51
|
Bisht K, Sharma K, Tremblay MÈ. Chronic stress as a risk factor for Alzheimer's disease: Roles of microglia-mediated synaptic remodeling, inflammation, and oxidative stress. Neurobiol Stress 2018; 9:9-21. [PMID: 29992181 PMCID: PMC6035903 DOI: 10.1016/j.ynstr.2018.05.003] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/23/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Microglia are the predominant immune cells of the central nervous system (CNS) that exert key physiological roles required for maintaining CNS homeostasis, notably in response to chronic stress, as well as mediating synaptic plasticity, learning and memory. The repeated exposure to stress confers a higher risk of developing neurodegenerative diseases including sporadic Alzheimer's disease (AD). While microglia have been causally linked to amyloid beta (Aβ) accumulation, tau pathology, neurodegeneration, and synaptic loss in AD, they were also attributed beneficial roles, notably in the phagocytic elimination of Aβ. In this review, we discuss the interactions between chronic stress and AD pathology, overview the roles played by microglia in AD, especially focusing on chronic stress as an environmental risk factor modulating their function, and present recently-described microglial phenotypes associated with neuroprotection in AD. These microglial phenotypes observed under both chronic stress and AD pathology may provide novel opportunities for the development of better-targeted therapeutic interventions.
Collapse
Key Words
- ABCA7, ATP-binding cassette transporter A7
- AD, Alzheimer's disease
- APOE, Apolipoprotein E
- APP, amyloid precursor protein
- Alzheimer's disease
- Aβ, Amyloid beta
- BDNF, brain derived neurotrophic factor
- CD11b, cluster of differentiation molecule 11B
- CD33, cluster of differentiation 33
- CNS, central nervous system
- CR, complement receptor
- CRF, corticotropin releasing factor
- DAM, disease associated microglia
- DAP12, DNAX-activation protein 12
- Dark microglia
- FAD, Familial Alzheimer's disease
- FCRLS, Fc receptor-like S scavenger receptor
- GR, glucocorticoid receptor
- HPA axis, hypothalamic pituitary adrenocortical axis
- IBA1, ionized calcium-binding adapter molecule 1
- IL, interleukin
- LTP, long-term potentiation
- MGnD, microglia with a neurodegenerative phenotype
- MR, mineralocorticoid receptor
- Microglia
- Microglial phenotypes
- NADPH, nicotinamide adenine dinucleotide phosphate
- NFT, neurofibrillary tangles
- Neurodegeneration
- Neuroinflammation
- PS, presenilin
- ROS, reactive oxygen species
- Stress
- Synaptic remodeling
- TGFβ, transforming growth factor β
- TLR, Toll-like receptors
- TMEM119, transmembrane protein 119
- TNFα, tumor necrosis factor-α
- TREM2, triggering receptor expressed in myeloid cells 2
- TYROBP, TYRO protein tyrosine kinase binding protein
- mPFC, medial prefrontal cortex
Collapse
Affiliation(s)
- Kanchan Bisht
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Kaushik Sharma
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, Québec, QC, Canada
- Département de médecine moléculaire, Université Laval, Québec, QC, Canada
| |
Collapse
|
52
|
Yuede CM, Timson BF, Hettinger JC, Yuede KM, Edwards HM, Lawson JE, Zimmerman SD, Cirrito JR. Interactions between stress and physical activity on Alzheimer's disease pathology. Neurobiol Stress 2018; 8:158-171. [PMID: 29888311 PMCID: PMC5991353 DOI: 10.1016/j.ynstr.2018.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022] Open
Abstract
Physical activity and stress are both environmental modifiers of Alzheimer's disease (AD) risk. Animal studies of physical activity in AD models have largely reported positive results, however benefits are not always observed in either cognitive or pathological outcomes and inconsistencies among findings remain. Studies using forced exercise may increase stress and mitigate some of the benefit of physical activity in AD models, while voluntary exercise regimens may not achieve optimal intensity to provide robust benefit. We evaluated the findings of studies of voluntary and forced exercise regimens in AD mouse models to determine the influence of stress, or the intensity of exercise needed to outweigh the negative effects of stress on AD measures. In addition, we show that chronic physical activity in a mouse model of AD can prevent the effects of acute restraint stress on Aβ levels in the hippocampus. Stress and physical activity have many overlapping and divergent effects on the body and some of the possible mechanisms through which physical activity may protect against stress-induced risk factors for AD are discussed. While the physiological effects of acute stress and acute exercise overlap, chronic effects of physical activity appear to directly oppose the effects of chronic stress on risk factors for AD. Further study is needed to identify optimal parameters for intensity, duration and frequency of physical activity to counterbalance effects of stress on the development and progression of AD.
Collapse
Affiliation(s)
- Carla M Yuede
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA.,Department of Psychiatry, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Benjamin F Timson
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - Jane C Hettinger
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Kayla M Yuede
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Hannah M Edwards
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| | - Justin E Lawson
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - Scott D Zimmerman
- Biomedical Science Department, Missouri State University, Springfield, MO, USA
| | - John R Cirrito
- Department of Neurology, Washington University in St. Louis, School of Medicine, St. Louis, MO, USA
| |
Collapse
|
53
|
Hoeijmakers L, Lesuis SL, Krugers H, Lucassen PJ, Korosi A. A preclinical perspective on the enhanced vulnerability to Alzheimer's disease after early-life stress. Neurobiol Stress 2018; 8:172-185. [PMID: 29888312 PMCID: PMC5991337 DOI: 10.1016/j.ynstr.2018.02.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022] Open
Abstract
Stress experienced early in life (ES), in the form of childhood maltreatment, maternal neglect or trauma, enhances the risk for cognitive decline in later life. Several epidemiological studies have now shown that environmental and adult life style factors influence AD incidence or age-of-onset and early-life environmental conditions have attracted attention in this respect. There is now emerging interest in understanding whether ES impacts the risk to develop age-related neurodegenerative disorders, and their severity, such as in Alzheimer's disease (AD), which is characterized by cognitive decline and extensive (hippocampal) neuropathology. While this might be relevant for the identification of individuals at risk and preventive strategies, this topic and its possible underlying mechanisms have been poorly studied to date. In this review, we discuss the role of ES in modulating AD risk and progression, primarily from a preclinical perspective. We focus on the possible involvement of stress-related, neuro-inflammatory and metabolic factors in mediating ES-induced effects on later neuropathology and the associated impairments in neuroplasticity. The available studies suggest that the age of onset and progression of AD-related neuropathology and cognitive decline can be affected by ES, and may aggravate the progression of AD neuropathology. These relevant changes in AD pathology after ES exposure in animal models call for future clinical studies to elucidate whether stress exposure during the early-life period in humans modulates later vulnerability for AD.
Collapse
Affiliation(s)
| | | | | | | | - Aniko Korosi
- Brain Plasticity Group, Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, Amsterdam, The Netherlands
| |
Collapse
|
54
|
Yan Y, Dominguez S, Fisher DW, Dong H. Sex differences in chronic stress responses and Alzheimer's disease. Neurobiol Stress 2018; 8:120-126. [PMID: 29888307 PMCID: PMC5991323 DOI: 10.1016/j.ynstr.2018.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/10/2018] [Accepted: 03/17/2018] [Indexed: 11/13/2022] Open
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and severity, but the mechanisms underlying this sex divergence are unknown. Though some have suggested this difference in risk is a reflection of known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities towards the disease. While a number of potential risk factors have been hypothesized to affect these differences in risks, none have been definitively verified. In this review, we discuss a novel hypothesis whereby women's susceptibility to chronic stress also mediates increased risk for AD. As stress is a risk factor for AD, and women are twice as likely to develop mood disorders where stress is a major etiology, it is possible that sex dimorphisms in stress responses contribute to the increase in women with AD. In line with this, sex divergence in biochemical responses to stress have been noted along the hypothalamic-pituitary-adrenal (HPA) axis and among known molecular effectors of AD, with crosstalk between these processes also being likely. In addition, activation of the cortical corticotrophin-releasing factor receptor 1 (CRF1) signaling pathway leads to distinct female-biased increases in molecules associated with AD pathogenesis. Therefore, the different biochemical responses to stress between women and men may represent an intrinsic, sex-dependent risk factor for AD.
Collapse
Affiliation(s)
- Yan Yan
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| | - Sky Dominguez
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Daniel W. Fisher
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| |
Collapse
|
55
|
Sindi S, Kåreholt I, Spulber G, Soininen H, Kivipelto M, Solomon A. Midlife Work-Related Stress is Associated with Late-Life Gray Matter Volume Atrophy. J Alzheimers Dis Rep 2017; 1:219-227. [PMID: 30480239 PMCID: PMC6159714 DOI: 10.3233/adr-170035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background: Work-related stress has been associated with an increased dementia risk. However, less is known about the mechanisms that underlie these associations. Objective: The goal is to examine associations between midlife work-related stress and late-life structural brain alterations. Methods: The Cardiovascular Risk Factors, Aging, and Dementia (CAIDE) study participants were randomly selected from independent population-based surveys (mean age 50) in Finland. MRI measurements included gray matter (GM) volume, white matter lesions (WML) and medial temporal atrophy (MTA) (1st re-examination, n = 102); and GM volume, hippocampal volume, WML volume, cortical thickness, and MTA (2nd re-examination, n = 64). Work-related stress comprised a score from two questions administered in midlife. Results: Higher levels of midlife work-related stress were associated with lower GM volume (β= –0.077, p = 0.033) at the first re-examination, even after adjusting for several confounders. No significant associations were found with MTA, WML, or MRI measurements at the second re-examination. Conclusion: Previously shown associations of midlife work-related stress with dementia risk may be at least partly explained by associations with lower GM volume in late-life. The lack of associations at the second re-examination may indicate a critical time window for the effects of midlife work-related stress, and/or selective survival/participation.
Collapse
Affiliation(s)
- Shireen Sindi
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Karolinska Institutet Center for Alzheimer Research, Stockholm, Sweden.,Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, UK
| | - Ingemar Kåreholt
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Gabriela Spulber
- Karolinska Institutet Center for Alzheimer Research, Stockholm, Sweden
| | - Hilkka Soininen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Neurology, University of Eastern Finland, and Kuopio University Hospital, Kuopio, Finland
| | - Miia Kivipelto
- Karolinska Institutet Center for Alzheimer Research, Stockholm, Sweden.,Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, UK.,Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Finland.,Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Alina Solomon
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden.,Karolinska Institutet Center for Alzheimer Research, Stockholm, Sweden.,Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Geriatrics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
56
|
Emerging role of amyloid beta in stress response: Implication for depression and diabetes. Eur J Pharmacol 2017; 817:22-29. [PMID: 28844871 DOI: 10.1016/j.ejphar.2017.08.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/11/2022]
|
57
|
Zhang C, Rissman RA. Corticotropin-releasing factor receptor-1 modulates biomarkers of DNA oxidation in Alzheimer's disease mice. PLoS One 2017; 12:e0181367. [PMID: 28750017 PMCID: PMC5531470 DOI: 10.1371/journal.pone.0181367] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/29/2017] [Indexed: 11/28/2022] Open
Abstract
Increased production of hydroxyl radical is the main source of oxidative damage in mammalian DNA that accumulates in Alzheimer’s disease (AD). Reactive oxygen species (ROS) react with both nuclear DNA (nDNA) and mitochondrial DNA (mtDNA) to generate 8-hydroxy-2’-deoxyguanosine (8-OHdG), both of which can be measured in the urine. Knowledge of this pathway has positioned measurement of urine 8-OHdG as a reliable index of DNA oxidation and a potential biomarker target for tracking early cellular dysfunction in AD. Furthermore, epigenetic studies demonstrate decreased global DNA methylation levels (e.g. 5-methyl-2’-deoxycytidine, 5-mdC) in AD tissues. Moreover, stress hormones can activate neuronal oxidative stress which will stimulate the release of additional stress hormones and result in damages to hippocampal neurons in the AD brain. Our previous work suggests that treating AD transgenic mice the type-1 corticotropin-releasing factor receptor (CRFR1) antagonist, R121919, to reduce stress signaling, prevented onset of cognitive impairment, synaptic/dendritic loss and Aβ plaque accumulation. Therefore, to investigate whether levels of DNA oxidation can be impacted by the same therapeutic approach, urine levels of hydrogen peroxide, 8-OHdG, 5-mdC and total antioxidant capacity (TAC) were analyzed using an AD Tg mouse model. We found that Tg animals had an 80% increase in hydrogen peroxide levels compared to wild type (Wt) counterparts, an effect that could be dramatically reversed by the chronic administration with R121919. A significant decrease of 8-OHdG levels was observed in Tg mice treated with CRFR1 antagonist. Collectively our data suggest that the beneficial effects of CRFR1 antagonism seen in Tg mice may be mechanistically linked to the modulation of oxidative stress pathways.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Neurosciences, University of California, San Diego School of Medicine, La Jolla, California, United States of America
| | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego School of Medicine, La Jolla, California, United States of America
- Veterans Affairs San Diego Healthcare System, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
58
|
Ennis GE, An Y, Resnick SM, Ferrucci L, O'Brien RJ, Moffat SD. Long-term cortisol measures predict Alzheimer disease risk. Neurology 2016; 88:371-378. [PMID: 27986873 DOI: 10.1212/wnl.0000000000003537] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/17/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To examine whether long-term measures of cortisol predict Alzheimer disease (AD) risk. METHOD We used a prospective longitudinal design to examine whether cortisol dysregulation was related to AD risk. Participants were from the Baltimore Longitudinal Study of Aging (BLSA) and submitted multiple 24-hour urine samples over an average interval of 10.56 years. Urinary free cortisol (UFC) and creatinine (Cr) were measured, and a UFC/Cr ratio was calculated to standardize UFC. To measure cortisol regulation, we used within-person UFC/Cr level (i.e., within-person mean), change in UFC/Cr over time (i.e., within-person slope), and UFC/Cr variability (i.e., within-person coefficient of variation). Cox regression was used to assess whether UFC/Cr measures predicted AD risk. RESULTS UFC/Cr level and UFC/Cr variability, but not UFC/Cr slope, were significant predictors of AD risk an average of 2.9 years before AD onset. Elevated UFC/Cr level and elevated UFC/Cr variability were related to a 1.31- and 1.38-times increase in AD risk, respectively. In a sensitivity analysis, increased UFC/Cr level and increased UFC/Cr variability predicted increased AD risk an average of 6 years before AD onset. CONCLUSIONS Cortisol dysregulation as manifested by high UFC/Cr level and high UFC/Cr variability may modulate the downstream clinical expression of AD pathology or be a preclinical marker of AD.
Collapse
Affiliation(s)
- Gilda E Ennis
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC
| | - Yang An
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC
| | - Susan M Resnick
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC
| | - Luigi Ferrucci
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC
| | - Richard J O'Brien
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC
| | - Scott D Moffat
- From the School of Psychology (G.E.E., S.D.M.), Georgia Institute of Technology, Atlanta; Laboratory of Behavioral Neuroscience (Y.A., S.M.R.) and Longitudinal Studies Section (L.F.), National Institute on Aging, Baltimore, MD; and School of Medicine (R.J.O.), Johns Hopkins University, Baltimore, MD. Dr. O'Brien is now at the School of Medicine, Duke University, Durham, NC.
| |
Collapse
|
59
|
Maternal separation exacerbates Alzheimer's disease-like behavioral and pathological changes in adult APPswe/PS1dE9 mice. Behav Brain Res 2016; 318:18-23. [PMID: 27771383 DOI: 10.1016/j.bbr.2016.10.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 10/17/2016] [Accepted: 10/19/2016] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder that gradually destroys memory and cognitive abilities in the elderly, makes a huge emotional and economic burden on the patients and their families. The presence of senile plaques and the loss of cholinergic neurons in the brain are two neuropathological hallmarks of AD. Maternal separation (MS) is an animal paradigm designed to make early life stress. Studies on wild type rodents showed that MS could induce AD-like cognitive deficit and pathological changes. However, the effects of MS on AD susceptible population or AD animal models are still unclear. In the present study, male APPswe/PS1dE9 transgenic mice were separated from dam and pups 3h per day from postnatal day 2 to day 21. After weaning, all animals were housed under normal conditions (4 mice per cage). At 9-month age, MWM tests were performed to evaluate the learning and memory abilities. Then the pathological changes in the brain were measured by histology staining. The results showed MS mice had more severe deficit of learning and memory. Compared to the control, there were more senile plaques in cortex and hippocampus, fewer cholinergic neurons in nucleus basalis of Meynert in MS mice. These results indicate that MS exacerbates Alzheimer's disease-like behavioral and pathological changes in APPswe/PS1dE9 mice.
Collapse
|
60
|
Pietrzak RH, Laws SM, Lim YY, Bender SJ, Porter T, Doecke J, Ames D, Fowler C, Masters CL, Milicic L, Rainey-Smith S, Villemagne VL, Rowe CC, Martins RN, Maruff P. Plasma Cortisol, Brain Amyloid-β, and Cognitive Decline in Preclinical Alzheimer's Disease: A 6-Year Prospective Cohort Study. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2016; 2:45-52. [PMID: 29560886 DOI: 10.1016/j.bpsc.2016.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/17/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hypothalamic-pituitary-adrenal axis dysregulation, which is typically assessed by measuring cortisol levels, is associated with cognitive dysfunction, hippocampal atrophy, and increased risk for mild cognitive impairment and Alzheimer's disease (AD). However, little is known about the role of hypothalamic-pituitary-adrenal axis dysregulation in moderating the effect of high levels of amyloid-β (Aβ+) on cognitive decline in the preclinical phase of AD, which is often protracted, and thus offers opportunities for prevention and early intervention. METHODS Using data from a 6-year multicenter prospective cohort study, we evaluated the relation between Aβ level, plasma cortisol level, and cognitive decline in 416 cognitively normal older adults. RESULTS Results revealed that Aβ+ older adults experienced faster decline than Aβ- older adults in all cognitive domains (Cohen's d at 6-year assessment = 0.37-0.65). They further indicated a significant interaction between Aβ and cortisol levels for global cognition (d = 0.32), episodic memory (d = 0.50), and executive function (d = 0.59) scores, with Aβ+ older adults with high cortisol levels having significantly faster decline in these domains compared with Aβ+ older adults with low cortisol levels. These effects were independent of age, sex, APOE genotype, anxiety symptoms, and radiotracer type. CONCLUSIONS In cognitively healthy older adults, Aβ+ is associated with greater cognitive decline and high plasma cortisol levels may accelerate the effect of Aβ+ on decline in global cognition, episodic memory, and executive function. These results suggest that therapies targeted toward lowering plasma cortisol and Aβ levels may be helpful in mitigating cognitive decline in the preclinical phase of AD.
Collapse
Affiliation(s)
- Robert H Pietrzak
- U.S. Department of Veterans Affairs National Center for Posttraumatic Stress Disorder, Clinical Neurosciences Division, VA Connecticut Healthcare System, West Haven; Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut.
| | - Simon M Laws
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia; Co-operative Research Centre for Mental Health
| | - Yen Ying Lim
- The Florey Institute, The University of Melbourne, Parkville, Victoria
| | - Sophie J Bender
- School of Health Sciences, University of Notre Dame Australia, Fremantle, Western Australia
| | - Tenielle Porter
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia; Co-operative Research Centre for Mental Health
| | - James Doecke
- The Commonwealth Scientific and Industrial Research Organization, Canberra
| | - David Ames
- Academic Unit for Psychiatry of Old Age, St. Vincent's Health, Department of Psychiatry, The University of Melbourne, Kew; National Ageing Research Institute, Parkville, Victoria
| | | | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, Victoria
| | - Lidija Milicic
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia
| | - Stephanie Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia
| | - Victor L Villemagne
- The Florey Institute, The University of Melbourne, Parkville, Victoria; Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, Western Australia; Department of Nuclear Medicine and Centre for PET, Austin Health
| | - Christopher C Rowe
- Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, Western Australia; Department of Nuclear Medicine and Centre for PET, Austin Health
| | - Ralph N Martins
- Centre of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, Western Australia; Sir James McCusker Alzheimer's Disease Research Unit, Hollywood Private Hospital, Perth, Western Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, Victoria; Department of Medicine, Austin Health, The University of Melbourne, Heidelberg; Cogstate Ltd., Melbourne, Victoria, Australia
| | | |
Collapse
|
61
|
Toda S, Iguchi Y, Lin Z, Nishikawa H, Nagasawa T, Watanabe H, Minabe Y. Reconsidering Animal Models of Major Depressive Disorder in the Elderly. Front Aging Neurosci 2016; 8:188. [PMID: 27551264 PMCID: PMC4976092 DOI: 10.3389/fnagi.2016.00188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 07/20/2016] [Indexed: 11/15/2022] Open
Affiliation(s)
- Shigenobu Toda
- Department of Psychiatry and Neurobiology, Kanazawa UniversityKanazawa, Japan; Research Center for Child Mental Development, Kanazawa UniversityKanazawa, Japan; Hokuriku Dementia Professional Physician Training PlanKanazawa, Japan
| | - Yoshio Iguchi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University Fukushima, Japan
| | - Ziqiao Lin
- Department of Psychiatry and Neurobiology, Kanazawa University Kanazawa, Japan
| | - Hiromi Nishikawa
- Department of Psychiatry and Neurobiology, Kanazawa University Kanazawa, Japan
| | - Tatsuya Nagasawa
- Department of Psychiatry and Neurobiology, Kanazawa UniversityKanazawa, Japan; Hokuriku Dementia Professional Physician Training PlanKanazawa, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine Tokyo, Japan
| | - Yoshio Minabe
- Department of Psychiatry and Neurobiology, Kanazawa UniversityKanazawa, Japan; Research Center for Child Mental Development, Kanazawa UniversityKanazawa, Japan; Hokuriku Dementia Professional Physician Training PlanKanazawa, Japan
| |
Collapse
|
62
|
Nehls M. Unified theory of Alzheimer's disease (UTAD): implications for prevention and curative therapy. J Mol Psychiatry 2016; 4:3. [PMID: 27429752 PMCID: PMC4947325 DOI: 10.1186/s40303-016-0018-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022] Open
Abstract
The aim of this review is to propose a Unified Theory of Alzheimer's disease (UTAD) that integrates all key behavioural, genetic and environmental risk factors in a causal chain of etiological and pathogenetic events. It is based on three concepts that emanate from human's evolutionary history: (1) The grandmother-hypothesis (GMH), which explains human longevity due to an evolutionary advantage in reproduction by trans-generational transfer of acquired knowledge. Consequently it is argued that mental health at old-age must be the default pathway of humans' genetic program and not development of AD. (2) Therefore, mechanism like neuronal rejuvenation (NRJ) and adult hippocampal neurogenesis (AHN) that still function efficiently even at old age provide the required lifelong ability to memorize personal experiences important for survival. Cumulative evidence from a multitude of experimental and epidemiological studies indicate that behavioural and environmental risk factors, which impair productive AHN, result in reduced episodic memory performance and in reduced psychological resilience. This leads to avoidance of novelty, dysregulation of the hypothalamic-pituitary-adrenal (HPA)-axis and cortisol hypersecretion, which drives key pathogenic mechanisms of AD like the accumulation and oligomerization of synaptotoxic amyloid beta, chronic neuroinflammation and neuronal insulin resistance. (3) By applying to AHN the law of the minimum (LOM), which defines the basic requirements of biological growth processes, the UTAD explains why and how different lifestyle deficiencies initiate the AD process by impairing AHN and causing dysregulation of the HPA-axis, and how environmental and genetic risk factors such as toxins or ApoE4, respectively, turn into disease accelerators under these unnatural conditions. Consequently, the UTAD provides a rational strategy for the prevention of mental decline and a system-biological approach for the causal treatment of AD, which might even be curative if the systemic intervention is initiated early enough in the disease process. Hence an individualized system-biological treatment of patients with early AD is proposed as a test for the validity of UTAD and outlined in this review.
Collapse
Affiliation(s)
- Michael Nehls
- Independent Researcher, Allmendweg 1, 79279 Vörstetten, Germany
| |
Collapse
|
63
|
Tamano H, Yusuke E, Ide K, Takeda A. Influences of yokukansankachimpihange on aggressive behavior of zinc-deficient mice and actions of the ingredients on excessive neural exocytosis in the hippocampus of zinc-deficient rats. Exp Anim 2016; 65:353-361. [PMID: 27245118 PMCID: PMC5111838 DOI: 10.1538/expanim.16-0028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We examined the effect of Yokukansankachimpihange (YKSCH), a form of Yokukansan
containing parts of two herbaceous plants, Citrus Unshiu Peel (Chimpi) and Pinellia Tuber
(Hange), on aggressive behavior of mice housed individually. Mice were fed a
zinc-deficient diet for 2 weeks. In a resident-intruder test, the cumulative duration of
aggressive behavior was decreased in zinc-deficient mice administrated drinking water
containing YKSCH (approximately 300 mg/kg body weight/day) for 2 weeks. We tested mice for
geissoschizine methyl ether (GM), which is contained in Uncaria Hook, and
18β-glycyrrhetinic acid (GA), a major metabolite of glycyrrhizin contained in Glycyrrhiza,
which were contained in YKS and YKSCH. In hippocampal slices from zinc-deficient rats,
excess exocytosis at mossy fiber boutons induced with 60 mM KCl was attenuated in the
presence of GA (100–500 µM) or GM (100 µM). The
intracellular Ca2+ level, which showed an increase induced by 60 mM KCl, was
also attenuated in the presence of GA (100–500 µM) or GM (100
µM). These results suggest that GA and GM ameliorate excess glutamate
release from mossy fiber boutons by suppressing the increase in intracellular
Ca2+ signaling. These ameliorative actions may contribute to decreasing the
aggressiveness of mice individually housed under zinc deficiency, potentially by
suppressing excess glutamatergic neuron activity in the hippocampus.
Collapse
Affiliation(s)
- Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52- 1 Yada, Shizuoka 422-8526, Japan
| | | | | | | |
Collapse
|
64
|
Zhang C, Kuo CC, Moghadam SH, Monte L, Campbell SN, Rice KC, Sawchenko PE, Masliah E, Rissman RA. Corticotropin-releasing factor receptor-1 antagonism mitigates beta amyloid pathology and cognitive and synaptic deficits in a mouse model of Alzheimer's disease. Alzheimers Dement 2016; 12:527-37. [PMID: 26555315 PMCID: PMC4860182 DOI: 10.1016/j.jalz.2015.09.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 09/27/2015] [Accepted: 09/28/2015] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Stress and corticotropin-releasing factor (CRF) have been implicated as mechanistically involved in Alzheimer's disease (AD), but agents that impact CRF signaling have not been carefully tested for therapeutic efficacy or long-term safety in animal models. METHODS To test whether antagonism of the type-1 corticotropin-releasing factor receptor (CRFR1) could be used as a disease-modifying treatment for AD, we used a preclinical prevention paradigm and treated 30-day-old AD transgenic mice with the small-molecule, CRFR1-selective antagonist, R121919, for 5 months, and examined AD pathologic and behavioral end points. RESULTS R121919 significantly prevented the onset of cognitive impairment in female mice and reduced cellular and synaptic deficits and beta amyloid and C-terminal fragment-β levels in both genders. We observed no tolerability or toxicity issues in mice treated with R121919. DISCUSSION CRFR1 antagonism presents a viable disease-modifying therapy for AD, recommending its advancement to early-phase human safety trials.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Ching-Chang Kuo
- NeuroInformatics Center, University of Oregon, Eugene, OR, USA
| | - Setareh H Moghadam
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Louise Monte
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Shannon N Campbell
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Kenner C Rice
- Chemical Biology Research Branch, National Institute on Drug Abuse and Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | | | - Eliezer Masliah
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
65
|
Campbell SN, Zhang C, Roe AD, Lee N, Lao KU, Monte L, Donohue MC, Rissman RA. Impact of CRFR1 Ablation on Amyloid-β Production and Accumulation in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 45:1175-84. [PMID: 25697705 DOI: 10.3233/jad-142844] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stress exposure and the corticotropin-releasing factor (CRF) system have been implicated as mechanistically involved in both Alzheimer's disease (AD) and associated rodent models. In particular, the major stress receptor, CRF receptor type 1 (CRFR1), modulates cellular activity in many AD-relevant brain areas, and has been demonstrated to impact both tau phosphorylation and amyloid-β (Aβ) pathways. The overarching goal of our laboratory is to develop and characterize agents that impact the CRF signaling system as disease-modifying treatments for AD. In the present study, we developed a novel transgenic mouse to determine whether partial or complete ablation of CRFR1 was feasible in an AD transgenic model and whether this type of treatment could impact Aβ pathology. Double transgenic AD mice (PSAPP) were crossed to mice null for CRFR1; resultant CRFR1 heterozygous (PSAPP-R1(+/-)) and homozygous (PSAPP-R1(-/-)) female offspring were used at 12 months of age to examine the impact of CRFR1 disruption on the severity of AD Aβ levels and pathology. We found that both PSAPP-R1(+/-) and PSAPP-R1(-/-) had significantly reduced Aβ burden in the hippocampus, insular, rhinal, and retrosplenial cortices. Accordingly, we observed dramatic reductions in Aβ peptides and AβPP-CTFs, providing support for a direct relationship between CRFR1 and Aβ production pathways. In summary, our results suggest that interference of CRFR1 in an AD model is tolerable and is efficacious in impacting Aβ neuropathology.
Collapse
Affiliation(s)
- Shannon N Campbell
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Cheng Zhang
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Allyson D Roe
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Nickey Lee
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Kathleen U Lao
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Louise Monte
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Michael C Donohue
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA Department of Family Preventive Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
66
|
Koenig AM, DeLozier IJ, Zmuda MD, Marron MM, Begley AE, Anderson SJ, Reynolds CF, Arnold SE, Becker JT, Butters MA. Neuropsychological functioning in the acute and remitted States of late-life depression. J Alzheimers Dis 2016; 45:175-85. [PMID: 25471193 DOI: 10.3233/jad-148006] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Late-life depression (LLD, major depression occurring in an adult 60 years or older) is a common condition that frequently presents with cognitive impairment. Up to half of individuals with LLD are estimated to have cognitive impairment greater than that of age- and education-matched comparators, with impairments of episodic memory, speed of information processing, executive functioning, and visuospatial ability being most common. To inform our understanding of the state- versus trait-effects of depression on neuropsychological functioning, and to overcome limitations of previous studies, we utilized baseline data from the longitudinal Pathways study to compare differences in single time point performance on a broad-based neuropsychological battery across three diagnostic groups of older adults, each comprised of unique participants (n = 438): currently depressed (n = 120), previously depressed but currently euthymic (n = 190), and never-depressed (n = 128). Consistent with our hypotheses, we found that participants with a history of depression (currently or previously depressed) performed significantly worse than never-depressed participants on most tests of global cognition, as well as on tests of episodic memory, attention and processing speed, verbal ability, and visuospatial ability; in general, differences were most pronounced within the domain of attention and processing speed. Contrary to our hypothesis, we did not observe differences in executive performance between the two depression groups, suggesting that certain aspects of executive functioning are "trait deficits" associated with LLD. These findings are in general agreement with the existing literature, and represent an enhancement in methodological rigor over previous studies given the cross-sectional approach that avoids practice effects on test performance.
Collapse
Affiliation(s)
- Aaron M Koenig
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Isaac J DeLozier
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michelle D Zmuda
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Megan M Marron
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy E Begley
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stewart J Anderson
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles F Reynolds
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA Department of Behavioral and Community Health Sciences, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven E Arnold
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - James T Becker
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
67
|
Citalopram Ameliorates Synaptic Plasticity Deficits in Different Cognition-Associated Brain Regions Induced by Social Isolation in Middle-Aged Rats. Mol Neurobiol 2016; 54:1927-1938. [PMID: 26899575 DOI: 10.1007/s12035-016-9781-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 02/08/2016] [Indexed: 12/19/2022]
Abstract
Our previous experiments demonstrated that social isolation (SI) caused AD-like tau hyperphosphorylation and spatial memory deficits in middle-aged rats. However, the underlying mechanisms of SI-induced spatial memory deficits remain elusive. Middle-aged rats (10 months) were group or isolation reared for 8 weeks. Following the initial 4-week period of rearing, citalopram (10 mg/kg i.p.) was administered for 28 days. Then, pathophysiological changes were assessed by performing behavioral, biochemical, and pathological analyses. We found that SI could cause cognitive dysfunction and decrease synaptic protein (synaptophysin or PSD93) expression in different brain regions associated with cognition, such as the prefrontal cortex, dorsal hippocampus, ventral hippocampus, amygdala, and caudal putamen, but not in the entorhinal cortex or posterior cingulate. Citalopram could significantly improve learning and memory and partially restore synaptophysin or PSD93 expression in the prefrontal cortex, hippocampus, and amygdala in SI rats. Moreover, SI decreased the number of dendritic spines in the prefrontal cortex, dorsal hippocampus, and ventral hippocampus, which could be reversed by citalopram. Furthermore, SI reduced the levels of BDNF, serine-473-phosphorylated Akt (active form), and serine-9-phosphorylated GSK-3β (inactive form) with no significant changes in the levels of total GSK-3β and Akt in the dorsal hippocampus, but not in the posterior cingulate. Our results suggest that decreased synaptic plasticity in cognition-associated regions might contribute to SI-induced cognitive deficits, and citalopram could ameliorate these deficits by promoting synaptic plasticity mainly in the prefrontal cortex, dorsal hippocampus, and ventral hippocampus. The BDNF/Akt/GSK-3β pathway plays an important role in regulating synaptic plasticity in SI rats.
Collapse
|
68
|
Tamano H, Ide K, Adlard PA, Bush AI, Takeda A. Involvement of hippocampal excitability in amyloid β-induced behavioral and psychological symptoms of dementia. J Toxicol Sci 2016; 41:449-57. [DOI: 10.2131/jts.41.449] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Kazuki Ide
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| | - Paul Anthony Adlard
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, At Genetics Lane on Royal Parade, The University of Melbourne, Australia
| | - Ashley Ian Bush
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, At Genetics Lane on Royal Parade, The University of Melbourne, Australia
| | - Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka
| |
Collapse
|
69
|
van Dijk G, van Heijningen S, Reijne AC, Nyakas C, van der Zee EA, Eisel ULM. Integrative neurobiology of metabolic diseases, neuroinflammation, and neurodegeneration. Front Neurosci 2015; 9:173. [PMID: 26041981 PMCID: PMC4434977 DOI: 10.3389/fnins.2015.00173] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/28/2015] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a complex, multifactorial disease with a number of leading mechanisms, including neuroinflammation, processing of amyloid precursor protein (APP) to amyloid β peptide, tau protein hyperphosphorylation, relocalization, and deposition. These mechanisms are propagated by obesity, the metabolic syndrome and type-2 diabetes mellitus. Stress, sedentariness, dietary overconsumption of saturated fat and refined sugars, and circadian derangements/disturbed sleep contribute to obesity and related metabolic diseases, but also accelerate age-related damage and senescence that all feed the risk of developing AD too. The complex and interacting mechanisms are not yet completely understood and will require further analysis. Instead of investigating AD as a mono- or oligocausal disease we should address the disease by understanding the multiple underlying mechanisms and how these interact. Future research therefore might concentrate on integrating these by “systems biology” approaches, but also to regard them from an evolutionary medicine point of view. The current review addresses several of these interacting mechanisms in animal models and compares them with clinical data giving an overview about our current knowledge and puts them into an integrated framework.
Collapse
Affiliation(s)
- Gertjan van Dijk
- Department Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands
| | - Steffen van Heijningen
- Department Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands
| | - Aaffien C Reijne
- Department Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands ; Systems Biology Centre for Energy Metabolism and Ageing, University Medical Center, University of Groningen Groningen, Netherlands
| | - Csaba Nyakas
- Department Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands
| | - Eddy A van der Zee
- Department Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands
| | - Ulrich L M Eisel
- Department Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences, University of Groningen Groningen, Netherlands ; University Centre of Psychiatry, University Medical Center Groningen, University of Groningen Groningen, Netherlands
| |
Collapse
|
70
|
Daulatzai MA. “Boomerang Neuropathology” of Late-Onset Alzheimer’s Disease is Shrouded in Harmful “BDDS”: Breathing, Diet, Drinking, and Sleep During Aging. Neurotox Res 2015; 28:55-93. [PMID: 25911292 DOI: 10.1007/s12640-015-9528-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 04/03/2015] [Accepted: 04/03/2015] [Indexed: 12/12/2022]
|
71
|
Huang H, Wang L, Cao M, Marshall C, Gao J, Xiao N, Hu G, Xiao M. Isolation Housing Exacerbates Alzheimer's Disease-Like Pathophysiology in Aged APP/PS1 Mice. Int J Neuropsychopharmacol 2015; 18:pyu116. [PMID: 25568286 PMCID: PMC4540096 DOI: 10.1093/ijnp/pyu116] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/03/2014] [Accepted: 12/23/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative disease characterized by gradual declines in social, cognitive, and emotional functions, leading to a loss of expected social behavior. Social isolation has been shown to have adverse effects on individual development and growth as well as health and aging. Previous experiments have shown that social isolation causes an early onset of Alzheimer's disease-like phenotypes in young APP695/PS1-dE9 transgenic mice. However, the interactions between social isolation and Alzheimer's disease still remain unknown. METHODS Seventeen-month-old male APP695/PS1-dE9 transgenic mice were either singly housed or continued group housing for 3 months. Then, Alzheimer's disease-like pathophysiological changes were evaluated by using behavioral, biochemical, and pathological analyses. RESULTS Isolation housing further promoted cognitive dysfunction and Aβ plaque accumulation in the hippocampus of aged APP695/PS1-dE9 transgenic mice, associated with increased γ-secretase and decreased neprilysin expression. Furthermore, exacerbated hippocampal atrophy, synapse and myelin associated protein loss, and glial neuroinflammatory reactions were observed in the hippocampus of isolated aged APP695/PS1-dE9 transgenic mice. CONCLUSIONS The results demonstrate that social isolation exacerbates Alzheimer's disease-like pathophysiology in aged APP695/PS1-dE9 transgenic mice, highlighting the potential role of group life for delaying or counteracting the Alzheimer's disease process.
Collapse
Affiliation(s)
- Huang Huang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Linmei Wang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Min Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Charles Marshall
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Na Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD)
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, China (Drs Huang MD, Wang MD, Cao Ms, Gao Ms, N. Xiao Ms, Hu MD, PhD, and M. Xiao MD, PhD); Department of Rehabilitation Sciences, University of Kentucky Center for Excellence in Rural Health, Hazard, KY (Dr Marshall PhD).
| |
Collapse
|
72
|
Du X, Pang TY. Is Dysregulation of the HPA-Axis a Core Pathophysiology Mediating Co-Morbid Depression in Neurodegenerative Diseases? Front Psychiatry 2015; 6:32. [PMID: 25806005 PMCID: PMC4353372 DOI: 10.3389/fpsyt.2015.00032] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/16/2015] [Indexed: 01/19/2023] Open
Abstract
There is increasing evidence of prodromal manifestation of neuropsychiatric symptoms in a variety of neurodegenerative diseases such as Parkinson's disease (PD) and Huntington's disease (HD). These affective symptoms may be observed many years before the core diagnostic symptoms of the neurological condition. It is becoming more apparent that depression is a significant modifying factor of the trajectory of disease progression and even treatment outcomes. It is therefore crucial that we understand the potential pathophysiologies related to the primary condition, which could contribute to the development of depression. The hypothalamic-pituitary-adrenal (HPA)-axis is a key neuroendocrine signaling system involved in physiological homeostasis and stress response. Disturbances of this system lead to severe hormonal imbalances, and the majority of such patients also present with behavioral deficits and/or mood disorders. Dysregulation of the HPA-axis is also strongly implicated in the pathology of major depressive disorder. Consistent with this, antidepressant drugs, such as the selective serotonin reuptake inhibitors have been shown to alter HPA-axis activity. In this review, we will summarize the current state of knowledge regarding HPA-axis pathology in Alzheimer's, PD and HD, differentiating between prodromal and later stages of disease progression when evidence is available. Both clinical and preclinical evidence will be examined, but we highlight animal model studies as being particularly useful for uncovering novel mechanisms of pathology related to co-morbid mood disorders. Finally, we purpose utilizing the preclinical evidence to better inform prospective, intervention studies.
Collapse
Affiliation(s)
- Xin Du
- Mental Health Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne , Melbourne, VIC , Australia
| | - Terence Y Pang
- Behavioural Neurosciences Division, Florey Institute of Neuroscience and Mental Health, University of Melbourne , Melbourne, VIC , Australia
| |
Collapse
|
73
|
Popp J, Wolfsgruber S, Heuser I, Peters O, Hüll M, Schröder J, Möller HJ, Lewczuk P, Schneider A, Jahn H, Luckhaus C, Perneczky R, Frölich L, Wagner M, Maier W, Wiltfang J, Kornhuber J, Jessen F. Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer's type. Neurobiol Aging 2014; 36:601-7. [PMID: 25435336 DOI: 10.1016/j.neurobiolaging.2014.10.031] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 10/08/2014] [Accepted: 10/24/2014] [Indexed: 11/25/2022]
Abstract
Increased peripheral and central nervous system cortisol levels have been reported in Alzheimer's disease (AD) and may reflect dysfunction of cerebral components of the hypothalamic-pituitary-adrenal (HPA) axis. However, brain exposure to high cortisol concentrations may also accelerate disease progression and cognitive decline. The objectives of this study were to investigate whether HPA-axis dysregulation occurs at early clinical stages of AD and whether plasma and CSF cortisol levels are associated with clinical disease progression. Morning plasma and CSF cortisol concentrations were obtained from the subjects with AD dementia, mild cognitive impairment of AD type (MCI-AD), MCI of other type (MCI-O), and controls with normal cognition included in a multicenter study from the German Dementia Competence Network. A clinical and neuropsychological follow-up was performed in a subgroup of participants with MCI-AD, MCI-O, and AD dementia. CSF cortisol concentrations were increased in the subjects with AD dementia or MCI-AD compared with subjects with MCI-O or normal cognition. After controlling for possible confounders including CSF measures of amyloid beta1-42 and total tau, higher baseline CSF cortisol levels were associated with faster clinical worsening and cognitive decline in MCI-AD. The findings suggest that HPA-axis dysregulation occurs at the MCI stage of AD and may accelerate disease progression and cognitive decline.
Collapse
Affiliation(s)
- Julius Popp
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany; Department of Psychiatry, Leenaards Memory Center, University Hospital of Lausanne, Lausanne, Switzerland; Department of Clinical Neurosciences, Leenaards Memory Center, University Hospital of Lausanne, Lausanne, Switzerland.
| | - Steffen Wolfsgruber
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Isabella Heuser
- Department of Psychiatry and Psychotherapy, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Oliver Peters
- Department of Psychiatry and Psychotherapy, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Michael Hüll
- Center for Geriatric Medicine and Gerontology, University Hospital Freiburg, Freiburg, Germany
| | - Johannes Schröder
- Department of Psychiatry and Psychotherapy, University of Heidelberg, Heidelberg, Germany
| | - Hans-Jürgen Möller
- Department of Psychiatry, Ludwig-Maximilians, University München, München, Germany
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Schneider
- Department of Psychiatry and Psychotherapy, University of Göttingen, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Holger Jahn
- Department of Psychiatry and Psychotherapy, University of Hamburg, Hamburg, Germany
| | - Christian Luckhaus
- Department of Psychiatry and Psychotherapy, University of Düsseldorf, Düsseldorf, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, Technische Universität München, München, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, Mannheim, Germany
| | - Michael Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August-University, Göttingen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
74
|
Abstract
Two-thirds of individuals with Alzheimer's disease (AD) are women, owing largely to the fact that women outlive men (https://www.alz.org/downloads/facts_figures_2012.pdf). Women's increased longevity, however, is not sufficient to explain the fact that women are 1.5 times more likely than men to develop the disease (Gaoet al., 1998). After age 80, the incidence of AD is much higher in women than in men, such that the proportion of women with AD is almost twice the proportion of men with the disease (e.g., Zandiet al., 2002; Plassmanet al., 2007). Moreover, once diagnosed with AD, women decline more rapidly, both cognitively and functionally, compared to men (Itoet al., 2011; Tschanzet al., 2011).
Collapse
|
75
|
Abstract
This brief report provides an introduction to the topic of cognitive functioning in late-life depression (LLD). In addition to providing a review of the literature, we present a framework for understanding the heterogeneity of cognitive outcomes in this highly prevalent disorder. In addition, we discuss the relationship between LLD and dementia, and highlight the importance of regularly assessing cognitive functioning in older adults who present with depressive symptoms. If cognitive deficits are discovered during a neuropsychological assessment, we recommend referral to a geriatric psychiatrist or cognitive neurologist, for evaluation and treatment of the patient's symptoms.
Collapse
|
76
|
Gordon BA, Blazey T, Benzinger TLS, Head D. Effects of aging and Alzheimer's disease along the longitudinal axis of the hippocampus. J Alzheimers Dis 2014; 37:41-50. [PMID: 23780659 DOI: 10.3233/jad-130011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The hippocampus is often treated as a uniform structure, but possesses differential projections to surrounding cortex along its longitudinal axis. This heterogeneity could create varied susceptibility to pathological influences, potentially leading to non-uniform volumetric associations with advancing age and Alzheimer's disease (AD). Previous examinations of aging and AD effects on hippocampal subdivisions have produced highly discrepant findings. To clarify these inconsistencies, we examined the hippocampal head, body, and tail in a large sample of 292 cognitively normal, 37 very mildly demented, and 18 mildly demented individuals, divided into two independent samples. As often done in the literature, we characterized qualitative patterns across these regions, but extended these results by explicitly testing for quantitative differences. In each sample of cognitively normal individuals, the head and body demonstrated greater age effects than the tail. In each sample contrasting AD and cognitively normal individuals, all three regions showed significant volume reductions, with the greatest effect on the head. When examining increasing severity of dementia, the hippocampal head showed progressive volume loss, while the body and tail did not. The patterns of results examining both aging and AD were relatively consistent across the independent samples. These results indicate that there is an anterior-to-posterior gradient of loss within the hippocampus with both advancing age and AD.
Collapse
Affiliation(s)
- Brian A Gordon
- Department of Psychology, Washington University in St. Louis, St. Louis, MO 63130-4899, USA.
| | | | | | | |
Collapse
|
77
|
Bennett DA, Arnold SE, Valenzuela MJ, Brayne C, Schneider JA. Cognitive and social lifestyle: links with neuropathology and cognition in late life. Acta Neuropathol 2014; 127:137-50. [PMID: 24356982 PMCID: PMC4054865 DOI: 10.1007/s00401-013-1226-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/17/2013] [Accepted: 11/30/2013] [Indexed: 12/26/2022]
Abstract
Many studies report an association of cognitive and social experiential factors and related traits with dementia risk. Further, many clinical-pathologic studies find a poor correspondence between levels of neuropathology and the presence of dementia and level of cognitive impairment. The poor correspondence suggests that other factors contribute to the maintenance or loss of cognitive function, with factors associated with the maintenance of function referred to as neural or cognitive reserve. This has led investigators to examine the associations of cognitive and social experiential factors with neuropathology as a first step in disentangling the complex associations between these experiential risk factors, neuropathology, and cognitive impairment. Despite the consistent associations of a range of cognitive and social lifestyle factors with cognitive decline and dementia risk, the extant clinical-pathologic data find only a single factor from one cohort, linguistic ability, related to AD pathology. Other factors, including education, harm avoidance, and emotional neglect, are associated with cerebrovascular disease. Overall, the associations are weak. Some factors, such as education, social networks, and purpose in life, modify the relation of neuropathology to cognition. Finally, some factors such as cognitive activity appear to bypass known pathologies altogether suggesting a more direct association with biologic indices that promote person-specific differences in reserve and resilience. Future work will first need to replicate findings across more studies to ensure the veracity of the existing data. Second, effort is needed to identify the molecular substrates of neural reserve as potential mediators of the association of lifestyle factors with cognition.
Collapse
Affiliation(s)
- David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA,
| | | | | | | | | |
Collapse
|
78
|
Khalaj L, Chavoshi Nejad S, Mohammadi M, Sarraf Zadeh S, Hossein Pour M, Ahmadiani A, Khodagholi F, Ashabi G, Zeighamy Alamdary S, Samami E. Gemfibrozil pretreatment proved protection against acute restraint stress-induced changes in the male rats' hippocampus. Brain Res 2013; 1527:117-30. [DOI: 10.1016/j.brainres.2013.06.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/25/2013] [Accepted: 06/26/2013] [Indexed: 02/06/2023]
|
79
|
Assessing competence of broccoli consumption on inflammatory and antioxidant pathways in restraint-induced models: estimation in rat hippocampus and prefrontal cortex. BIOMED RESEARCH INTERNATIONAL 2013; 2013:590379. [PMID: 23936822 PMCID: PMC3725709 DOI: 10.1155/2013/590379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/03/2013] [Indexed: 01/23/2023]
Abstract
A growing body of evidence advocated the protective and therapeutic potential of natural compounds and phytochemicals used in diets against pathological conditions. Herein, the outcome of dietary whole broccoli consumption prior to restraint stress has been investigated in the hippocampus and prefrontal cortex of male rats, two important regions involved in the processing of responses to stressful events. Interestingly, a region-specific effect was detected regarding some of antioxidant defense system factors: nuclear factor erythroid-derived 2-related factor 2 (Nrf-2) antioxidant pathway, mitochondrial prosurvival proteins involved in mitochondrial biogenesis, and apoptotic cell death proteins. Dietary broccoli supplementation modulated the restraint-induced changes towards a consistent overall protection in the hippocampus. In the prefrontal cortex, however, despite activation of most of the protective factors, presumably as an attempt to save the system against the stress insult, some detrimental outcomes such as induced malate dehydrogenase (MDA) level and cleaved form of caspase-3 were detectable. Such diversity may be attributed in one hand to the different basic levels and/or availability of defensive mechanisms within the two studied cerebral regions, and on the other hand to the probable dose-dependent and hormetic effects of whole broccoli. More experiments are essential to demonstrate these assumptions.
Collapse
|
80
|
Dar-Nimrod I, Chapman BP, Franks P, Robbins J, Porsteinsson A, Mapstone M, Duberstein PR. Personality factors moderate the associations between apolipoprotein genotype and cognitive function as well as late onset Alzheimer disease. Am J Geriatr Psychiatry 2012; 20:1026-35. [PMID: 23079898 PMCID: PMC4184145 DOI: 10.1097/jgp.0b013e318267016b] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES We tested the hypothesis that neuroticism moderates the association between APOE (apolipoprotein E) genotype and two major outcomes, cognitive function and Alzheimer disease. We also explored whether other personality dimensions (extraversion, openness to experience, agreeableness, and conscientiousness) moderate the associations of APOE with these outcomes. DESIGN Primary analyses of existing randomized clinical trial data. SAMPLE Six-hundred two older adults (mean age of 78 years at baseline). MEASUREMENTS APOE genotype, the NEO-Five Factor Inventory, the Alzheimer's Disease Assessment Scale-Cognitive: measured every 6 months for 6.5 years) and relevant covariates. RESULTS Fully adjusted multivariate analyses showed that the association between the presence of APOE [Latin Small Letter Open E]-4 allele(s) and both outcomes was evident among individuals with high levels of neuroticism and extraversion but not among persons with low levels of these traits. CONCLUSIONS Phenotypic personality dimensions, primarily neuroticism and extraversion, moderate the relationship between APOE [Latin Small Letter Open E]-4 genotype and cognitive outcomes among older adults. Future research is needed to elucidate the physiological processes involved in these particular phenotype-genotype interactions.
Collapse
|
81
|
Dong H, Murphy KM, Meng L, Montalvo-Ortiz J, Zeng Z, Kolber BJ, Zhang S, Muglia LJ, Csernansky JG. Corticotrophin releasing factor accelerates neuropathology and cognitive decline in a mouse model of Alzheimer's disease. J Alzheimers Dis 2012; 28:579-92. [PMID: 22045495 DOI: 10.3233/jad-2011-111328] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic stress has been suggested to influence the pathogenesis of Alzheimer's disease (AD); however, the mechanism underlying this influence remains unknown. In this study, we created a triple transgenic mouse model that overexpresses corticotrophin-releasing factor (CRF) and human amyloid-β protein precursor (AβPP), to investigate whether increases in the expression of CRF can mimic the effects of stress on amyloid metabolism and the neurodegeneration. Tg2576 mice that overexpresses human AβPP gene were crossbreed with Tetop-CRF (CRF) mice and CaMKII-tTA (tTA) mice to create a novel triple transgenic mouse model that conditioned overexpresses CRF in forebrain and overexpresses human AβPP (called AβPP+/CRF+/tTA+, or TT mice). Then we evaluated serial neuro-anatomical and behavioral phenotypes on TT mice using histological, biochemical, and behavioral assays. TT mice showed a Cushingoid-like phenotype starting at 3 months of age. At 6 months of age, these mice demonstrated increases in tissue-soluble amyloid-β (Aβ) and Aβ plaques in the cortex and hippocampus, as compared to control mice. Moreover, TT mice characterized substantial decreases in dendritic branching and dendritic spine density in pyramidal neurons in layer 4 of the frontal cortex and CA1 of the hippocampus. Finally, TT mice showed significantly impaired working memory and contextual memory, with a modest increase in anxiety-like behavior. Our results suggested genetic increases in the brain of CRF expression mimicked chronic stress on the effects of amyloid deposition, neurodegeneration, and behavioral deficits. The novel transgenic mouse model will provide a unique tool to further investigate the mechanisms between stress and AD.
Collapse
Affiliation(s)
- Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Pomara N, Bruno D, Sarreal AS, Hernando RT, Nierenberg J, Petkova E, Sidtis JJ, Wisniewski TM, Mehta PD, Pratico D, Zetterberg H, Blennow K. Lower CSF amyloid beta peptides and higher F2-isoprostanes in cognitively intact elderly individuals with major depressive disorder. Am J Psychiatry 2012; 169:523-30. [PMID: 22764362 PMCID: PMC3586557 DOI: 10.1176/appi.ajp.2011.11081153] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Major depressive disorder is common in the elderly, and symptoms are often not responsive to conventional antidepressant treatment, especially in the long term. Soluble oligomeric and aggregated forms of amyloid beta peptides, especially amyloid beta 42, impair neuronal and synaptic function. Amyloid beta 42 is the main component of plaques and is implicated in Alzheimer's disease. Amyloid beta peptides also induce a depressive state in rodents and disrupt major neurotransmitter systems linked to depression. The authors assessed whether major depression was associated with CSF levels of amyloid beta, tau protein, and F2-isoprostanes in elderly individuals with major depressive disorder and age-matched nondepressed comparison subjects. METHOD CSF was obtained from 47 cognitively intact volunteers (major depression group, N=28; comparison group, N=19) and analyzed for levels of soluble amyloid beta, total and phosphorylated tau proteins, and isoprostanes. RESULTS Amyloid beta 42 levels were significantly lower in the major depression group relative to the comparison group, and amyloid beta 40 levels were lower but only approaching statistical significance. In contrast, isoprostane levels were higher in the major depression group. No differences were observed in total and phosphorylated tau proteins across conditions. Antidepressant use was not associated with differences in amyloid beta 42 levels. CONCLUSIONS Reduction in CSF levels of amyloid beta 42 may be related to increased brain amyloid beta plaques or decreased soluble amyloid beta production in elderly individuals with major depression relative to nondepressed comparison subjects. These results may have implications for our understanding of the pathophysiology of major depression and for the development of treatment strategies.
Collapse
|
83
|
Martinho A, Gonçalves I, Costa M, Santos CR. Stress and Glucocorticoids Increase Transthyretin Expression in Rat Choroid Plexus via Mineralocorticoid and Glucocorticoid Receptors. J Mol Neurosci 2012; 48:1-13. [DOI: 10.1007/s12031-012-9715-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 01/27/2012] [Indexed: 01/18/2023]
|
84
|
Restraint stress in rats alters gene transcription and protein translation in the hippocampus. Neurochem Res 2012; 37:958-64. [PMID: 22219132 DOI: 10.1007/s11064-011-0688-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 12/18/2011] [Accepted: 12/23/2011] [Indexed: 10/14/2022]
Abstract
Stress is a relatively new and emerging risk factor for Alzheimer's disease (AD). Severe stress can alter brain characteristics such as neuronal plasticity, due to changes in the metabolism of cytoskeletal proteins. In this study, male Wistar rats were exposed to restraint stress (RS) for 5 h daily for different time periods. At the end of the exposure periods, the amounts of β-actin, cofilin, amyloid precursor protein (APP) and mitogen-activated protein kinase 1 (MAPK-1) RNAs and proteins were investigated. The mRNA expressions of β-actin, cofilin and MAPK-1 followed U-shaped time course. Acute (3 days) and chronic (21 days) RS caused a fourfold and tenfold increases, respectively, in hippocampal β-actin mRNA expression. In the case of cofilin mRNA expression, elevations were detected in the hippocampus on days 3, 7 and 21. The APP mRNA level was increased on day 21. On protein level, chronic stress elevated the levels of β-actin, cofilin and APP in the hippocampus. These results suggest that stress causes the induction of some genes and proteins that are also elevated in AD selectively in the hippocampal region of the rat brain.
Collapse
|
85
|
Sawyer K, Corsentino E, Sachs-Ericsson N, Steffens DC. Depression, hippocampal volume changes, and cognitive decline in a clinical sample of older depressed outpatients and non-depressed controls. Aging Ment Health 2012; 16:753-62. [PMID: 22548411 PMCID: PMC3430833 DOI: 10.1080/13607863.2012.678478] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The aim of this study was to develop and test a model of depression, hippocampal changes, and cognitive decline. METHOD Participants were 248 community-dwelling, depressed patients and 147 healthy, non-depressed individuals 60 years and older. Participants received a structured interview assessing current depressive symptoms and past depressive episodes, completed cognitive testing with the Mini Mental State Examination (MMSE), and underwent structural Magnetic Resonance Imaging (MRI) of the brain. For up to 10 years, assessment of depressive symptoms and MMSE administration was repeated at least annually, and MRI was repeated every two years. RESULTS Regression analyses demonstrated that depression diagnosis at baseline predicted decrease in right (but not left) hippocampal volume over a four-year period. Analyses using structural equation modeling demonstrated that a decrease in left and right hippocampal volumes predicted decrease in MMSE score over four years. CONCLUSION Results provide some evidence for relationships between depression and decrease in right hippocampal volume, and between hippocampal volume and MMSE score. This would be consistent with depression as a causal factor in subsequent cognitive decline. Plausible biological mechanisms include a glucocorticoid cascade or a facilitating effect of depression on amyloid-beta plaque formation. Future studies should examine the relationship between hippocampal volume and specialized memory measures, as well as between depression diagnosis and volume of other brain structures.
Collapse
Affiliation(s)
| | | | | | - David C. Steffens
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center
| |
Collapse
|
86
|
Andrus BM, Blizinsky K, Vedell PT, Dennis K, Shukla PK, Schaffer DJ, Radulovic J, Churchill GA, Redei EE. Gene expression patterns in the hippocampus and amygdala of endogenous depression and chronic stress models. Mol Psychiatry 2012; 17:49-61. [PMID: 21079605 PMCID: PMC3117129 DOI: 10.1038/mp.2010.119] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2010] [Revised: 10/05/2010] [Accepted: 10/11/2010] [Indexed: 12/24/2022]
Abstract
The etiology of depression is still poorly understood, but two major causative hypotheses have been put forth: the monoamine deficiency and the stress hypotheses of depression. We evaluate these hypotheses using animal models of endogenous depression and chronic stress. The endogenously depressed rat and its control strain were developed by bidirectional selective breeding from the Wistar-Kyoto (WKY) rat, an accepted model of major depressive disorder (MDD). The WKY More Immobile (WMI) substrain shows high immobility/despair-like behavior in the forced swim test (FST), while the control substrain, WKY Less Immobile (WLI), shows no depressive behavior in the FST. Chronic stress responses were investigated by using Brown Norway, Fischer 344, Lewis and WKY, genetically and behaviorally distinct strains of rats. Animals were either not stressed (NS) or exposed to chronic restraint stress (CRS). Genome-wide microarray analyses identified differentially expressed genes in hippocampi and amygdalae of the endogenous depression and the chronic stress models. No significant difference was observed in the expression of monoaminergic transmission-related genes in either model. Furthermore, very few genes showed overlapping changes in the WMI vs WLI and CRS vs NS comparisons, strongly suggesting divergence between endogenous depressive behavior- and chronic stress-related molecular mechanisms. Taken together, these results posit that although chronic stress may induce depressive behavior, its molecular underpinnings differ from those of endogenous depression in animals and possibly in humans, suggesting the need for different treatments. The identification of novel endogenous depression-related and chronic stress response genes suggests that unexplored molecular mechanisms could be targeted for the development of novel therapeutic agents.
Collapse
Affiliation(s)
- B M Andrus
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - K Blizinsky
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - P T Vedell
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - K Dennis
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - P K Shukla
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - D J Schaffer
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - J Radulovic
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - E E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
87
|
Chronic stress exacerbates tau pathology, neurodegeneration, and cognitive performance through a corticotropin-releasing factor receptor-dependent mechanism in a transgenic mouse model of tauopathy. J Neurosci 2011; 31:14436-49. [PMID: 21976528 DOI: 10.1523/jneurosci.3836-11.2011] [Citation(s) in RCA: 190] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Because overactivation of the hypothalamic-pituitary-adrenal (HPA) axis occurs in Alzheimer's disease (AD), dysregulation of stress neuromediators may play a mechanistic role in the pathophysiology of AD. However, the effects of stress on tau phosphorylation are poorly understood, and the relationship between corticosterone and corticotropin-releasing factor (CRF) on both β-amyloid (Aβ) and tau pathology remain unclear. Therefore, we first established a model of chronic stress, which exacerbates Aβ accumulation in Tg2576 mice and then extended this stress paradigm to a tau transgenic mouse model with the P301S mutation (PS19) that displays tau hyperphosphorylation, insoluble tau inclusions and neurodegeneration. We show for the first time that both Tg2576 and PS19 mice demonstrate a heightened HPA stress profile in the unstressed state. In Tg2576 mice, 1 month of restraint/isolation (RI) stress increased Aβ levels, suppressed microglial activation, and worsened spatial and fear memory compared with nonstressed mice. In PS19 mice, RI stress promoted tau hyperphosphorylation, insoluble tau aggregation, neurodegeneration, and fear-memory impairments. These effects were not mimicked by chronic corticosterone administration but were prevented by pre-stress administration of a CRF receptor type 1 (CRF(1)) antagonist. The role for a CRF(1)-dependent mechanism was further supported by the finding that mice overexpressing CRF had increased hyperphosphorylated tau compared with wild-type littermates. Together, these results implicate HPA dysregulation in AD neuropathogenesis and suggest that prolonged stress may increase Aβ and tau hyperphosphorylation. These studies also implicate CRF in AD pathophysiology and suggest that pharmacological manipulation of this neuropeptide may be a potential therapeutic strategy for AD.
Collapse
|
88
|
Takeda A, Tamano H. Zinc signaling through glucocorticoid and glutamate signaling in stressful circumstances. J Neurosci Res 2011; 88:3002-10. [PMID: 20568287 DOI: 10.1002/jnr.22456] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Humans and animals are constantly exposed to environmental stress. The hypothalamic-pituitary-adrenal (HPA) axis responds to stress, followed by glucocorticoid secretion from the adrenal glands. This response serves to maintain homeostasis in the living body through energy mobilization or to restore it. The brain is an important target for glucocorticoids. The hippocampus participates in the regulation of the HPA axis. Stress activates glutamatergic neurons in the hippocampus, and serious stress induces dyshomeostasis of extracellular glutamate. This dyshomeostasis, which is potentiated by glucocorticoids, modifies cognitive and emotional behavior. On the other hand, zinc is necessary for glucocorticoid signaling and is released from glutamatergic (zincergic) neurons to modulate synaptic glutamate signaling. Stress also induces dyshomeostasis of extracellular zinc, which may be linked to dyshomeostasis of extracellular glutamate. Thus, glucocorticoid signaling might also contribute to dyshomeostasis of extracellular zinc. It is likely that zinc signaling participates in cognitive and emotional behavior through glucocorticoid and glutamate signaling under stressful circumstances. This Mini-Review analyzes the relationship among signals of glucocorticoid, glutamate, and zinc under stressful circumstances to elucidate the significance of the zinc signaling in response to stress.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| | | |
Collapse
|
89
|
Butterworth P, Cherbuin N, Sachdev P, Anstey KJ. The association between financial hardship and amygdala and hippocampal volumes: results from the PATH through life project. Soc Cogn Affect Neurosci 2011; 7:548-56. [PMID: 21551226 DOI: 10.1093/scan/nsr027] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study examined whether middle-aged adults exposed to poverty in childhood or current financial hardship have detectable brain differences from those who have not experienced such adversity. Structural magnetic resonance imaging (MRI) was conducted as one aspect of the Personality and Total Health (PATH) through life study: a large longitudinal community survey measuring the health and well-being of three cohorts from south-eastern Australia. This analysis considers data from 431 middle-aged adults in the aged 44-48 years at the time of the interview. Volumetric segmentation was performed with the Freesurfer image analysis suite. Data on socio-demographic circumstances, mental health and cognitive performance were collected through the survey interview. Results showed that, after controlling for well-established risk factors for atrophy, adults who reported financial hardship had smaller left and right hippocampal and amygdalar volumes than those who did not report hardship. In contrast, there was no reliable association between hardship and intra-cranial volume or between childhood poverty and any of the volumetric measures. Financial hardship may be considered a potent stressor and the observed results are consistent with the view that hardship influences hippocampal and amygdalar volumes through hypothalamic-pituitary-adrenal axis function and other stress-related pathways.
Collapse
Affiliation(s)
- Peter Butterworth
- Centre for Mental Health Research, Building 63, The Australian National University, Canberra ACT 0200, Australia.
| | | | | | | |
Collapse
|
90
|
Jarvis CI, Goncalves MB, Clarke E, Dogruel M, Kalindjian SB, Thomas SA, Maden M, Corcoran JPT. Retinoic acid receptor-α signalling antagonizes both intracellular and extracellular amyloid-β production and prevents neuronal cell death caused by amyloid-β. Eur J Neurosci 2011; 32:1246-55. [PMID: 20950278 PMCID: PMC3003897 DOI: 10.1111/j.1460-9568.2010.07426.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer’s disease (AD) is characterized by amyloid-β (Aβ) deposition in the brain, neuronal cell loss and cognitive decline. We show here that retinoic acid receptor (RAR)α signalling in vitro can prevent both intracellular and extracellular Aβ accumulation. RARα signalling increases the expression of a disintegrin and metalloprotease 10, an α-secretase that processes the amyloid precursor protein into the non-amyloidic pathway, thus reducing Aβ production. We also show that RARα agonists are neuroprotective, as they prevent Aβ-induced neuronal cell death in cortical cultures. If RARα agonists are given to the Tg2576 mouse, the normal Aβ production in their brains is suppressed. In contrast, neither RARβ nor γ-agonists affect Aβ production or Aβ-mediated neuronal cell death. Therefore, RARα agonists have therapeutic potential for the treatment of AD.
Collapse
Affiliation(s)
- C I Jarvis
- The Wolfson Centre For Age-Related Diseases, King's College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Involvement of 5-lipoxygenase in the corticosteroid-dependent amyloid beta formation: in vitro and in vivo evidence. PLoS One 2011; 6:e15163. [PMID: 21253592 PMCID: PMC3017076 DOI: 10.1371/journal.pone.0015163] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 10/27/2010] [Indexed: 11/19/2022] Open
Abstract
Background Numerous studies show that high circulating level of glucocorticosteroids is a biochemical characteristic of Alzheimer's disease (AD). These stress hormones can increase the amount of AD-like pathology in animal models of the disease. Since they also up-regulate the 5-Lipoxygenase (5-LO), an enzyme which modulates amyloid beta (Aβ) formation, in the present paper we tested the hypothesis that this enzymatic pathway is involved in the glucocorticoid-induced pro-amyloidotic effect. Methodology/Principal Findings Incubation of neuronal cells with dexamethasone resulted in a significant increase in 5-LO activity and Aβ formation. By contrast, pharmacological inhibition of 5-LO prevented the dexamethasone-dependent increase in Aβ levels. Mouse embryonic fibroblasts responded with a significant increase in Aβ formation after dexamethasone challenge. However, this effect was abolished when dexamethasone was incubated with fibroblasts genetically deficient for 5-LO. No difference in the glucocorticoid receptor levels was observed between the two groups. Finally, treatment of wild type mice with dexamethasone resulted in a significant increase in endogenous brain Aβ levels, which was prevented in mice genetically lacking 5-LO. Conclusions These findings suggest that 5-LO plays a functional role in the glucocorticoid-induced brain AD-like amyloid pathology.
Collapse
|
92
|
Takeda A. Insight into glutamate excitotoxicity from synaptic zinc homeostasis. Int J Alzheimers Dis 2010; 2011:491597. [PMID: 21234391 PMCID: PMC3017909 DOI: 10.4061/2011/491597] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 11/05/2010] [Indexed: 02/06/2023] Open
Abstract
Zinc is released from glutamatergic (zincergic) neuron terminals in the hippocampus, followed by the increase in Zn(2+) concentration in the intracellular (cytosol) compartment, as well as that in the extracellular compartment. The increase in Zn(2+) concentration in the intracellular compartment during synaptic excitation is mainly due to Zn(2+) influx through calcium-permeable channels and serves as Zn(2+) signaling as well as the case in the extracellular compartment. Synaptic Zn(2+) homeostasis is important for glutamate signaling and altered under numerous pathological processes such as Alzheimer's disease. Synaptic Zn(2+) homeostasis might be altered in old age, and this alteration might be involved in the pathogenesis and progression of Alzheimer's disease; Zinc may play as a key-mediating factor in the pathophysiology of Alzheimer's disease. This paper summarizes the role of Zn(2+) signaling in glutamate excitotoxicity, which is involved in Alzheimer's disease, to understand the significance of synaptic Zn(2+) homeostasis in the pathophysiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Global COE, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
93
|
Pomara N, Sidtis JJ. Brain neurotoxic amyloid-beta peptides: their potential role in the pathophysiology of depression and as molecular therapeutic targets. Br J Pharmacol 2010; 161:768-70. [PMID: 21105218 DOI: 10.1111/j.1476-5381.2010.00948.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The monoamine hypothesis ascribes an important role to the under activity of brain monoamines such as 5-HT, noradrenaline and dopamine to the pathophysiology of depression. This view emerged more than 50 years ago and has guided development of most medications currently used for the treatment of this disorder. However, large numbers of depressed individuals treated with currently available antidepressant agents, or even with various combinations, do not respond. Residual symptoms, relapses and recurrences are common while receiving adequate doses of these medications. In a recent issue of the BJP, Colaianna et al.describe results suggesting that a new neurobiological mechanism with treatment implications should be considered for the development of depression in humans, namely, elevations in potentially neurotoxic brain amyloid-ß peptides.
Collapse
Affiliation(s)
- Nunzio Pomara
- The Geriatric Psychiatry Program, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | | |
Collapse
|
94
|
Ameliorative effect of Yokukansan on social isolation-induced aggressive behavior of zinc-deficient young mice. Brain Res Bull 2010; 83:351-5. [PMID: 20813168 DOI: 10.1016/j.brainresbull.2010.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 08/05/2010] [Accepted: 08/23/2010] [Indexed: 12/20/2022]
Abstract
Yokukansan, a traditional Japanese medicine has been used to cure neuropsychological disorders. In the present study, the effect of Yokukansan on social isolation-induced aggressive behavior was examined in zinc-deficient mice, which were fed a zinc-deficient diet and a drinking water containing Yokukansan for 2 weeks. In the resident-intruder test, the rate of mice that exhibited aggressive behavior in zinc-deficient mice, which was significantly higher than that in the control mice, was significantly decreased by administration of Yokukansan. The basal level of serum glucocorticoid, which was significantly higher in zinc-deficient mice, was lowered by administration of Yokukansan. On the other hand, serum glucocorticoid levels after the resident-intruder test were almost the same between the control and zinc-deficient mice. However, administration of Yokukansan to zinc-deficient mice significantly increased serum glucocorticoid level after the resident-intruder test and the significant difference in the rate of serum corticosterone level after the test to the basal level between the control and zinc-deficient mice was abolished. Dietary zinc deficiency increases the basal levels of serum glucocorticoid, while may insufficiently increase serum glucocorticoid levels in the resident-intruder test. The concentrations of glutamate and GABA (γ-aminobutyric acid) in the brain were significantly higher in zinc-deficient mice, while Yokukansan ameliorated the significant increases. These results indicate that Yokukansan ameliorates social isolation-induced aggressive behavior of zinc-deficient mice, probably via amelioration of abnormal glucocorticoid secretion. The ameliorative effect seems to be linked to the modification of glutamatergic neuron activity after administration of Yokukansan.
Collapse
|
95
|
|
96
|
Impact of the Hypothalamic–pituitary–adrenal/gonadal Axes on Trajectory of Age-Related Cognitive Decline. PROGRESS IN BRAIN RESEARCH 2010; 182:31-76. [DOI: 10.1016/s0079-6123(10)82002-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|