleration, which gave results comparable with nonaccelerated sequences. ApoE status and baseline amyloid-beta positron emission tomography data improved statistical power. Among healthy, EMCI, and LMCI participants, sample size requirements were significantly lower in the amyloid+/ApoE4+ group than for the amyloid−/ApoE4− group. ApoE4 strongly predicted atrophy rates across brain regions most affected by AD, but the remaining 9 of the top 10 AD risk genes offered no added predictive value in this cohort.
Collapse
Affiliation(s)
- Xue Hua
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA; Interdepartmental Neuroscience Graduate Program, University of California, Los Angeles, School of Medicine, Los Angeles, CA, USA
| | - Adam Mezher
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA
| | - Boris A Gutman
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA
| | - Derrek P Hibar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Priya Bhatt
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA
| | - Alex D Leow
- Department of Psychiatry, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | - Michael W Weiner
- Department of Radiology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine and Psychiatry, University of California, San Francisco, San Francisco, CA, USA; Department Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Marina del Rey, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Psychiatry, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Engineering, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | | |
Collapse
|
144
|
Lin KA, Choudhury KR, Rathakrishnan BG, Marks DM, Petrella JR, Doraiswamy PM. Marked gender differences in progression of mild cognitive impairment over 8 years. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2015; 1:103-110. [PMID: 26451386 PMCID: PMC4593067 DOI: 10.1016/j.trci.2015.07.001] [Citation(s) in RCA: 269] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Introduction This study examined whether, among subjects with mild cognitive impairment (MCI), women progressed at faster rates than men. Methods We examine longitudinal rates of change from baseline in 398 MCI subjects (141 females and 257 males) in the Alzheimer's Disease Neuroimaging Initiative-1, followed for up to 8 years (mean, 4.1 ± 2.5 years) using mixed-effects models incorporating all follow-ups (mean, 8 ± 4 visits). Results Women progressed at faster rates than men on the Alzheimer's disease assessment scale-cognitive subscale (ADAS-Cog; P = .001) and clinical dementia rating-sum of boxes (CDR-SB; P = .003). Quadratic fit for change over time was significant for both ADAS-Cog (P = .001) and CDR-SB (P = .004), and the additional acceleration in women was 100% for ADAS-Cog and 143% for CDR-SB. The variability of change was greater in women. The gender effect was greater in apolipoprotein E (APOE) ε4 carriers. Discussion Women with MCI have greater longitudinal rates of cognitive and functional progression than men. Studies to confirm and uncover potential mechanisms appear to be warranted. Trial Registration ADNI ClinicalTrials.gov identifier: NCT00106899.
Collapse
Affiliation(s)
- Katherine A Lin
- Department of Psychiatry, Duke University Medical Center, 2301 Erwin Road, Durham, NC 27710 ; Duke Institute for Brain Sciences, Duke University, Box 91003, Levine Science Research Center, Room B107, 450 Research Drive, Durham, North Carolina 27708
| | - Kingshuk Roy Choudhury
- Department of Radiology, Duke University Medical Center, Box 3808, 2301 Erwin Road, Durham, NC 27710
| | | | - David M Marks
- Department of Psychiatry, Duke University Medical Center, 2301 Erwin Road, Durham, NC 27710
| | - Jeffrey R Petrella
- Department of Radiology, Duke University Medical Center, Box 3808, 2301 Erwin Road, Durham, NC 27710
| | - P Murali Doraiswamy
- Department of Psychiatry, Duke University Medical Center, 2301 Erwin Road, Durham, NC 27710 ; Duke Institute for Brain Sciences, Duke University, Box 91003, Levine Science Research Center, Room B107, 450 Research Drive, Durham, North Carolina 27708
| | | |
Collapse
|
145
|
Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, Green RC, Harvey D, Jack CR, Jagust W, Luthman J, Morris JC, Petersen RC, Saykin AJ, Shaw L, Shen L, Schwarz A, Toga AW, Trojanowski JQ. 2014 Update of the Alzheimer's Disease Neuroimaging Initiative: A review of papers published since its inception. Alzheimers Dement 2015; 11:e1-120. [PMID: 26073027 PMCID: PMC5469297 DOI: 10.1016/j.jalz.2014.11.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/18/2013] [Indexed: 01/18/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) is an ongoing, longitudinal, multicenter study designed to develop clinical, imaging, genetic, and biochemical biomarkers for the early detection and tracking of Alzheimer's disease (AD). The initial study, ADNI-1, enrolled 400 subjects with early mild cognitive impairment (MCI), 200 with early AD, and 200 cognitively normal elderly controls. ADNI-1 was extended by a 2-year Grand Opportunities grant in 2009 and by a competitive renewal, ADNI-2, which enrolled an additional 550 participants and will run until 2015. This article reviews all papers published since the inception of the initiative and summarizes the results to the end of 2013. The major accomplishments of ADNI have been as follows: (1) the development of standardized methods for clinical tests, magnetic resonance imaging (MRI), positron emission tomography (PET), and cerebrospinal fluid (CSF) biomarkers in a multicenter setting; (2) elucidation of the patterns and rates of change of imaging and CSF biomarker measurements in control subjects, MCI patients, and AD patients. CSF biomarkers are largely consistent with disease trajectories predicted by β-amyloid cascade (Hardy, J Alzheimer's Dis 2006;9(Suppl 3):151-3) and tau-mediated neurodegeneration hypotheses for AD, whereas brain atrophy and hypometabolism levels show predicted patterns but exhibit differing rates of change depending on region and disease severity; (3) the assessment of alternative methods of diagnostic categorization. Currently, the best classifiers select and combine optimum features from multiple modalities, including MRI, [(18)F]-fluorodeoxyglucose-PET, amyloid PET, CSF biomarkers, and clinical tests; (4) the development of blood biomarkers for AD as potentially noninvasive and low-cost alternatives to CSF biomarkers for AD diagnosis and the assessment of α-syn as an additional biomarker; (5) the development of methods for the early detection of AD. CSF biomarkers, β-amyloid 42 and tau, as well as amyloid PET may reflect the earliest steps in AD pathology in mildly symptomatic or even nonsymptomatic subjects and are leading candidates for the detection of AD in its preclinical stages; (6) the improvement of clinical trial efficiency through the identification of subjects most likely to undergo imminent future clinical decline and the use of more sensitive outcome measures to reduce sample sizes. Multimodal methods incorporating APOE status and longitudinal MRI proved most highly predictive of future decline. Refinements of clinical tests used as outcome measures such as clinical dementia rating-sum of boxes further reduced sample sizes; (7) the pioneering of genome-wide association studies that leverage quantitative imaging and biomarker phenotypes, including longitudinal data, to confirm recently identified loci, CR1, CLU, and PICALM and to identify novel AD risk loci; (8) worldwide impact through the establishment of ADNI-like programs in Japan, Australia, Argentina, Taiwan, China, Korea, Europe, and Italy; (9) understanding the biology and pathobiology of normal aging, MCI, and AD through integration of ADNI biomarker and clinical data to stimulate research that will resolve controversies about competing hypotheses on the etiopathogenesis of AD, thereby advancing efforts to find disease-modifying drugs for AD; and (10) the establishment of infrastructure to allow sharing of all raw and processed data without embargo to interested scientific investigators throughout the world.
Collapse
Affiliation(s)
- Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Laurel A Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse Cedarbaum
- Neurology Early Clinical Development, Biogen Idec, Cambridge, MA, USA
| | - Robert C Green
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | | | - William Jagust
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Johan Luthman
- Neuroscience Clinical Development, Neuroscience & General Medicine Product Creation Unit, Eisai Inc., Philadelphia, PA, USA
| | - John C Morris
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leslie Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adam Schwarz
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, Institute of Neuroimaging and Informatics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - John Q Trojanowski
- Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Alzheimer's Disease Core Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Udall Parkinson's Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
146
|
Lin KA, Doraiswamy PM. When Mars Versus Venus is Not a Cliché: Gender Differences in the Neurobiology of Alzheimer's Disease. Front Neurol 2015; 5:288. [PMID: 25628598 PMCID: PMC4290582 DOI: 10.3389/fneur.2014.00288] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/18/2014] [Indexed: 11/13/2022] Open
Affiliation(s)
- Katherine Amy Lin
- Division of Translational Neuroscience, Department of Psychiatry, Duke University Medical Center , Durham, NC , USA ; Duke Institute for Brain Sciences , Durham, NC , USA
| | - P Murali Doraiswamy
- Division of Translational Neuroscience, Department of Psychiatry, Duke University Medical Center , Durham, NC , USA ; Duke Institute for Brain Sciences , Durham, NC , USA
| |
Collapse
|
147
|
Turner JA. The rise of large-scale imaging studies in psychiatry. Gigascience 2014; 3:29. [PMID: 25793106 PMCID: PMC4365768 DOI: 10.1186/2047-217x-3-29] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/07/2014] [Indexed: 11/13/2022] Open
Abstract
From the initial arguments over whether 12 to 20 subjects were sufficient for an fMRI study, sample sizes in psychiatric neuroimaging studies have expanded into the tens of thousands. These large-scale imaging studies fall into several categories, each of which has specific advantages and challenges. The different study types can be grouped based on their level of control: meta-analyses, at one extreme of the spectrum, control nothing about the imaging protocol or subject selection criteria in the datasets they include, On the other hand, planned multi-site mega studies pour intense efforts into strictly having the same protocols. However, there are several other combinations possible, each of which is best used to address certain questions. The growing investment of all these studies is delivering on the promises of neuroimaging for psychiatry, and holds incredible potential for impact at the level of the individual patient. However, to realize this potential requires both standardized data-sharing efforts, so that there is more staying power in the datasets for re-use and new applications, as well as training the next generation of neuropsychiatric researchers in "Big Data" techniques in addition to traditional experimental methods. The increased access to thousands of datasets along with the needed informatics demands a new emphasis on integrative scientific methods.
Collapse
Affiliation(s)
- Jessica A Turner
- Department of Psychology and Neuroscience Institute, Georgia State University, P.O .Box 5010, Atlanta, GA 30302 USA
| |
Collapse
|
148
|
Duriez Q, Crivello F, Mazoyer B. Sex-related and tissue-specific effects of tobacco smoking on brain atrophy: assessment in a large longitudinal cohort of healthy elderly. Front Aging Neurosci 2014; 6:299. [PMID: 25404916 PMCID: PMC4217345 DOI: 10.3389/fnagi.2014.00299] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/15/2014] [Indexed: 01/07/2023] Open
Abstract
We investigated the cross-sectional and longitudinal effects of tobacco smoking on brain atrophy in a large cohort of healthy elderly participants (65–80 years). MRI was used for measuring whole brain (WB), gray matter (GM), white matter (WM), and hippocampus (HIP) volumes at study entry time (baseline, N = 1451), and the annualized rates of variation of these volumes using a 4-year follow-up MRI in a subpart of the cohort (N = 1111). Effects of smoking status (never, former, or current smoker) at study entry and of lifetime tobacco consumption on these brain phenotypes were studied using sex-stratified AN(C)OVAs, including other health parameters as covariates. At baseline, male current smokers had lower GM, while female current smokers had lower WM. In addition, female former smokers exhibited reduced baseline HIP, the reduction being correlated with lifetime tobacco consumption. Longitudinal analyses demonstrated that current smokers, whether men or women, had larger annualized rates of HIP atrophy, as compared to either non or former smokers, independent of their lifetime consumption of tobacco. There was no effect of smoking on the annualized rate of WM loss. In all cases, measured sizes of these tobacco-smoking effects were of the same order of magnitude than those of age, and larger than effect sizes of any other covariate. These results demonstrate that tobacco smoking is a major factor of brain aging, with sex- and tissue specific effects, notably on the HIP annualized rate of atrophy after the age of 65.
Collapse
Affiliation(s)
- Quentin Duriez
- Life Sciences, University of Bordeaux, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Centre National de la Recherche Scientifique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Commisariat à l'Energie Atomique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France
| | - Fabrice Crivello
- Life Sciences, University of Bordeaux, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Centre National de la Recherche Scientifique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Commisariat à l'Energie Atomique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France
| | - Bernard Mazoyer
- Life Sciences, University of Bordeaux, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Centre National de la Recherche Scientifique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France ; Commisariat à l'Energie Atomique, Neurofunctional Imaging Group (GIN) UMR5296 Bordeaux, France
| |
Collapse
|
149
|
Fouquet M, Besson FL, Gonneaud J, La Joie R, Chételat G. Imaging Brain Effects of APOE4 in Cognitively Normal Individuals Across the Lifespan. Neuropsychol Rev 2014; 24:290-9. [DOI: 10.1007/s11065-014-9263-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/23/2014] [Indexed: 12/21/2022]
|
150
|
Shen L, Thompson PM, Potkin SG, Bertram L, Farrer LA, Foroud TM, Green RC, Hu X, Huentelman MJ, Kim S, Kauwe JSK, Li Q, Liu E, Macciardi F, Moore JH, Munsie L, Nho K, Ramanan VK, Risacher SL, Stone DJ, Swaminathan S, Toga AW, Weiner MW, Saykin AJ, for the Alzheimer’s Disease Neuroimaging Initiative. Genetic analysis of quantitative phenotypes in AD and MCI: imaging, cognition and biomarkers. Brain Imaging Behav 2014; 8:183-207. [PMID: 24092460 PMCID: PMC3976843 DOI: 10.1007/s11682-013-9262-z] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The Genetics Core of the Alzheimer's Disease Neuroimaging Initiative (ADNI), formally established in 2009, aims to provide resources and facilitate research related to genetic predictors of multidimensional Alzheimer's disease (AD)-related phenotypes. Here, we provide a systematic review of genetic studies published between 2009 and 2012 where either ADNI APOE genotype or genome-wide association study (GWAS) data were used. We review and synthesize ADNI genetic associations with disease status or quantitative disease endophenotypes including structural and functional neuroimaging, fluid biomarker assays, and cognitive performance. We also discuss the diverse analytical strategies used in these studies, including univariate and multivariate analysis, meta-analysis, pathway analysis, and interaction and network analysis. Finally, we perform pathway and network enrichment analyses of these ADNI genetic associations to highlight key mechanisms that may drive disease onset and trajectory. Major ADNI findings included all the top 10 AD genes and several of these (e.g., APOE, BIN1, CLU, CR1, and PICALM) were corroborated by ADNI imaging, fluid and cognitive phenotypes. ADNI imaging genetics studies discovered novel findings (e.g., FRMD6) that were later replicated on different data sets. Several other genes (e.g., APOC1, FTO, GRIN2B, MAGI2, and TOMM40) were associated with multiple ADNI phenotypes, warranting further investigation on other data sets. The broad availability and wide scope of ADNI genetic and phenotypic data has advanced our understanding of the genetic basis of AD and has nominated novel targets for future studies employing next-generation sequencing and convergent multi-omics approaches, and for clinical drug and biomarker development.
Collapse
Affiliation(s)
- Li Shen
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Paul M. Thompson
- Imaging Genetics Center, Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
| | - Steven G. Potkin
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
| | - Lars Bertram
- Neuropsychiatric Genetics Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lindsay A. Farrer
- Biomedical Genetics L320, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA
| | - Tatiana M. Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Robert C. Green
- Division of Genetics and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115 USA
| | - Xiaolan Hu
- Clinical Genetics, Exploratory Clinical & Translational Research, Bristol-Myers Squibbs, Pennington, NJ 08534 USA
| | - Matthew J. Huentelman
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ 85004 USA
| | - Sungeun Kim
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - John S. K. Kauwe
- Departments of Biology, Neuroscience, Brigham Young University, 675 WIDB, Provo, UT 84602 USA
| | - Qingqin Li
- Department of Neuroscience Biomarkers, Janssen Research and Development, LLC, Raritan, NJ 08869 USA
| | - Enchi Liu
- Biomarker Discovery, Janssen Alzheimer Immunotherapy Research and Development, LLC, South San Francisco, CA 94080 USA
| | - Fabio Macciardi
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
- Department of Sciences and Biomedical Technologies, University of Milan, Segrate, MI Italy
| | - Jason H. Moore
- Department of Genetics, Computational Genetics Laboratory, Dartmouth Medical School, Lebanon, NH 03756 USA
| | - Leanne Munsie
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN 46285 USA
| | - Kwangsik Nho
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Vijay K. Ramanan
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Shannon L. Risacher
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - David J. Stone
- Merck Research Laboratories, 770 Sumneytown Pike, WP53B-120, West Point, PA 19486 USA
| | - Shanker Swaminathan
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
| | - Michael W. Weiner
- Departments of Radiology, Medicine and Psychiatry, UC San Francisco, San Francisco, CA 94143 USA
| | - Andrew J. Saykin
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Center for Neuroimaging and Indiana Alzheimer’s Disease Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 16th Street, Suite 4100, Indianapolis, IN 46202 USA
- Imaging Genetics Center, Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92617 USA
- Neuropsychiatric Genetics Group, Max-Planck Institute for Molecular Genetics, Berlin, Germany
- Biomedical Genetics L320, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118 USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Division of Genetics and Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115 USA
- Clinical Genetics, Exploratory Clinical & Translational Research, Bristol-Myers Squibbs, Pennington, NJ 08534 USA
- Neurogenomics Division, The Translational Genomics Research Institute, Phoenix, AZ 85004 USA
- Departments of Biology, Neuroscience, Brigham Young University, 675 WIDB, Provo, UT 84602 USA
- Department of Neuroscience Biomarkers, Janssen Research and Development, LLC, Raritan, NJ 08869 USA
- Biomarker Discovery, Janssen Alzheimer Immunotherapy Research and Development, LLC, South San Francisco, CA 94080 USA
- Department of Sciences and Biomedical Technologies, University of Milan, Segrate, MI Italy
- Department of Genetics, Computational Genetics Laboratory, Dartmouth Medical School, Lebanon, NH 03756 USA
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN 46285 USA
- Merck Research Laboratories, 770 Sumneytown Pike, WP53B-120, West Point, PA 19486 USA
- Laboratory of Neuro Imaging, Department of Neurology, UCLA School of Medicine, Los Angeles, CA 90095 USA
- Departments of Radiology, Medicine and Psychiatry, UC San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|