51
|
Assessment of diets containing curcumin, epigallocatechin-3-gallate, docosahexaenoic acid and α-lipoic acid on amyloid load and inflammation in a male transgenic mouse model of Alzheimer's disease: Are combinations more effective? Neurobiol Dis 2019; 124:505-519. [DOI: 10.1016/j.nbd.2018.11.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/25/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
|
52
|
Buckley RF, Mormino EC, Chhatwal J, Schultz AP, Rabin JS, Rentz DM, Acar D, Properzi MJ, Dumurgier J, Jacobs H, Gomez-Isla T, Johnson KA, Sperling RA, Hanseeuw BJ. Associations between baseline amyloid, sex, and APOE on subsequent tau accumulation in cerebrospinal fluid. Neurobiol Aging 2019; 78:178-185. [PMID: 30947113 DOI: 10.1016/j.neurobiolaging.2019.02.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 01/01/2023]
Abstract
We investigated the effect of baseline Aβ, sex, and APOE on longitudinal tau accumulation in cerebrospinal fluid (CSF) in clinically normal older adults. Two hundred thirty-nine participants (aged 56-89 years, clinical dementia rating = 0) underwent serial CSF collection for Aβ1-42, total-tau (t-tau) and phospho-tau181P (p-tau). We used preprocessed data from fully automated Roche Elecsys immunoassays. A series of linear regressions were used to examine cross-sectional effects of Aβ1-42, sex, and APOEε4 on baseline CSF tau and linear mixed models for longitudinal changes in CSF tau. Cross-sectionally, CSF t-tau and p-tau were associated with abnormal Aβ1-42 and APOEε4 but not with sex. Longitudinally, low baseline CSF Aβ1-42 levels, but not APOEε4 or sex, predicted faster p-tau accumulation. The relationship between baseline CSF Aβ1-42 and tau accumulation was strongest in APOEε4 carriers, and particularly female carriers, relative to other groups. The current findings support an association between baseline CSF Aβ1-42 and changes in CSF tau. Elevated risk in females, apparent only in carriers, reinforces findings of sex-related vulnerability in those with genetic predisposition for Alzheimer's disease.
Collapse
Affiliation(s)
- Rachel F Buckley
- Florey Institutes of Neuroscience and Mental Health, Melbourne, Victoria, Australia; Melbourne School of Psychological Science, University of Melbourne, Victoria, Australia; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Santa Clara County, CA, USA
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Aaron P Schultz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer S Rabin
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Dorene M Rentz
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Diler Acar
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael J Properzi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Julien Dumurgier
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Heidi Jacobs
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gomez-Isla
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
| | - Keith A Johnson
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Reisa A Sperling
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Bernard J Hanseeuw
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Gordon Center for Medical Imaging, Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | | |
Collapse
|
53
|
Höfling C, Shehabi E, Kuhn PH, Lichtenthaler SF, Hartlage-Rübsamen M, Roßner S. Cell Type-Specific Human APP Transgene Expression by Hippocampal Interneurons in the Tg2576 Mouse Model of Alzheimer's Disease. Front Neurosci 2019; 13:137. [PMID: 30853883 PMCID: PMC6395433 DOI: 10.3389/fnins.2019.00137] [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] [Received: 12/11/2018] [Accepted: 02/06/2019] [Indexed: 01/21/2023] Open
Abstract
Amyloid precursor protein (APP) transgenic animal models of Alzheimer’s disease have become versatile tools for basic and translational research. However, there is great heterogeneity of histological, biochemical, and functional data between transgenic mouse lines, which might be due to different transgene expression patterns. Here, the expression of human APP (hAPP) by GABAergic hippocampal interneurons immunoreactive for the calcium binding proteins parvalbumin, calbindin, calretinin, and for the peptide hormone somatostatin was analyzed in Tg2576 mice by double immunofluorescent microscopy. Overall, there was no GABAergic interneuron subpopulation that did not express the transgene. On the other hand, in no case all neurons of such a subpopulation expressed hAPP. In dentate gyrus molecular layer and in stratum lacunosum moleculare less than 10% of hAPP-positive interneurons co-express any of these interneuron markers, whereas in stratum oriens hAPP-expressing neurons frequently co-express these interneuron markers to different proportions. We conclude that these neurons differentially contribute to deficits in young Tg2576 mice before the onset of Abeta plaque pathology. The detailed analysis of distinct brain region and neuron type-specific APP transgene expression patterns is indispensable to understand particular pathological features and mouse line-specific differences in neuronal and systemic functions.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Emira Shehabi
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| | - Peer-Hendrik Kuhn
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany.,Institute for Advanced Study, Technical University of Munich, Garching, Germany
| | | | - Steffen Roßner
- Paul-Flechsig-Institute for Brain Research, Leipzig University, Leipzig, Germany
| |
Collapse
|
54
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
55
|
Nyarko JNK, Quartey MO, Baker GB, Mousseau DD. Can Animal Models Inform on the Relationship between Depression and Alzheimer Disease? CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2019; 64:18-29. [PMID: 29685068 PMCID: PMC6364140 DOI: 10.1177/0706743718772514] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The focus on the β-amyloid (Aβ) peptide in clinical Alzheimer disease (AD) as well as in animal models of AD has perhaps biased our understanding of what contributes to the heterogeneity in disease onset and progression. Part of this heterogeneity could reflect the various neuropsychiatric risk factors that present with common symptomatology and can predispose the brain to AD-like changes. One such risk factor is depression. Animal models, particularly mouse models carrying variants of AD-related gene(s), many of which lead to an accumulation of Aβ, suggest that a fundamental shift in depression-related monoaminergic systems (including serotonin and noradrenaline) is a strong indicator of the altered cellular function associated with the earlier(est) stages of AD-related pathology. These changes in monoaminergic neurochemistry could provide for relevant targets for intervention in clinical AD and/or could support a polypharmacy strategy, which might include the targeting of Aβ, in vulnerable populations. Future studies must also include female mice as well as male mice in animal model studies on the relationship between depression and AD.
Collapse
Affiliation(s)
- Jennifer N K Nyarko
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Maa O Quartey
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Glen B Baker
- 2 Department of Psychiatry, Neuroscience and Mental Health Institute, Neurochemical Research Unit, University of Alberta, Edmonton, Alberta, Canada
| | - Darrell D Mousseau
- 1 Cell Signalling Laboratory, Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
56
|
Buckley RF, Mormino EC, Amariglio RE, Properzi MJ, Rabin JS, Lim YY, Papp KV, Jacobs HIL, Burnham S, Hanseeuw BJ, Doré V, Dobson A, Masters CL, Waller M, Rowe CC, Maruff P, Donohue MC, Rentz DM, Kirn D, Hedden T, Chhatwal J, Schultz AP, Johnson KA, Villemagne VL, Sperling RA. Sex, amyloid, and APOE ε4 and risk of cognitive decline in preclinical Alzheimer's disease: Findings from three well-characterized cohorts. Alzheimers Dement 2018; 14:1193-1203. [PMID: 29803541 PMCID: PMC6131023 DOI: 10.1016/j.jalz.2018.04.010] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/19/2018] [Accepted: 04/10/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Our objective was to investigate the effect of sex on cognitive decline within the context of amyloid β (Aβ) burden and apolipoprotein E genotype. METHODS We analyzed sex-specific effects on Aβ-positron emission tomography, apolipoprotein, and rates of change on the Preclinical Alzheimer Cognitive Composite-5 across three cohorts, such as the Alzheimer's Disease Neuroimaging Initiative, Australian Imaging, Biomarker and Lifestyle, and Harvard Aging Brain Study (n = 755; clinical dementia rating = 0; age (standard deviation) = 73.6 (6.5); female = 55%). Mixed-effects models of cognitive change by sex, Aβ-positron emission tomography, and apolipoprotein ε4 were examined with quadratic time effects over a median of 4 years of follow-up. RESULTS Apolipoprotein ε4 prevalence and Aβ burden did not differ by sex. Sex did not directly influence cognitive decline. Females with higher Aβ exhibited faster decline than males. Post hoc contrasts suggested that females who were Aβ and apolipoprotein ε4 positive declined faster than their male counterparts. DISCUSSION Although Aβ did not differ by sex, cognitive decline was greater in females with higher Aβ. Our findings suggest that sex may play a modifying role on risk of Alzheimer's disease-related cognitive decline.
Collapse
Affiliation(s)
- Rachel F. Buckley
- The Florey Institute, The University of Melbourne, Victoria, Australia
- Melbourne School of Psychological Science, University of Melbourne, Victoria, Australia
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - Rebecca E. Amariglio
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Michael J. Properzi
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jennifer S. Rabin
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yen Ying Lim
- The Florey Institute, The University of Melbourne, Victoria, Australia
| | - Kathryn V. Papp
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Heidi I. L. Jacobs
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, The Netherlands
| | - Samantha Burnham
- The Australian eHealth Research Centre, CSIRO Health & Biosecurity, Victoria, Australia
| | - Bernard J. Hanseeuw
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Neurology, Cliniques Universitaires Saint-Luc, Institute of Neuroscience, Université Catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Vincent Doré
- The Australian eHealth Research Centre, CSIRO Health & Biosecurity, Queensland, Australia
| | - Annette Dobson
- The University of Queensland, School of Public Health, Faculty of Medicine, Queensland, Australia
| | - Colin L. Masters
- The Florey Institute, The University of Melbourne, Victoria, Australia
| | - Michael Waller
- The University of Queensland, School of Public Health, Faculty of Medicine, Queensland, Australia
| | - Christopher C. Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Victoria, Australia
- The Department of Medicine, Austin Health, The University of Melbourne, Victoria, Australia
| | | | - Michael C. Donohue
- Department of Neurology, University of Southern California, San Diego, California, USA Words: 4122
| | - Dorene M. Rentz
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Dylan Kirn
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Trey Hedden
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Jasmeer Chhatwal
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aaron P. Schultz
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Keith A. Johnson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School, Boston, MA
| | - Victor L. Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Victoria, Australia
- The Department of Medicine, Austin Health, The University of Melbourne, Victoria, Australia
| | - Reisa A. Sperling
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | - the Australian Imaging, Biomarker and Lifestyle study of ageing
- Corresponding author: Rachel F. Buckley, PhD, Address: Department of Neurology, Level 10, Athinoula A. Martinos Center for Biomedical Imaging, 149 13th St, Charlestown, MA, USA 02129,
| | | |
Collapse
|
57
|
Precision medicine and drug development in Alzheimer's disease: the importance of sexual dimorphism and patient stratification. Front Neuroendocrinol 2018; 50:31-51. [PMID: 29902481 DOI: 10.1016/j.yfrne.2018.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/29/2018] [Accepted: 06/07/2018] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (ND) are among the leading causes of disability and mortality. Considerable sex differences exist in the occurrence of the various manifestations leading to cognitive decline. Alzheimer's disease (AD) exhibits substantial sexual dimorphisms and disproportionately affects women. Women have a higher life expectancy compared to men and, consequently, have more lifespan to develop AD. The emerging precision medicine and pharmacology concepts - taking into account the individual genetic and biological variability relevant for disease risk, prevention, detection, diagnosis, and treatment - are expected to substantially enhance our knowledge and management of AD. Stratifying the affected individuals by sex and gender is an important basic step towards personalization of scientific research, drug development, and care. We hypothesize that sex and gender differences, extending from genetic to psychosocial domains, are highly relevant for the understanding of AD pathophysiology, and for the conceptualization of basic/translational research and for clinical therapy trial design.
Collapse
|
58
|
Congdon EE. Sex Differences in Autophagy Contribute to Female Vulnerability in Alzheimer's Disease. Front Neurosci 2018; 12:372. [PMID: 29988365 PMCID: PMC6023994 DOI: 10.3389/fnins.2018.00372] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with over 5. 4 million cases in the US alone (Alzheimer's Association, 2016). Clinically, AD is defined by the presence of plaques composed of Aβ and neurofibrillary pathology composed of the microtubule associated protein tau. Another key feature is the dysregulation of autophagy at key steps in the pathway. In AD, disrupted autophagy contributes to disease progression through the failure to clear pathological protein aggregates, insulin resistance, and its role in the synthesis of Aβ. Like many psychiatric and neurodegenerative diseases, the risk of developing AD, and disease course are dependent on the sex of the patient. One potential mechanism through which these differences occur, is the effects of sex hormones on autophagy. In women, the loss of hormones with menopause presents both a risk factor for developing AD, and an obvious example of where sex differences in AD can stem from. However, because AD pathology can begin decades before menopause, this does not provide the full answer. We propose that sex-based differences in autophagy regulation during the lifespan contribute to the increased risk of AD, and greater severity of pathology seen in women.
Collapse
Affiliation(s)
- Erin E Congdon
- Neuroscience and Physiology, School of Medicine, New York University, New York City, NY, United States
| |
Collapse
|
59
|
Ziegler-Waldkirch S, Marksteiner K, Stoll J, d´Errico P, Friesen M, Eiler D, Neudel L, Sturn V, Opper I, Datta M, Prinz M, Meyer-Luehmann M. Environmental enrichment reverses Aβ pathology during pregnancy in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2018; 6:44. [PMID: 29855361 PMCID: PMC5984325 DOI: 10.1186/s40478-018-0549-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 05/20/2018] [Indexed: 11/10/2022] Open
Abstract
Several studies suggest that women have a higher risk to develop Alzheimer's disease (AD) than men. In particular, the number of pregnancies was shown to be a risk factor for AD and women with several pregnancies on average had an earlier onset of the disease, thus making childbearing a risk factor. However, the impact of being pregnant on Aβ plaque pathology and adult neurogenesis still remains elusive. Postmortem analysis revealed that pregnant 5xFAD transgenic mice had significantly more Aβ plaques in the hippocampus from G10 onwards and that the number of Ki67 and DCX positive cells dramatically decreased during the postpartum period. Furthermore, 5 months old 5xFAD transgenic mice that also nursed their offsprings for 4 weeks had a similar Aβ plaque load than merely pregnant mice, indicating that pregnancy alone is sufficient to elevate Aβ plaque levels. Interestingly, housing in an enriched environment reduced the Aβ plaque load and vivified neurogenesis. Our results suggest that pregnancy alters Aβ plaque deposition in 5xFAD transgenic mice and diminishes the generation of newborn neurons. We conclude that pregnancy alone is sufficient to induce this phenotype that can be reversed upon environmental enrichment.
Collapse
|
60
|
Maruyama T, Abe Y, Niikura T. SENP1 and SENP2 regulate SUMOylation of amyloid precursor protein. Heliyon 2018; 4:e00601. [PMID: 29862363 PMCID: PMC5968171 DOI: 10.1016/j.heliyon.2018.e00601] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 11/29/2022] Open
Abstract
Amyloid β, a key molecule in the pathogenesis of Alzheimer's disease (AD), is produced from amyloid precursor protein (APP) by the cleavage of secretases. APP is SUMOylated near the cleavage site of β-secretase. SUMOylation of APP reduces amyloid β production, but its regulatory system is still unclear. SUMOylation, a modification at a lysine residue of a target protein, is mediated by activating, conjugating, and ligating enzymes and is reversed by a family of sentrin/SUMO-specific proteases (SENPs). Here, we found that both SENP1 and SENP2 induced de-SUMOylation of APP. Using quantitative PCR, we also found that expression of SENP1 but not SENP2 increased in an age-dependent manner only in female mice. The results of immunoblot analyses showed that the protein expression was consistent with the PCR results. Females, compared to males, have a higher incidence of AD in humans and show more aggressive amyloid pathology in AD mouse models. Our results provide a clue to understanding the role of SUMOylation in the sex difference in AD pathogenesis.
Collapse
Affiliation(s)
- Takuma Maruyama
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, Japan
| | - Yoichiro Abe
- Department of Pharmacology, Keio University School of Medicine, Japan
| | - Takako Niikura
- Department of Information and Communication Sciences, Faculty of Science and Technology, Sophia University, Japan
| |
Collapse
|
61
|
Keyvani K, Münster Y, Kurapati NK, Rubach S, Schönborn A, Kocakavuk E, Karout M, Hammesfahr P, Wang YC, Hermann DM, Teuber-Hanselmann S, Herring A. Higher levels of kallikrein-8 in female brain may increase the risk for Alzheimer's disease. Brain Pathol 2018; 28:947-964. [PMID: 29505099 DOI: 10.1111/bpa.12599] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 02/28/2018] [Indexed: 01/21/2023] Open
Abstract
Women seem to have a higher vulnerability to Alzheimer's disease (AD), but the underlying mechanisms of this sex dichotomy are not well understood. Here, we first determined the influence of sex on various aspects of Alzheimer's pathology in transgenic CRND8 mice. We demonstrate that beta-amyloid (Aβ) plaque burden starts to be more severe around P180 (moderate disease stage) in female transgenics when compared to males and that aging aggravates this sex-specific difference. Furthermore, we show that female transgenics suffer from higher levels of neurovascular dysfunction around P180, resulting in impaired Aβ peptide clearance across the blood-brain-barrier at P360. Female transgenics show also higher levels of diffuse microgliosis and inflammation, but the density of microglial cells surrounding Aβ plaques is less in females. In line with this finding, testosterone compared to estradiol was able to improve microglial viability and Aβ clearance in vitro. The spatial memory of transgenics was in general poorer than in wildtypes and at P360 worse in females irrespective of their genotype. This difference was accompanied by a slightly diminished dendritic complexity in females. While all the above-named sex-differences emerged after the onset of Aβ pathology, kallikrein-8 (KLK8) protease levels were, as an exception, higher in female than in male brains very early when virtually no plaques were detectable. In a second step, we quantified cerebral KLK8 levels in AD patients and healthy controls, and could ascertain, similar to mice, higher KLK8 levels not only in AD-affected but also in healthy brains of women. Accordingly, we could demonstrate that estradiol but not testosterone induces KLK8 synthesis in neuronal and microglial cells. In conclusion, multiple features of AD are more pronounced in females. Here, we show for the first time that this sex-specific difference may be meditated by estrogen-induced KLK8 overproduction long before AD pathology emerges.
Collapse
Affiliation(s)
- Kathy Keyvani
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Yvonne Münster
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Nirup K Kurapati
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Sebastian Rubach
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Andreas Schönborn
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Emre Kocakavuk
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Mohamed Karout
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Pia Hammesfahr
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Ya-Chao Wang
- Department of Neurology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Sarah Teuber-Hanselmann
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Arne Herring
- Institute of Neuropathology, University of Duisburg-Essen, Hufelandstr. 55, 45122 Essen, Germany
| |
Collapse
|
62
|
Ruiz-Riquelme A, Lau HHC, Stuart E, Goczi AN, Wang Z, Schmitt-Ulms G, Watts JC. Prion-like propagation of β-amyloid aggregates in the absence of APP overexpression. Acta Neuropathol Commun 2018; 6:26. [PMID: 29615128 PMCID: PMC5883524 DOI: 10.1186/s40478-018-0529-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/21/2018] [Indexed: 11/10/2022] Open
Abstract
The amyloid cascade hypothesis posits that the initiating event in Alzheimer's disease (AD) is the aggregation and deposition of the β-amyloid (Aβ) peptide, which is a proteolytic cleavage product of the amyloid precursor protein (APP). Mounting evidence suggests that the formation and spread of prion-like Aβ aggregates during AD may contribute to disease progression. Inoculation of transgenic mice that overexpress APP with pre-formed Aβ aggregates results in the prion-like induction of cerebral Aβ deposition. To determine whether Aβ deposition can also be induced when physiological APP levels are present in the brain, we inoculated AppNL-F mice, a knock-in model of AD that avoids potential artifacts associated with APP overexpression, with Aβ aggregates derived from the brains of AD patients or transgenic mice. In all cases, induced Aβ deposition was apparent in the corpus callosum, olfactory bulb, and meningeal blood vessels of inoculated mice at 130-150 days post-inoculation, whereas uninoculated and buffer-inoculated animals exhibited minimal or no Aβ deposits at these ages. Interestingly, despite being predominantly composed of protease-resistant Aβ42 aggregates, the induced parenchymal Aβ deposits were largely diffuse and were unreactive to an amyloid-binding dye. These results demonstrate that APP overexpression is not a prerequisite for the prion-like induction of cerebral Aβ deposition. Accordingly, spreading of Aβ deposition may contribute to disease progression in AD patients.
Collapse
|
63
|
Liu P, Reichl JH, Rao ER, McNellis BM, Huang ES, Hemmy LS, Forster CL, Kuskowski MA, Borchelt DR, Vassar R, Ashe KH, Zahs KR. Quantitative Comparison of Dense-Core Amyloid Plaque Accumulation in Amyloid-β Protein Precursor Transgenic Mice. J Alzheimers Dis 2018; 56:743-761. [PMID: 28059792 DOI: 10.3233/jad-161027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
There exist several dozen lines of transgenic mice that express human amyloid-β protein precursor (AβPP) with Alzheimer's disease (AD)-linked mutations. AβPP transgenic mouse lines differ in the types and amounts of Aβ that they generate and in their spatiotemporal patterns of expression of Aβ assemblies, providing a toolkit to study Aβ amyloidosis and the influence of Aβ aggregation on brain function. More complete quantitative descriptions of the types of Aβ assemblies present in transgenic mice and in humans during disease progression should add to our understanding of how Aβ toxicity in mice relates to the pathogenesis of AD. Here, we provide a direct quantitative comparison of amyloid plaque burdens and plaque sizes in four lines of AβPP transgenic mice. We measured the fraction of cortex and hippocampus occupied by dense-core plaques, visualized by staining with Thioflavin S, in mice from young adulthood through advanced age. We found that the plaque burdens among the transgenic lines varied by an order of magnitude: at 15 months of age, the oldest age studied, the median cortical plaque burden in 5XFAD mice was already ∼4.5 times that of 21-month-old Tg2576 mice and ∼15 times that of 21-24-month-old rTg9191 mice. Plaque-size distributions changed across the lifespan in a line- and region-dependent manner. We also compared the dense-core plaque burdens in the mice to those measured in a set of pathologically-confirmed AD cases from the Nun Study. Cortical plaque burdens in Tg2576, APPSwePS1ΔE9, and 5XFAD mice eventually far exceeded those measured in the human cohort.
Collapse
Affiliation(s)
- Peng Liu
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA
| | - John H Reichl
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA
| | - Eshaan R Rao
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Brittany M McNellis
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Eric S Huang
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA
| | - Laura S Hemmy
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA.,GRECC, VA Medical Center, Minneapolis, MN, USA
| | - Colleen L Forster
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,UMN Academic Health Center Biological Materials Procurement Network, University of Minnesota, Minneapolis, MN, USA
| | | | - David R Borchelt
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Robert Vassar
- Department of Cellular and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karen H Ashe
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA.,Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.,GRECC, VA Medical Center, Minneapolis, MN, USA
| | - Kathleen R Zahs
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA.,N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
64
|
Tschiffely AE, Schuh RA, Prokai-Tatrai K, Ottinger MA, Prokai L. An exploratory investigation of brain-selective estrogen treatment in males using a mouse model of Alzheimer's disease. Horm Behav 2018; 98:16-21. [PMID: 29183688 PMCID: PMC5999339 DOI: 10.1016/j.yhbeh.2017.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/24/2017] [Accepted: 11/21/2017] [Indexed: 11/19/2022]
Abstract
Estrogens are neuroprotective, and studies suggest that they may mitigate the pathology and symptoms of Alzheimer's disease (AD) in female models. However, central estrogen effects have not been examined in males in the context of AD. The purpose of this follow-up study was to assess the benefits of a brain-selective 17β-estradiol estrogen prodrug, 10β,17β-hydroxyestra-1,4-dien-3-one (DHED), also in the male APPswe/PS1dE9 double-transgenic mouse model of the disease. After continuously exposing 6-month old animals to DHED for two months, their brains showed decreased amyloid precursor and amyloid-β protein levels. The DHED-treated APPswe/PS1dE9 double transgenic subjects also exhibited enhanced performance in a cognitive task, while 17β-estradiol treatment did not reach statistical significance. Taken together, data presented here suggest that DHED may also have therapeutic benefit in males and warrant further investigations to fully elucidate the potential of targeted estrogen therapy for a gender-independent treatment of early-stage AD.
Collapse
Affiliation(s)
- Anna E Tschiffely
- Neuroscience and Cognitive Science Graduate Program, University of Maryland College Park, MD 20742, USA; Department of Animal and Avian Sciences, University of Maryland College Park, MD 20742, USA
| | - Rosemary A Schuh
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Research Service, VAMHCS, Baltimore, MD 21201, USA
| | - Katalin Prokai-Tatrai
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Mary Ann Ottinger
- Department of Animal and Avian Sciences, University of Maryland College Park, MD 20742, USA.
| | - Laszlo Prokai
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
65
|
Jankowsky JL, Zheng H. Practical considerations for choosing a mouse model of Alzheimer's disease. Mol Neurodegener 2017; 12:89. [PMID: 29273078 PMCID: PMC5741956 DOI: 10.1186/s13024-017-0231-7] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 01/06/2023] Open
Abstract
Alzheimer’s disease (AD) is behaviorally identified by progressive memory impairment and pathologically characterized by the triad of β-amyloid plaques, neurofibrillary tangles, and neurodegeneration. Genetic mutations and risk factors have been identified that are either causal or modify the disease progression. These genetic and pathological features serve as basis for the creation and validation of mouse models of AD. Efforts made in the past quarter-century have produced over 100 genetically engineered mouse lines that recapitulate some aspects of AD clinicopathology. These models have been valuable resources for understanding genetic interactions that contribute to disease and cellular reactions that are engaged in response. Here we focus on mouse models that have been widely used stalwarts of the field or that are recently developed bellwethers of the future. Rather than providing a summary of each model, we endeavor to compare and contrast the genetic approaches employed and to discuss their respective advantages and limitations. We offer a critical account of the variables which may contribute to inconsistent findings and the factors that should be considered when choosing a model and interpreting the results. We hope to present an insightful review of current AD mouse models and to provide a practical guide for selecting models best matched to the experimental question at hand.
Collapse
Affiliation(s)
- Joanna L Jankowsky
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neurology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Hui Zheng
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
66
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
67
|
Pike CJ. Sex and the development of Alzheimer's disease. J Neurosci Res 2017; 95:671-680. [PMID: 27870425 DOI: 10.1002/jnr.23827] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022]
Abstract
Men and women exhibit differences in the development and progression of Alzheimer's disease (AD). The factors underlying the sex differences in AD are not well understood. This Review emphasizes the contributions of sex steroid hormones to the relationship between sex and AD. In women, events that decrease lifetime exposure to estrogens are generally associated with increased AD risk, whereas estrogen-based hormone therapy administered near the time of menopause may reduce AD risk. In men, estrogens do not exhibit age-related reduction and are not significantly associated with AD risk. Rather, normal age-related depletions of testosterone in plasma and brain predict enhanced vulnerability to AD. Both estrogens and androgens exert numerous protective actions in the adult brain that increase neural functioning and resilience as well as specifically attenuating multiple aspects of AD-related neuropathology. Aging diminishes the activational effects of sex hormones in sex-specific manners, which is hypothesized to contribute to the relationship between aging and AD. Sex steroid hormones may also drive sex differences in AD through their organizational effects during developmental sexual differentiation of the brain. Specifically, sex hormone actions during early development may confer inherent vulnerability of the female brain to development of AD in advanced age. The combined effects of organizational and activational effects of sex steroids yield distinct sex differences in AD pathogenesis, a significant variable that must be more rigorously considered in future research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|
68
|
Grant DA, Serpa R, Moattari CR, Brown A, Greco T, Prins ML, Teng E. Repeat Mild Traumatic Brain Injury in Adolescent Rats Increases Subsequent β-Amyloid Pathogenesis. J Neurotrauma 2017; 35:94-104. [PMID: 28728464 DOI: 10.1089/neu.2017.5042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Single moderate-to-severe traumatic brain injuries (TBIs) may increase subsequent risk for neurodegenerative disease by facilitating β-amyloid (Aβ) deposition. However, the chronic effects on Aβ pathogenesis of repetitive mild TBIs (rTBI), which are common in adolescents and young adults, remain uncertain. We examined the effects of rTBI sustained during adolescence on subsequent deposition of Aβ pathology in a transgenic APP/PS1 rat model. Transgenic rats received sham or four individual mild TBIs (rTBIs) separated by either 24- or 72-h intervals at post-natal day 35 (before Aβ plaque deposition). Animals were euthanized at 12 months of age and underwent immunohistochemical analyses of Aβ plaque deposition. Significantly greater hippocampal Aβ plaque deposition was observed after rTBI separated by 24 h relative to rTBI separated by 72 h or sham injuries. These increases in hippocampal Aβ plaque load were driven by increases in both plaque number and size. Similar, though less-pronounced, effects were observed in extrahippocampal regions. Increases in Aβ plaque deposition were observed both ipsilaterally and contralaterally to the injury site and in both males and females. rTBIs sustained in adolescence can increase subsequent deposition of Aβ pathology, and these effects are critically dependent on interinjury interval.
Collapse
Affiliation(s)
- Daya A Grant
- 1 Department of Neurosurgery, UCLA , Los Angeles, California.,3 Interdeparmental Program for Neuroscience, UCLA , Los Angeles, California
| | - Rebecka Serpa
- 1 Department of Neurosurgery, UCLA , Los Angeles, California
| | - Cameron R Moattari
- 3 Interdeparmental Program for Neuroscience, UCLA , Los Angeles, California
| | - Ari Brown
- 1 Department of Neurosurgery, UCLA , Los Angeles, California
| | - Tiffany Greco
- 1 Department of Neurosurgery, UCLA , Los Angeles, California
| | - Mayumi L Prins
- 1 Department of Neurosurgery, UCLA , Los Angeles, California
| | - Edmond Teng
- 2 Department of Neurology, David Geffen School of Medicine at UCLA, UCLA , Los Angeles, California.,4 Veterans Affairs Greater Los Angeles Healthcare System , Los Angeles, California
| |
Collapse
|
69
|
Sex- and age-specific modulation of brain GABA levels in a mouse model of Alzheimer's disease. Neurobiol Aging 2017; 62:168-179. [PMID: 29154037 DOI: 10.1016/j.neurobiolaging.2017.10.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/05/2017] [Accepted: 10/16/2017] [Indexed: 01/12/2023]
Abstract
Age and sex are risk factors of Alzheimer's disease (AD). Among the neurotransmitter systems, gamma-aminobutyric acid (GABA) has been implicated in AD pathogenesis but the relevance of sex-specific GABAergic dysfunction during AD progression remains unknown. In the present study, we utilized state-of-the-art high-resolution magic angle spinning nuclear magnetic resonance to systematically monitor the brain region-, age-, and sex-specific modulation of GABA levels in wild-type and Tg2576 mice with amyloid pathology. In addition, we followed the possible role of reactive astrocytes in sex-specific GABA modulation. In female Tg2576 mice, hippocampal GABA levels were significantly elevated, along with higher number of reactive astrocytes and amyloid deposition. The elevated GABA was found to be produced via the monoamine oxidase-B route from putrescine in reactive astrocytes, more substantially in female than male mice, thus suggesting a role of astrocytes in memory impairment and sex-related differences in AD. Our results paint a coherent model of memory impairment in AD and signify that dynamic changes in regional GABA may be at the root of marked sex disparities observed in AD.
Collapse
|
70
|
Richetin K, Petsophonsakul P, Roybon L, Guiard BP, Rampon C. Differential alteration of hippocampal function and plasticity in females and males of the APPxPS1 mouse model of Alzheimer's disease. Neurobiol Aging 2017; 57:220-231. [DOI: 10.1016/j.neurobiolaging.2017.05.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/19/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022]
|
71
|
Zhang J, Zhou W, Wang L, Zhang X. Gender differences of neuropsychological profiles in cognitively normal older people without amyloid pathology. Compr Psychiatry 2017; 75:22-26. [PMID: 28285181 PMCID: PMC5783637 DOI: 10.1016/j.comppsych.2017.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/16/2017] [Accepted: 02/24/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To investigate the impact of sex on cognition among cognitively normal older people without amyloid pathology. METHODS The study sample consisted of 83 males (mean age 74years, ranging from 65 to 88years) and 111 females (mean age 72years, ranging from 65 to 89years) who were cognitively normal without amyloid pathology confirmed by Pittsburgh compound B PET scan. We examined the sex discrepancies in cognition using a comprehensive neuropsychological battery. RESULTS Our data showed significantly greater advantage for women than men on the tasks of verbal memory and category fluency while better performance of naming in men than women when age, education and depressive symptoms were considered. CONCLUSION These findings highlight the importance of considering sex differences in the interpretation of cognitive data which contribute to clinical diagnosis of neurodegenerative disorders, such as Alzheimer's disease.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Wenjun Zhou
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijun Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xiangyang Zhang
- Beijing Huilongguan Hospital, Peking University, Beijing, China.
| |
Collapse
|
72
|
Zhao ZA, Ning YL, Li P, Yang N, Peng Y, Xiong RP, Zhao Y, Liu D, Zeng XJ, Chen JF, Zhou YG. Widespread hyperphosphorylated tau in the working memory circuit early after cortical impact injury of brain (Original study). Behav Brain Res 2017; 323:146-153. [DOI: 10.1016/j.bbr.2017.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 02/01/2017] [Indexed: 12/14/2022]
|
73
|
Ito K, Tatebe T, Suzuki K, Hirayama T, Hayakawa M, Kubo H, Tomita T, Makino M. Memantine reduces the production of amyloid-β peptides through modulation of amyloid precursor protein trafficking. Eur J Pharmacol 2017; 798:16-25. [PMID: 28167259 DOI: 10.1016/j.ejphar.2017.02.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 01/30/2023]
Abstract
Memantine, an uncompetitive glutamatergic N-methyl-D-aspartate (NMDA) receptor antagonist, is widely used as medication for the treatment of Alzheimer's disease (AD). It has been reported that memantine reduces amyloid-β peptide (Aβ) levels in both neuronal cultures and in brains of animal models of AD. However, the underlying mechanism of these effects is unclear. Here we examined the effect of memantine on Aβ production. Memantine was administered to 9-month-old Tg2576 mice, a transgenic mouse model of AD, at 10 or 20mg/kg/day in drinking water for 1 month. Memantine significantly reduced the amounts of both CHAPS-soluble and CHAPS-insoluble Aβ in the brains of Tg2576 mice. Memantine at 10mg/kg/day for 1 month also reduced the levels of insoluble Aβ42 in the brains of aged F344 rats. Moreover, memantine reduced Aβ and sAPPβ levels in conditioned media from rat primary cortical cultures without affecting the enzymatic activities of α-secretase, β-secretase, or γ-secretase. Notably, in a cell-surface biotinylation assay, memantine increased the amount of amyloid precursor protein (APP) at the cell surface without changing the total amount of APP. Collectively, our results indicate that chronic treatment with memantine reduces the levels of Aβ both in AD models and in aged animals, and that memantine affects the endocytosis pathway of APP, which is required for β-secretase-mediated cleavage. This leads to a reduction in Aβ production. These results suggest that memantine reduces Aβ production and plaque deposition through the regulation of intracellular trafficking of APP.
Collapse
Affiliation(s)
- Kaori Ito
- Venture Science Laboratories, R&D Division, Daiichi-Sankyo Co. Ltd., Tokyo 140-8710, Japan.
| | - Takuya Tatebe
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Kunimichi Suzuki
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Takashi Hirayama
- Biological Research Department, Daiichi Sankyo RD Novare Co. Ltd., Tokyo 134-8630, Japan.
| | - Maki Hayakawa
- Biological Research Department, Daiichi Sankyo RD Novare Co. Ltd., Tokyo 134-8630, Japan.
| | - Hideo Kubo
- Biological Research Department, Daiichi Sankyo RD Novare Co. Ltd., Tokyo 134-8630, Japan.
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Mitsuhiro Makino
- Venture Science Laboratories, R&D Division, Daiichi-Sankyo Co. Ltd., Tokyo 140-8710, Japan.
| |
Collapse
|
74
|
Long Z, Zeng Q, Wang K, Sharma A, He G. Gender difference in valproic acid-induced neuroprotective effects on APP/PS1 double transgenic mice modeling Alzheimer's disease. Acta Biochim Biophys Sin (Shanghai) 2016; 48:930-938. [PMID: 27614317 DOI: 10.1093/abbs/gmw085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/26/2016] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes progressive memory and cognitive impairment with gender difference in specific cognitive ability domains, pathology, and risk of AD. Since valproic acid (VPA) is a widely used mood stabilizer and an antiepileptic drug, which exhibits multiple neuroprotective activities on AD, this study intended to investigate the gender difference in the effect of VPA on APP/PS1 double transgenic mice modeling AD. Behavioral experiments showed that VPA reduced the autonomous behaviors, improved learning and memory, and exhibited gender differences in AD mice compared with the control mice. The decrease in senile plaque, amyloid β (Aβ) 40, and Aβ42 caused by VPA in the male AD mice was more notable than that in the female AD mice. Meanwhile, VPA protected brain cells from dying notably in the male AD mice but only slightly in the female AD mice, and VPA treatment thickened the postsynaptic density and markedly increased the number and density of presynaptic vesicles in both male and female AD mice. However, the effects of rescuing early synaptic structural and functional deficits by VPA were more obvious in the male mice. Overall, these results supported the hypothesis that gender difference significantly influences AD and indicated that VPA may be a promising remedy for AD if basic biological differences and gender specificity were prudently taken into account.
Collapse
Affiliation(s)
- Zhimin Long
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - Qinghua Zeng
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
| | - Kejian Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| | - Akhilesh Sharma
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China
| | - Guiqiong He
- Chongqing Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing 400016, China Department of Anatomy, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
75
|
Melnikova T, Park D, Becker L, Lee D, Cho E, Sayyida N, Tian J, Bandeen-Roche K, Borchelt DR, Savonenko AV. Sex-related dimorphism in dentate gyrus atrophy and behavioral phenotypes in an inducible tTa:APPsi transgenic model of Alzheimer's disease. Neurobiol Dis 2016; 96:171-185. [PMID: 27569580 DOI: 10.1016/j.nbd.2016.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 07/07/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
Sex differences are a well-known phenomenon in Alzheimer's disease (AD), with women having a higher risk for AD than men. Many AD mouse models display a similar sex-dependent pattern, with females showing earlier cognitive deficits and more severe neuropathology than males. However, whether those differences are relevant to human disease is unclear. Here we show that in AD mouse models that overexpress amyloid precursor protein (APP) under control of the prion protein promoter (PrP), female transgenic mice have higher APP expression than males, complicating interpretations of the role of sex-related factors in such models. By contrast, in a tTa:APPsi model, in which APP expression is driven by the tetracycline transactivator (tTa) from the CaMKIIα promoter, there are no sex-related differences in expression or processing of APP. In addition, the levels of Aβ dimers and tetramers, as well as Aβ peptide accumulation, are similar between sexes. Behavioral testing demonstrated that both male and female tTa:APPsi mice develop age-dependent deficits in spatial recognition memory and conditional freezing to context. These cognitive deficits were accompanied by habituation-associated hyperlocomotion and startle hyper-reactivity. Significant sex-related dimorphisms were observed, due to females showing earlier onsets of the deficits in conditioned freezing and hyperlocomotion. In addition, tTa:APPsi males but not females demonstrated a lack of novelty-induced activation. Both males and females showed atrophy of the dentate gyrus (DG) of the dorsal hippocampus, associated with widening of the pyramidal layer of the CA1 area in both sexes. Ventral DG was preserved. Sex-related differences were limited to the DG, with females showing more advanced degeneration than males. Collectively, our data show that the tTa:APPsi model is characterized by a lack of sex-related differences in APP expression, making this model useful in deciphering the mechanisms of sex differences in AD pathogenesis. Sex-related dimorphisms observed in this model under conditions of equal APP expression between sexes suggest a higher sensitivity of females to the effects of APP and/or Aβ production.
Collapse
Affiliation(s)
- Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - DaMin Park
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Lauren Becker
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Deidre Lee
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Eugenia Cho
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Nuzhat Sayyida
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| | - Jing Tian
- Department of Biostatistics, School of Public Health, The Johns Hopkins University, 615 N Wolfe St E3527, Baltimore, MD 21205, USA.
| | - Karen Bandeen-Roche
- Department of Biostatistics, School of Public Health, The Johns Hopkins University, 615 N Wolfe St E3527, Baltimore, MD 21205, USA.
| | - David R Borchelt
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, 100 Newell Drive, Gainesville, FL 32610, USA.
| | - Alena V Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, 720 Rutland Ave, Ross 558, Baltimore, MD 21205, USA.
| |
Collapse
|
76
|
Huang Y, Skwarek-Maruszewska A, Horré K, Vandewyer E, Wolfs L, Snellinx A, Saito T, Radaelli E, Corthout N, Colombelli J, Lo AC, Van Aerschot L, Callaerts-Vegh Z, Trabzuni D, Bossers K, Verhaagen J, Ryten M, Munck S, D'Hooge R, Swaab DF, Hardy J, Saido TC, De Strooper B, Thathiah A. Loss of GPR3 reduces the amyloid plaque burden and improves memory in Alzheimer's disease mouse models. Sci Transl Med 2016; 7:309ra164. [PMID: 26468326 DOI: 10.1126/scitranslmed.aab3492] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The orphan G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR) GPR3 regulates activity of the γ-secretase complex in the absence of an effect on Notch proteolysis, providing a potential therapeutic target for Alzheimer's disease (AD). However, given the vast resources required to develop and evaluate any new therapy for AD and the multiple failures involved in translational research, demonstration of the pathophysiological relevance of research findings in multiple disease-relevant models is necessary before initiating costly drug development programs. We evaluated the physiological consequences of loss of Gpr3 in four AD transgenic mouse models, including two that contain the humanized murine Aβ sequence and express similar amyloid precursor protein (APP) levels as wild-type mice, thereby reducing potential artificial phenotypes. Our findings reveal that genetic deletion of Gpr3 reduced amyloid pathology in all of the AD mouse models and alleviated cognitive deficits in APP/PS1 mice. Additional three-dimensional visualization and analysis of the amyloid plaque burden provided accurate information on the amyloid load, distribution, and volume in the structurally intact adult mouse brain. Analysis of 10 different regions in healthy human postmortem brain tissue indicated that GPR3 expression was stable during aging. However, two cohorts of human AD postmortem brain tissue samples showed a correlation between elevated GPR3 and AD progression. Collectively, these studies provide evidence that GPR3 mediates the amyloidogenic proteolysis of APP in four AD transgenic mouse models as well as the physiological processing of APP in wild-type mice, suggesting that GPR3 may be a potential therapeutic target for AD drug development.
Collapse
Affiliation(s)
- Yunhong Huang
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Aneta Skwarek-Maruszewska
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Katrien Horré
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Elke Vandewyer
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Leen Wolfs
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - An Snellinx
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan. Japan Science and Technology Agency, 332-0012 Saitama, Japan
| | - Enrico Radaelli
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Julien Colombelli
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Adrian C Lo
- Department of Psychology, Laboratory of Biological Psychology, University of Leuven, 3000 Leuven, Belgium
| | - Leen Van Aerschot
- Department of Psychology, Laboratory of Biological Psychology, University of Leuven, 3000 Leuven, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Department of Psychology, Laboratory of Biological Psychology, University of Leuven, 3000 Leuven, Belgium
| | - Daniah Trabzuni
- Departments of Molecular Neuroscience and Clinical Neuroscience, Reta Lila Weston Research Laboratories, Institute of Neurology, University College London, WC1N 3BG London, UK. Department of Genetics, King Faisal Specialist Hospital and Research Centre, 11211 Riyadh, Saudi Arabia
| | - Koen Bossers
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, Netherlands
| | - Joost Verhaagen
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, Netherlands
| | - Mina Ryten
- Departments of Molecular Neuroscience and Clinical Neuroscience, Reta Lila Weston Research Laboratories, Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Sebastian Munck
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium
| | - Rudi D'Hooge
- Department of Psychology, Laboratory of Biological Psychology, University of Leuven, 3000 Leuven, Belgium
| | - Dick F Swaab
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, Netherlands
| | - John Hardy
- Departments of Molecular Neuroscience and Clinical Neuroscience, Reta Lila Weston Research Laboratories, Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, 351-0198 Saitama, Japan
| | - Bart De Strooper
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium.
| | - Amantha Thathiah
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium. KU Leuven Center for Human Genetics and Leuven Institute for Neurodegenerative Diseases, 3000 Leuven, Belgium.
| |
Collapse
|
77
|
Grimm A, Mensah-Nyagan AG, Eckert A. Alzheimer, mitochondria and gender. Neurosci Biobehav Rev 2016; 67:89-101. [DOI: 10.1016/j.neubiorev.2016.04.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
|
78
|
Levels and actions of neuroactive steroids in the nervous system under physiological and pathological conditions: Sex-specific features. Neurosci Biobehav Rev 2016; 67:25-40. [DOI: 10.1016/j.neubiorev.2015.09.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 01/21/2023]
|
79
|
Sex and Gender Differences in Central Nervous System-Related Disorders. NEUROSCIENCE JOURNAL 2016; 2016:2827090. [PMID: 27314003 PMCID: PMC4904110 DOI: 10.1155/2016/2827090] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/08/2016] [Indexed: 12/12/2022]
Abstract
There are important sex differences in the brain that seem to arise from biology as well as psychosocial influences. Sex differences in several aspects of human behavior and cognition have been reported. Gonadal sex steroids or genes found on sex chromosomes influence sex differences in neuroanatomy, neurochemistry and neuronal structure, and connectivity. There has been some resistance to accept that sex differences in the human brain exist and have biological relevance; however, a few years ago, it has been recommended by the USA National Institute of Mental Health to incorporate sex as a variable in experimental and clinical neurological and psychiatric studies. We here review the clinical literature on sex differences in pain and neurological and psychiatric diseases, with the aim to further stimulate interest in sexual dimorphisms in the brain and brain diseases, possibly encouraging more research in the field of the implications of sex differences for treating these conditions.
Collapse
|
80
|
Chan ES, Shetty MS, Sajikumar S, Chen C, Soong TW, Wong BS. ApoE4 expression accelerates hippocampus-dependent cognitive deficits by enhancing Aβ impairment of insulin signaling in an Alzheimer's disease mouse model. Sci Rep 2016; 6:26119. [PMID: 27189808 PMCID: PMC4870502 DOI: 10.1038/srep26119] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/27/2016] [Indexed: 02/01/2023] Open
Abstract
The apolipoprotein E4 (ApoE4) is the strongest genetic risk factor for Alzheimer's disease (AD). The AD brain was shown to be insulin resistant at end stage, but the interplay between insulin signaling, ApoE4 and Aβ across time, and their involvement in memory decline is unclear. To investigate insulin response in the ageing mouse hippocampus, we crossed the human ApoE-targeted replacement mice with the mutant human amyloid precursor protein (APP) mice (ApoExAPP). While hippocampal Aβ levels were comparable between ApoE3xAPP and ApoE4xAPP mice at 26 weeks, insulin response was impaired in the ApoE4xAPP hippocampus. Insulin treatment was only able to stimulate insulin signaling and increased AMPA-GluR1 phosphorylation in forskolin pre-treated hippocampal slices from ApoE3xAPP mice. In ApoE4xAPP mice, insulin dysfunction was also associated with poorer spatial memory performance. Using dissociated hippocampal neuron in vitro, we showed that insulin response in ApoE3 and ApoE4 neurons increased AMPA receptor-mediated miniature excitatory postsynaptic current (mEPSC) amplitudes and GluR1-subunit insertion. Pre-treatment of ApoE3 neurons with Aβ42 did not affect insulin-mediated GluR1 subunit insertion. However, impaired insulin sensitivity observed only in the presence of ApoE4 and Aβ42, attenuated GluR1-subunit insertion. Taken together, our results suggest that ApoE4 enhances Aβ inhibition of insulin-stimulated AMPA receptor function, which accelerates memory impairment in ApoE4xAPP mice.
Collapse
Affiliation(s)
- Elizabeth S Chan
- Departments of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 117456, Singapore
| | - Mahesh Shivarama Shetty
- Departments of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 117456, Singapore.,Memory Networks Program, Neurobiology and Ageing Program, Life Sciences Institute, National University of Singapore 117456, Singapore
| | - Sreedharan Sajikumar
- Departments of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 117456, Singapore.,Memory Networks Program, Neurobiology and Ageing Program, Life Sciences Institute, National University of Singapore 117456, Singapore
| | - Christopher Chen
- Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore.,Memory Ageing and Cognition Centre (MACC), National University Health System (NUHS) 117599, Singapore
| | - Tuck Wah Soong
- Departments of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 117456, Singapore.,Memory Networks Program, Neurobiology and Ageing Program, Life Sciences Institute, National University of Singapore 117456, Singapore
| | - Boon-Seng Wong
- Departments of Physiology, Yong Loo Lin School of Medicine, National University of Singapore 117456, Singapore
| |
Collapse
|
81
|
Granger MW, Franko B, Taylor MW, Messier C, George-Hyslop PS, Bennett SA. A TgCRND8 Mouse Model of Alzheimer’s Disease Exhibits Sexual Dimorphisms in Behavioral Indices of Cognitive Reserve. J Alzheimers Dis 2016; 51:757-73. [DOI: 10.3233/jad-150587] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Matthew W. Granger
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Bettina Franko
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew W. Taylor
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Claude Messier
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Peter St. George-Hyslop
- Department of Clinical Neurosciences, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK and Tanz Centre for Research in Neurodegenerative Diseases University of Toronto, Toronto, ON, Canada
| | - Steffany A.L. Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
82
|
Grimm A, Biliouris EE, Lang UE, Götz J, Mensah-Nyagan AG, Eckert A. Sex hormone-related neurosteroids differentially rescue bioenergetic deficits induced by amyloid-β or hyperphosphorylated tau protein. Cell Mol Life Sci 2016; 73:201-15. [PMID: 26198711 PMCID: PMC4700074 DOI: 10.1007/s00018-015-1988-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/11/2015] [Accepted: 07/09/2015] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease marked by a progressive cognitive decline. Metabolic impairments are common hallmarks of AD, and amyloid-β (Aβ) peptide and hyperphosphorylated tau protein--the two foremost histopathological signs of AD--have been implicated in mitochondrial dysfunction. Neurosteroids have recently shown promise in alleviating cognitive and neuronal sequelae of AD. The present study evaluates the impact of neurosteroids belonging to the sex hormone family (progesterone, estradiol, estrone, testosterone, 3α-androstanediol) on mitochondrial dysfunction in cellular models of AD: human neuroblastoma cells (SH-SY5Y) stably transfected with constructs encoding (1) the human amyloid precursor protein (APP) resulting in overexpression of APP and Aβ, (2) wild-type tau (wtTau), and (3) mutant tau (P301L), that induces abnormal tau hyperphosphorylation. We show that while APP and P301L cells both display a drop in ATP levels, they present distinct mitochondrial impairments with regard to their bioenergetic profiles. The P301L cells presented a decreased maximal respiration and spare respiratory capacity, while APP cells exhibited, in addition, a decrease in basal respiration, ATP turnover, and glycolytic reserve. All neurosteroids showed beneficial effects on ATP production and mitochondrial membrane potential in APP/Aβ overexpressing cells while only progesterone and estradiol increased ATP levels in mutant tau cells. Of note, testosterone was more efficient in alleviating Aβ-induced mitochondrial deficits, while progesterone and estrogen were the most effective neurosteroids in our model of AD-related tauopathy. Our findings lend further support to the neuroprotective effects of neurosteroids in AD and may open new avenues for the development of gender-specific therapeutic approaches in AD.
Collapse
Affiliation(s)
- Amandine Grimm
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular and Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland
- Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000, Strasbourg, France
| | - Emily E Biliouris
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular and Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland
- Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland
| | - Undine E Lang
- Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Ayikoe Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 11 rue Humann, 67 000, Strasbourg, France
| | - Anne Eckert
- Neurobiology Laboratory for Brain Aging and Mental Health, Transfaculty Research Platform, Molecular and Cognitive Neuroscience, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland.
- Psychiatric University Clinics, University of Basel, Wilhelm Klein-Str. 27, 4012, Basel, Switzerland.
| |
Collapse
|
83
|
Chronic Mild Stress Assay Leading to Early Onset and Propagation of Alzheimer's Disease Phenotype in Mouse Models. Methods Mol Biol 2016; 1303:241-6. [PMID: 26235071 DOI: 10.1007/978-1-4939-2627-5_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A comprehensive chronic mild stress (CMS) procedure is presented, which consists in the application of unpredictable mild stressors to animal models in a random order for several weeks. This assay can be applied to Alzheimer's disease (AD) mouse models, leading to accelerated onset and increased severity of AD phenotypes and signs, including memory deficits and the accumulation of amyloid-β and phospho-tau. These assays open the way towards advanced studies on the influence of sustained mild stress, stress responses and pathways on the onset and propagation of Alzheimer's disease.
Collapse
|
84
|
Jiao SS, Bu XL, Liu YH, Zhu C, Wang QH, Shen LL, Liu CH, Wang YR, Yao XQ, Wang YJ. Sex Dimorphism Profile of Alzheimer's Disease-Type Pathologies in an APP/PS1 Mouse Model. Neurotox Res 2015; 29:256-66. [PMID: 26707129 DOI: 10.1007/s12640-015-9589-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/06/2015] [Accepted: 12/10/2015] [Indexed: 01/23/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly, characterized by parenchymal and vascular beta-amyloid (Aβ) burden, tau pathology, neuroinflammation, and loss of neurons and synapses. There is a clear sex difference in the prevalence of AD. However, sex differences in AD-type pathologies have not been systematically documented. Applying 12-month-old female and male APP/PS1 mice as a model, we investigated the sex dimorphism in these major pathological indices. Compared with male APP/PS1 mice, the females exhibited higher parenchymal Aβ burdens, with the sex difference in hippocampus being the most significant. Female APP/PS1 mice had more severe cerebral amyloid angiopathy and subsequent microhemorrhage. In addition, female APP/PS1 mice also showed higher levels of phosphorylated tau and proinflammatory cytokines, more severe astrocytosis and microgliosis, and greater neuronal and synaptic degenerations. The present study systematically described a sex dimorphism in AD-type pathologic indices, suggesting that gender should be taken into account in designing studies involving these pathological indices and when interpreting the relevant findings in those studies.
Collapse
Affiliation(s)
- Shu-Sheng Jiao
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xian-Le Bu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Hui Liu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Chi Zhu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Qing-Hua Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Lin-Lin Shen
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Cheng-Hui Liu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ye-Ran Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiu-Qing Yao
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
85
|
Murakami K, Watanabe T, Koike T, Kamata M, Igari T, Kondo S. Pharmacological properties of a novel and potent γ-secretase modulator as a therapeutic option for the treatment of Alzheimer's disease. Brain Res 2015; 1633:73-86. [PMID: 26707977 DOI: 10.1016/j.brainres.2015.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 11/17/2022]
Abstract
Previous studies of γ-secretase inhibitors (GSIs) and Notch-sparing GSIs have shown reduced amyloid-β (Aβ) peptide levels but increased Notch-related and -unrelated adverse effects. In this study, we examined the effects of compound-1 on Aβ processing and cognitive function and assessed Notch-related and -unrelated adverse effects. Compound-1 reduced Aβ40 and Aβ42 levels but inversely increased Aβ37 in Neuro2a cells, leading to no net changes in total Aβ levels, indicating that compound-1 is a γ-secretase modulator (GSM). In time-course experiments, compound-1 reduced Aβ40 and Aβ42 levels in tris-soluble fractions, with peak reduction at approximately 3h after oral administration in C57BL mice. Moreover, at >1mg/kg, compound-1 dose dependently reduced Aβ40 and Aβ42 levels in Tg2576 mice. Chronic treatment with compound-1 in Tg2576 mice for 4 months significantly reduced both soluble and insoluble Aβ42 levels and ameliorated cognitive impairments, even after drug withdrawal for 10 days following oral administration for 2 months. In contrast with compound-1, at 100-fold higher doses (100mg/kg), the GSI LY450139 decreased HES1 mRNA expression in thymus tissues and increased the intensity of periodic acid-Schiff (PAS)-positive areas in the intestine. Moreover, the Notch-sparing GSI BMS708163 led to amyloid precursor protein (APP)-β-C-terminal fragment accumulation in mouse primary neurons. BMS708163 significantly hampered cognitive function in normal mice 1 month after administration, whereas compound-1 significantly improved cognitive function. Taken together, the present novel and orally active GSM is a promising molecule for the treatment of pathologies associated with Aβ42 and Aβ40.
Collapse
Affiliation(s)
- Koji Murakami
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomomichi Watanabe
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tatsuki Koike
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Makoto Kamata
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomoko Igari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Shinichi Kondo
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan.
| |
Collapse
|
86
|
Fernández-Fernández D, Dorner-Ciossek C, Kroker KS, Rosenbrock H. Age-related synaptic dysfunction in Tg2576 mice starts as a failure in early long-term potentiation which develops into a full abolishment of late long-term potentiation. J Neurosci Res 2015; 94:266-81. [PMID: 26629777 DOI: 10.1002/jnr.23701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/30/2015] [Accepted: 11/17/2015] [Indexed: 11/12/2022]
Abstract
Tg2576 mice are widely used to study amyloid-dependent synaptic dysfunction related to Alzheimer's disease. However, conflicting data have been reported for these mice with regard to basal transmission as well as the in vitro correlate of memory, long-term potentiation (LTP). Some studies show clear impairments, whereas others report no deficiency. The present study uses hippocampal slices from 3-, 10-, and 15-month-old wild-type (WT) and Tg2576 mice to evaluate synaptic function in each group, including experiments to investigate basal synaptic transmission, short- and long-term plasticity by inducing paired-pulse facilitation, and both early and late LTP. We show that synaptic function remains intact in hippocampal slices from Tg2576 mice at 3 months of age. However, both early and late LTP decline progressively during aging in these mice. This deterioration of synaptic plasticity starts affecting early LTP, ultimately leading to the abolishment of both forms of LTP in 15-month-old animals. In comparison, WT littermates display normal synaptic parameters during aging. Additional pharmacological investigation into the involvement of NMDA receptors and L-type voltage-gated calcium channels in LTP suggests a distinct mechanism of induction among age groups, demonstrating that both early and late LTP are differentially affected by these channels in Tg2576 mice during aging.
Collapse
Affiliation(s)
- Diego Fernández-Fernández
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Cornelia Dorner-Ciossek
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Katja S Kroker
- Deptartment of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| | - Holger Rosenbrock
- Deparment of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach (Riss), Germany
| |
Collapse
|
87
|
Neuronal-Targeted TFEB Accelerates Lysosomal Degradation of APP, Reducing Aβ Generation and Amyloid Plaque Pathogenesis. J Neurosci 2015; 35:12137-51. [PMID: 26338325 DOI: 10.1523/jneurosci.0705-15.2015] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED In AD, an imbalance between Aβ production and removal drives elevated brain Aβ levels and eventual amyloid plaque deposition. APP undergoes nonamyloidogenic processing via α-cleavage at the plasma membrane, amyloidogenic β- and γ-cleavage within endosomes to generate Aβ, or lysosomal degradation in neurons. Considering multiple reports implicating impaired lysosome function as a driver of increased amyloidogenic processing of APP, we explored the efficacy of targeting transcription factor EB (TFEB), a master regulator of lysosomal pathways, to reduce Aβ levels. CMV promoter-driven TFEB, transduced via stereotactic hippocampal injections of adeno-associated virus particles in APP/PS1 mice, localized primarily to neuronal nuclei and upregulated lysosome biogenesis. This resulted in reduction of APP protein, the α and β C-terminal APP fragments (CTFs), and in the steady-state Aβ levels in the brain interstitial fluid. In aged mice, total Aβ levels and amyloid plaque load were selectively reduced in the TFEB-transduced hippocampi. TFEB transfection in N2a cells stably expressing APP695, stimulated lysosome biogenesis, reduced steady-state levels of APP and α- and β-CTFs, and attenuated Aβ generation by accelerating flux through the endosome-lysosome pathway. Cycloheximide chase assays revealed a shortening of APP half-life with exogenous TFEB expression, which was prevented by concomitant inhibition of lysosomal acidification. These data indicate that TFEB enhances flux through lysosomal degradative pathways to induce APP degradation and reduce Aβ generation. Activation of TFEB in neurons is an effective strategy to attenuate Aβ generation and attenuate amyloid plaque deposition in AD. SIGNIFICANCE STATEMENT A key driver for AD pathogenesis is the net balance between production and clearance of Aβ, the major component of amyloid plaques. Here we demonstrate that lysosomal degradation of holo-APP influences Aβ production by limiting the availability of APP for amyloidogenic processing. Using viral gene transfer of transcription factor EB (TFEB), a master regulator of lysosome biogenesis in neurons of APP/PS1 mice, steady-state levels of APP were reduced, resulting in decreased interstitial fluid Aβ levels and attenuated amyloid deposits. These effects were caused by accelerated lysosomal degradation of endocytosed APP, reflected by reduced APP half-life and steady-state levels in TFEB-expressing cells, with resultant decrease in Aβ production and release. Additional studies are needed to explore the therapeutic potential of this approach.
Collapse
|
88
|
Ye L, Hamaguchi T, Fritschi SK, Eisele YS, Obermüller U, Jucker M, Walker LC. Progression of Seed-Induced Aβ Deposition within the Limbic Connectome. Brain Pathol 2015; 25:743-52. [PMID: 25677332 PMCID: PMC4530099 DOI: 10.1111/bpa.12252] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/02/2015] [Indexed: 12/16/2022] Open
Abstract
An important early event in the pathogenesis of Alzheimer's disease (AD) is the aberrant polymerization and extracellular accumulation of amyloid-β peptide (Aβ). In young transgenic mice expressing the human Aβ-precursor protein (APP), deposits of Aβ can be induced by the inoculation of minute amounts of brain extract containing Aβ aggregates ("Aβ seeds"), indicative of a prion-like seeding phenomenon. Moreover, focal intracerebral injection of Aβ seeds can induce deposits not only in the immediate vicinity of the injection site, but, with time, also in distal regions of the brain. However, it remains uncertain whether the spatial progression of Aβ deposits occurs via nonsystematic diffusion from the injection site to proximal regions or via directed transit along neuroanatomical pathways. To address this question, we analyzed the spatiotemporal emergence of Aβ deposits in two different APP-transgenic mouse models that had been previously inoculated with Aβ seeds into the hippocampal formation. The results revealed a specific, neuroanatomically constrained pattern of induced Aβ deposits in structures corresponding to the limbic connectome, supporting the hypothesis that neuronal pathways act as conduits for the movement of proteopathic agents among brain regions, thereby facilitating the progression of disease.
Collapse
Affiliation(s)
- Lan Ye
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- Graduate School of Cellular and Molecular NeuroscienceUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Tsuyoshi Hamaguchi
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Sarah K. Fritschi
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- Graduate School of Cellular and Molecular NeuroscienceUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Yvonne S. Eisele
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Ulrike Obermüller
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Mathias Jucker
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Lary C. Walker
- Department of Cellular NeurologyHertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- Yerkes National Primate Research CenterEmory UniversityAtlantaGA
- Department of NeurologyEmory UniversityAtlantaGA
| |
Collapse
|
89
|
Caspase-dependent degradation of MDMx/MDM4 cell cycle regulatory protein in amyloid β-induced neuronal damage. Neurosci Lett 2015; 609:182-8. [PMID: 26477779 DOI: 10.1016/j.neulet.2015.10.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/07/2015] [Accepted: 10/10/2015] [Indexed: 11/23/2022]
Abstract
MDMx/MDM4 is a negative regulator of the p53 tumor suppressor protein and is necessary for survival in dividing cells. MDMx is also expressed in postmitotic neurons, with prosurvival roles that are independent of its extensively described roles in carcinogenesis. We and others have shown a role for MDMx loss in neuronal death in vitro and in vivo in several neurodegenerative diseases. Further, we have recently shown that MDMx is targeted for proteolytic degradation by calcium-dependent proteases, calpains, in neurons in vitro, and that MDMx overexpression provided partial neuroprotection in a model of HIV-associated neurodegeneration. Here, we assessed whether amyloid β (Aβ)-induced MDMx degradation occurred in Alzheimer's Disease (AD) models. Our data shows an age-dependent reduction in MDMx levels in cholinergic neurons within the cortex of adult mice expressing the swedish mutant of the amyloid precursor protein, APP in the Tg2576 murine model of AD. In vitro, Aβ treatment of primary cortical neurons led to the caspase-dependent MDMx degradation. Our findings suggest that MDMx degradation associated with neuronal death occurs via caspase activation in neurons, and that the progressive loss of MDMx protein represents a potential mechanism of Aβ-induced neuronal death during disease progression in AD.
Collapse
|
90
|
Differential interaction of Apolipoprotein-E isoforms with insulin receptors modulates brain insulin signaling in mutant human amyloid precursor protein transgenic mice. Sci Rep 2015; 5:13842. [PMID: 26346625 PMCID: PMC4561911 DOI: 10.1038/srep13842] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 08/07/2015] [Indexed: 12/12/2022] Open
Abstract
It is unclear how human apolipoprotein E4 (ApoE4) increases the risk for Alzheimer’s disease (AD). Although Aβ levels can lead to insulin signaling impairment, these experiments were done in the absence of human ApoE. To examine ApoE role, we crossed the human ApoE-targeted replacement mice with mutant human amyloid precursor protein (APP) mice. In 26 week old mice with lower Aβ levels, the expression and phosphorylation of insulin signaling proteins remained comparable among APP, ApoE3xAPP and ApoE4xAPP mouse brains. When the mice aged to 78 weeks, these proteins were markedly reduced in APP and ApoE4xAPP mouse brains. While Aβ can bind to insulin receptor, how ApoE isoforms modulate this interaction remains unknown. Here, we showed that ApoE3 had greater association with insulin receptor as compared to ApoE4, regardless of Aβ42 concentration. In contrast, ApoE4 bound more Aβ42 with increasing peptide levels. Using primary hippocampal neurons, we showed that ApoE3 and ApoE4 neurons are equally sensitive to physiological levels of insulin. However, in the presence of Aβ42, insulin failed to elicit a downstream response only in ApoE4 hippocampal neurons. Taken together, our data show that ApoE genotypes can modulate this Aβ-mediated insulin signaling impairment.
Collapse
|
91
|
Vandal M, White PJ, Chevrier G, Tremblay C, St.‐Amour I, Planel E, Marette A, Calon F. Age‐dependent impairment of glucose tolerance in the 3xTg‐AD mouse model of Alzheimer's disease. FASEB J 2015; 29:4273-84. [DOI: 10.1096/fj.14-268482] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 06/22/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Milene Vandal
- Faculté de PharmacieUniversité LavalQuébecQuébecCanada
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université LavalQuébecQuébecCanada
- Axe NeurosciencesCentre de Recherche du Centre Hospitalier de l'Université Laval (CHUL)QuébecQuébecCanada
| | - Phillip J. White
- Faculté de PharmacieUniversité LavalQuébecQuébecCanada
- Département de Medicine, Axe de Cardiologie, Faculté de MédicineUniversité LavalQuébecQuébecCanada
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology InstituteDurhamNorth CarolinaUSA
| | - Geneviève Chevrier
- Département de Medicine, Axe de Cardiologie, Faculté de MédicineUniversité LavalQuébecQuébecCanada
- Institut Universitaire de Pneumologie et de CardiologieQuébecQuébecCanada
| | - Cyntia Tremblay
- Axe NeurosciencesCentre de Recherche du Centre Hospitalier de l'Université Laval (CHUL)QuébecQuébecCanada
| | - Isabelle St.‐Amour
- Faculté de PharmacieUniversité LavalQuébecQuébecCanada
- Axe NeurosciencesCentre de Recherche du Centre Hospitalier de l'Université Laval (CHUL)QuébecQuébecCanada
| | - Emmanuel Planel
- Axe NeurosciencesCentre de Recherche du Centre Hospitalier de l'Université Laval (CHUL)QuébecQuébecCanada
| | - Andre Marette
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université LavalQuébecQuébecCanada
- Département de Medicine, Axe de Cardiologie, Faculté de MédicineUniversité LavalQuébecQuébecCanada
- Institut Universitaire de Pneumologie et de CardiologieQuébecQuébecCanada
| | - Frederic Calon
- Faculté de PharmacieUniversité LavalQuébecQuébecCanada
- Institut des Nutraceutiques et des Aliments Fonctionnels, Université LavalQuébecQuébecCanada
- Axe NeurosciencesCentre de Recherche du Centre Hospitalier de l'Université Laval (CHUL)QuébecQuébecCanada
| |
Collapse
|
92
|
Enhancement of β-amyloid oligomer accumulation after intracerebroventricular injection of streptozotocin, which involves central insulin signaling in a transgenic mouse model. Neuroreport 2015; 25:1289-95. [PMID: 25202927 DOI: 10.1097/wnr.0000000000000261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The β-amyloid (Aβ) oligomer rather than fibrillar Aβ has become the important focus of recent studies on the pathogenesis of Alzheimer's disease (AD). Insulin signaling plays important roles in cognitive disease, such as AD. However, in-vivo evidence for the link between central insulin signaling and the Aβ oligomer are lacking, and the mechanisms underlying the effect of central insulin signaling on AD are still elusive. Our team has established the Presenilin-1 Val97Leu mutant transgenic (PS1V97L) AD mouse model with the intraneuronal Aβ oligomer as the potential initiator for other pathologies, but without extracellular amyloid plaque formation. Using this model, we investigated the roles of disturbed central insulin signaling induced by intracerebroventricular injection of streptozotocin (STZ) in the progression of AD. We observed that PS1V97L mice after intracerebroventricular injection of STZ showed increased Aβ oligomer accumulation and aggravated spatial learning and memory deficit in the absence of diabetes symptoms. Furthermore, STZ administration inhibited the activation of the insulin receptor and enhanced the activation of c-Jun NH2-terminal kinase, which was accompanied by increased production of carboxy-terminal fragments from the amyloid precursor protein, in the brain of PS1V97L mice. Overall, our study provided in-vivo evidence for a role of central insulin signaling in AD progression.
Collapse
|
93
|
Oxidative Stress during the Progression of β-Amyloid Pathology in the Neocortex of the Tg2576 Mouse Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:967203. [PMID: 25973140 PMCID: PMC4418010 DOI: 10.1155/2015/967203] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/05/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive neurodegeneration. Pathogenetic mechanisms, triggered by β-amyloid (Aβ) accumulation, include oxidative stress, derived from energy homeostasis deregulation and involving mitochondria and peroxisomes. We here addressed the oxidative stress status and the elicited cellular response at the onset and during the progression of Aβ pathology, studying the neocortex of Tg2576 model of AD. Age-dependent changes of oxidative damage markers, antioxidant enzymes, and related transcription factors were analysed in relation to the distribution of Aβ peptide and oligomers, by a combined molecular/morphological approach. Nucleic acid oxidative damage, accompanied by defective antioxidant defences, and decreased PGC1α expression are already detected in 3-month-old Tg2576 neurons. Conversely, PPARα is increased in these cells, with its cytoplasmic localization suggesting nongenomic, anti-inflammatory actions. At 6 months, when intracellular Aβ accumulates, PMP70 is downregulated, indicating impairment of fatty acids peroxisomal translocation and their consequent harmful accumulation. In 9-month-old Tg2576 neocortex, Aβ oligomers and acrolein deposition correlate with GFAP, GPX1, and PMP70 increases, supporting a compensatory response, involving astroglial peroxisomes. At severe pathological stages, when senile plaques disrupt cortical cytoarchitecture, antioxidant capacity is gradually lost. Overall, our data suggest early therapeutic intervention in AD, also targeting peroxisomes.
Collapse
|
94
|
Chan J, Jones NC, Bush AI, O'Brien TJ, Kwan P. A mouse model of Alzheimer's disease displays increased susceptibility to kindling and seizure-associated death. Epilepsia 2015; 56:e73-7. [DOI: 10.1111/epi.12993] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Jianxiong Chan
- Department of Medicine (RMH); The University of Melbourne; Parkville Victoria Australia
| | - Nigel C. Jones
- Department of Medicine (RMH); The University of Melbourne; Parkville Victoria Australia
| | - Ashley I. Bush
- Florey Institute of Neuroscience and Mental Health; The University of Melbourne; Parkville Victoria Australia
| | - Terence J. O'Brien
- Department of Medicine (RMH); The University of Melbourne; Parkville Victoria Australia
- Department of Neurology; Royal Melbourne Hospital; The University of Melbourne; Parkville Victoria Australia
| | - Patrick Kwan
- Department of Medicine (RMH); The University of Melbourne; Parkville Victoria Australia
- Department of Neurology; Royal Melbourne Hospital; The University of Melbourne; Parkville Victoria Australia
| |
Collapse
|
95
|
Sadleir KR, Eimer WA, Cole SL, Vassar R. Aβ reduction in BACE1 heterozygous null 5XFAD mice is associated with transgenic APP level. Mol Neurodegener 2015; 10:1. [PMID: 25567526 PMCID: PMC4297413 DOI: 10.1186/1750-1326-10-1] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/21/2014] [Indexed: 11/10/2022] Open
Abstract
Background The β-secretase, BACE1, cleaves APP to initiate generation of the β-amyloid peptide, Aβ, that comprises amyloid plaques in Alzheimer’s disease (AD). Reducing BACE1 activity is an attractive therapeutic approach to AD, but complete inhibition of BACE1 could have mechanism-based side-effects as BACE1−/− mice show deficits in axon guidance, myelination, memory, and other neurological processes. Since BACE1+/− mice appear normal there is interest in determining whether 50% reduction in BACE1 is potentially effective in preventing or treating AD. APP transgenic mice heterozygous for BACE1 have decreased Aβ but the extent of reduction varies greatly from study to study. Here we assess the effects of 50% BACE1 reduction on the widely used 5XFAD mouse model of AD. Results 50% BACE1 reduction reduces Aβ42, plaques, and BACE1-cleaved APP fragments in female, but not in male, 5XFAD/BACE1+/− mice. 5XFAD/BACE1+/+ females have higher levels of Aβ42 and steady-state transgenic APP than males, likely caused by an estrogen response element in the transgene Thy-1 promoter. We hypothesize that higher transgenic APP level in female 5XFAD mice causes BACE1 to no longer be in excess over APP so that 50% BACE1 reduction has a significant Aβ42 lowering effect. In contrast, the lower APP level in 5XFAD males allows BACE1 to be in excess over APP even at 50% BACE1 reduction, preventing lowering of Aβ42 in 5XFAD/BACE1+/− males. We also developed and validated a dot blot assay with an Aβ42-selective antibody as an accurate and cost-effective alternative to ELISA for measuring cerebral Aβ42 levels. Conclusions 50% BACE1 reduction lowers Aβ42 in female 5XFAD mice only, potentially because BACE1 is not in excess over APP in 5XFAD females with higher transgene expression, while BACE1 is in excess over APP in 5XFAD males with lower transgene expression. Our results suggest that greater than 50% BACE1 inhibition might be necessary to significantly lower Aβ, given that BACE1 is likely to be in excess over APP in the human brain. Additionally, in experiments using the 5XFAD mouse model, or other Thy-1 promoter transgenic mice, equal numbers of male and female mice should be used, in order to avoid artifactual gender-related differences.
Collapse
Affiliation(s)
| | | | | | - Robert Vassar
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60605, USA.
| |
Collapse
|
96
|
Wang B, Tanaka K, Ji B, Ono M, Fang Y, Ninomiya Y, Maruyama K, Izumi-Nakajima N, Begum N, Higuchi M, Fujimori A, Uehara Y, Nakajima T, Suhara T, Nenoi M. Low-dose total-body carbon-ion irradiations induce early transcriptional alteration without late Alzheimer's disease-like pathogenesis and memory impairment in mice. J Neurosci Res 2015; 92:915-26. [PMID: 24936619 DOI: 10.1002/jnr.23363] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The cause and risk factors of Alzheimer's disease (AD) are largely unknown. Studies on possible radiation-induced AD-like pathogenesis and behavioral consequences are important because humans are exposed to ionizing radiation (IR) from various sources. It was reported that total-body irradiations (TBI) at 10 cGy of low linear energy transfer (LET) X-rays to mice triggered acute transcriptional alterations in genes associated with cognitive dysfunctions. However, it was unknown whether low doses of IR could induce AD-like changes late after exposure. We reported previously that 10 cGy X-rays induced early transcriptional response of several AD-related genes in hippocampi without late AD-like pathogenesis and memory impairment in mice. Here, further studies on two low doses (5 or 10 cGy) of high LET carbonion irradiations are reported. On expression of 84 AD-related genes in hippocampi, at 4 hr after TBI, 5 cGy induced a significant upregulation of three genes (Abca1, Casp3, and Chat) and 10 cGy led to a marked upregulation of one gene (Chat) and a downregulation of three genes (Apoe, Ctsd, and Il1α), and, at 1 year after TBI, one gene (Il1α) was significantly downregulated in 10 cGy-irradiated animals. Changes in spatial learning ability and memory and induction of AD-like pathogenesis were not detected by in vivo brain imaging for amyloid-β peptide accumulation and by immunohistochemical staining of amyloid precursor protein, amyloid-β protein, tau, and phosphorylated tau protein. These findings indicate that low doses of carbon-ion irradiations did not cause behavioral impairment or AD-like pathological change in mice.
Collapse
|
97
|
Schafer MJ, Alldred MJ, Lee SH, Calhoun ME, Petkova E, Mathews PM, Ginsberg SD. Reduction of β-amyloid and γ-secretase by calorie restriction in female Tg2576 mice. Neurobiol Aging 2014; 36:1293-302. [PMID: 25556162 DOI: 10.1016/j.neurobiolaging.2014.10.043] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 09/22/2014] [Accepted: 10/07/2014] [Indexed: 12/21/2022]
Abstract
Research indicates that female risk of developing Alzheimer's disease (AD) is greater than that of males. Moderate reduction of calorie intake, known as calorie restriction (CR), reduces pathology in AD mouse models and is a potentially translatable prevention measure for individuals at-risk for AD, as well as an important tool for understanding how the brain endogenously attenuates age-related pathology. Whether sex influences the response to CR remains unknown. In this study, we assessed the effect of CR on beta-amyloid peptide (Aβ) pathology and hippocampal CA1 neuron specific gene expression in the Tg2576 mouse model of cerebral amyloidosis. Relative to ad libitum (AL) feeding, CR feeding significantly reduced hippocampal Aβ burden in 15-month-old female, but not age-matched male, Tg2576 mice. Sustained CR also significantly reduced expression of presenilin enhancer 2 (Psenen) and presenilin 1, components of the γ-secretase complex, in Tg2576 females. These results indicate that long-term CR significantly reduces age-dependent female Tg2576 Aβ pathology, which is likely to involve CR-mediated reductions in γ-secretase-dependent amyloid precursor protein (APP) metabolism.
Collapse
Affiliation(s)
- Marissa J Schafer
- Cell and Molecular Biology Program, New York University Langone Medical Center, New York, NY, USA; Department of Cell Biology, New York University Langone Medical Center, New York, NY, USA; Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| | - Melissa J Alldred
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Sang Han Lee
- Division of Medical Physics, Nathan Kline Institute, Orangeburg, NY, USA
| | | | - Eva Petkova
- Department of Child and Adolescent Psychiatry, New York University Langone Medical Center, New York, NY, USA; Division of Child Psychiatry, Nathan Kline Institute, Orangeburg, NY, USA
| | - Paul M Mathews
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Stephen D Ginsberg
- Cell and Molecular Biology Program, New York University Langone Medical Center, New York, NY, USA; Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA; Department of Physiology & Neuroscience, New York University Langone Medical Center, New York, NY, USA.
| |
Collapse
|
98
|
Cañete T, Blázquez G, Tobeña A, Giménez-Llort L, Fernández-Teruel A. Cognitive and emotional alterations in young Alzheimer's disease (3xTgAD) mice: effects of neonatal handling stimulation and sexual dimorphism. Behav Brain Res 2014; 281:156-71. [PMID: 25446741 DOI: 10.1016/j.bbr.2014.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
Alzheimer disease is the most common neurodegenerative disorder and cause of senile dementia. It is characterized by an accelerated memory loss, and alterations of mood, reason, judgment and language. The main neuropathological hallmarks of the disorder are β-amyloid (βA) plaques and neurofibrillary Tau tangles. The triple transgenic 3xTgAD mouse model develops βA and Tau pathologies in a progressive manner which mimicks the pattern that takes place in the human brain with AD, and showing cognitive alterations characteristic of the disease. The present study intended to examine whether 3xTgAD mice of both sexes present cognitive, emotional and other behavioral alterations at the early age of 4 months, an age in which only some intraneuronal amyloid accumulation is found. Neonatal handling (H) is an early-life treatment known to produce profound and long-lasting behavioral and neurobiological effects in rodents, as well as improvements in cognitive functions. Therefore, we also aimed at evaluating the effects of H on the behavioral/cognitive profile of 4-month-old male and female 3xTgAD mice. The results indicate that, (1) 3xTgAD mice present spatial learning/memory deficits and emotional alterations already at the early age of 4 months, (2) there exists sexual dimorphism effects on several behavioral variables at this age, (3) neonatal handling exerts a preventive effect on some cognitive (spatial learning) and emotional alterations appearing in 3xTgAD mice already at early ages, and 4) H treatment appears to produce stronger positive effects in females than in males in several spatial learning measures and in the open field test.
Collapse
Affiliation(s)
- T Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| | - G Blázquez
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - L Giménez-Llort
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - A Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
99
|
Zeiss CJ. Improving the predictive value of interventional animal models data. Drug Discov Today 2014; 20:475-82. [PMID: 25448761 DOI: 10.1016/j.drudis.2014.10.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/03/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022]
Abstract
For many chronic diseases, translational success using the animal model paradigm has reached an impasse. Using Alzheimer's disease as an example, this review employs a networks-based method to assess repeatability of outcomes across species, by intervention and mechanism. Over 75% of animal studies reported an improved outcome. Strain background was a significant potential confounder. Five percent of interventions had been tested across animals and humans, or examined across three or more animal models. Positive outcomes across species emerged for donepezil, memantine and exercise. Repeatable positive outcomes in animals were identified for the amyloid hypothesis and three additional mechanisms. This approach supports in silico reduction of positive outcomes bias in animal studies.
Collapse
Affiliation(s)
- Caroline J Zeiss
- Section of Comparative Medicine, Yale University School of Medicine, 375 Congress Ave, New Haven, CT 06520, USA.
| |
Collapse
|
100
|
Fragkouli A, Tsilibary EC, Tzinia AK. Neuroprotective role of MMP-9 overexpression in the brain of Alzheimer's 5xFAD mice. Neurobiol Dis 2014; 70:179-89. [DOI: 10.1016/j.nbd.2014.06.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/05/2014] [Accepted: 06/28/2014] [Indexed: 12/20/2022] Open
|