1
|
Bansal VA, Tan JM, Soon HR, Zainolabidin N, Saido T, Ch'ng TH. Aβ-driven nuclear pore complex dysfunction alters activation of necroptosis proteins in a mouse model of Alzheimer's disease. eLife 2025; 13:RP92069. [PMID: 40132021 PMCID: PMC11936419 DOI: 10.7554/elife.92069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
The emergence of Aβ pathology is one of the hallmarks of Alzheimer's disease (AD), but the mechanisms and impact of Aβ in progression of the disease is unclear. The nuclear pore complex (NPC) is a multi-protein assembly in mammalian cells that regulates movement of macromolecules across the nuclear envelope; its function is shown to undergo age-dependent decline during normal aging and is also impaired in multiple neurodegenerative disorders. Yet not much is known about the impact of Aβ on NPC function in neurons. Here, we examined NPC and nucleoporin (NUP) distribution and nucleocytoplasmic transport using a mouse model of AD (AppNL-G-F/NL-G-F) that expresses Aβ in young animals. Our studies revealed that a time-dependent accumulation of intracellular Aβ corresponded with a reduction of NPCs and NUPs in the nuclear envelope which resulted in the degradation of the permeability barrier and inefficient segregation of nucleocytoplasmic proteins, and active transport. As a result of the NPC dysfunction App KI neurons become more vulnerable to inflammation-induced necroptosis - a programmed cell death pathway where the core components are activated via phosphorylation through nucleocytoplasmic shutting. Collectively, our data implicates Aβ in progressive impairment of nuclear pore function and further confirms that the protein complex is vulnerable to disruption in various neurodegenerative diseases and is a potential therapeutic target.
Collapse
Affiliation(s)
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | - Hui Rong Soon
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| | | | - Takaomi Saido
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingaporeSingapore
- School of Biological Science, Nanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
2
|
Isidro F. Brain aging and Alzheimer's disease, a perspective from non-human primates. Aging (Albany NY) 2024; 16:13145-13171. [PMID: 39475348 PMCID: PMC11552644 DOI: 10.18632/aging.206143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/03/2024] [Indexed: 11/07/2024]
Abstract
Brain aging is compared between Cercopithecinae (macaques and baboons), non-human Hominidae (chimpanzees, orangutans, and gorillas), and their close relative, humans. β-amyloid deposition in the form of senile plaques (SPs) and cerebral β-amyloid angiopathy (CAA) is a frequent neuropathological change in non-human primate brain aging. SPs are usually diffuse, whereas SPs with dystrophic neurites are rare. Tau pathology, if present, appears later, and it is generally mild or moderate, with rare exceptions in rhesus macaques and chimpanzees. Behavior and cognitive impairment are usually mild or moderate in aged non-human primates. In contrast, human brain aging is characterized by early tau pathology manifested as neurofibrillary tangles (NFTs), composed of paired helical filaments (PHFs), progressing from the entorhinal cortex, hippocampus, temporal cortex, and limbic system to other brain regions. β-amyloid pathology appears decades later, involves the neocortex, and progresses to the paleocortex, diencephalon, brain stem, and cerebellum. SPs with dystrophic neurites containing PHFs and CAA are common. Cognitive impairment and dementia of Alzheimer's type occur in about 1-5% of humans aged 65 and about 25% aged 85. In addition, other proteinopathies, such as limbic-predominant TDP-43 encephalopathy, amygdala-predominant Lewy body disease, and argyrophilic grain disease, primarily affecting the archicortex, paleocortex, and amygdala, are common in aged humans but non-existent in non-human primates. These observations show that human brain aging differs from brain aging in non-human primates, and humans constitute the exception among primates in terms of severity and extent of brain aging damage.
Collapse
Affiliation(s)
- Ferrer Isidro
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
- Reial Acadèmia de Medicina de Catalunya, Barcelona, Spain
| |
Collapse
|
3
|
Barnes CA, Permenter MR, Vogt JA, Chen K, Beach TG. Human Alzheimer's Disease ATN/ABC Staging Applied to Aging Rhesus Macaque Brains: Association With Cognition and MRI-Based Regional Gray Matter Volume. J Comp Neurol 2024; 532:e25670. [PMID: 39315417 PMCID: PMC11451939 DOI: 10.1002/cne.25670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
The brain changes of Alzheimer's disease (AD) include Abeta (Aβ) amyloid plaques ("A"), abnormally phosphorylated tau tangles ("T"), and neurodegeneration ("N"). These have been used to construct in vivo and postmortem diagnostic and staging classifications for evaluating the spectrum of AD in the "ATN" and "ABC" ("B" for Braak tau stage, "C" for Consortium to Establish a Registry for Alzheimer's Disease [CERAD] neuritic plaque density) systems. Another common AD feature involves cerebral amyloid angiopathy (CAA). We report the first experiment to examine relationships among cognition, brain distribution of amyloid plaques, CAA, tau/tangles, and magnetic resonance imaging (MRI)-determined volume changes (as a measure of "N") in the same group of behaviorally characterized nonhuman primates. Both ATN and ABC systems were applied to a group of 32 rhesus macaques aged between 7 and 33 years. When an immunohistochemical method for "T" and "B" was used, some monkeys were "triple positive" on ATN, with a maximum ABC status of A1B2C3. With silver or thioflavin S methods, however, all monkeys were classified as T-negative and B0, indicating the absence of mature neurofibrillary tangles (NFTs) and hence neuropathologically defined AD. Although monkeys at extremes of the ATN and ABC classifications, or with frequent CAA, had significantly lower scores on some cognitive tests, the lack of fully mature NFTs or dementia-consistent cognitive impairment indicates that fully developed AD may not occur in rhesus macaques. There were sex differences noted in the types of histopathology present, and only CAA was significantly related to gray matter volume.
Collapse
Affiliation(s)
- Carol A Barnes
- Departments of Psychology, Neurology and Neuroscience, Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Tucson, Arizona, USA
| | - Michele R Permenter
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Julie A Vogt
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Kewei Chen
- Arizona State University, Tempe, Arizona, USA
| | - Thomas G Beach
- Department of Neuroscience, Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
4
|
Ferrer I. Amyloid-β Pathology Is the Common Nominator Proteinopathy of the Primate Brain Aging. J Alzheimers Dis 2024; 100:S153-S164. [PMID: 39031364 PMCID: PMC11380266 DOI: 10.3233/jad-240389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2024] [Indexed: 07/22/2024]
Abstract
Senile plaques, mainly diffuse, and cerebral amyloid-β (Aβ) angiopathy are prevalent in the aging brain of non-human primates, from lemurs to non-human Hominidae. Aβ but not hyper-phosphorylated tau (HPtau) pathology is the common nominator proteinopathy of non-human primate brain aging. The abundance of Aβ in the aging primate brain is well tolerated, and the impact on cognitive functions is usually limited to particular tasks. In contrast, human brain aging is characterized by the early appearance of HPtau pathology, mainly forming neurofibrillary tangles, dystrophic neurites of neuritic plaques, and neuropil threads, preceding Aβ deposits by several decades and by its severity progressing from selected nuclei of the brain stem, entorhinal cortex, and hippocampus to the limbic system, neocortex, and other brain regions. Neurofibrillary tangles correlate with cognitive impairment and dementia in advanced cases. Aβ pathology is linked in humans to altered membrane protein and lipid composition, particularly involving lipid rafts. Although similar membrane alterations are unknown in non-human primates, membrane senescence is postulated to cause the activated β-amyloidogenic pathway, and Aβ pathology is the prevailing signature of non-human and human primate brain aging.
Collapse
Affiliation(s)
- Isidro Ferrer
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
IWAIDE S, NAKAYAMA Y, CHAMBERS JK, UCHIDA K, NAKAGAWA D, YAMANASHI Y, BANDO H, MURAKAMI T. Senile plaques and phosphorylated tau deposition in a super-aged rhesus monkey (Macaca mulatta). J Vet Med Sci 2023; 85:1296-1300. [PMID: 37821381 PMCID: PMC10788178 DOI: 10.1292/jvms.23-0313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/01/2023] [Indexed: 10/13/2023] Open
Abstract
The brain of a rhesus monkey that died at 43 years of age with symptoms of suspected cognitive dysfunction was analyzed. pathological analyses revealed characteristic Alzheimer's disease-related lesions: the aggregation of amyloid β (Aβ) in the form of senile plaques and phosphorylated tau proteins. We also revealed that Aβ43, which is prone to aggregation and toxicity in humans, is involved in senile plaques in the brain of the rhesus monkey, as well as several other Aβ species. Comparative studies of neuropathology using aged nonhuman primates lack behavioral descriptions compared to human medicine. This case report showed behavioral abnormalities and the detailed pathological changes that may have caused it in a super-aged rhesus monkey.
Collapse
Affiliation(s)
- Susumu IWAIDE
- Laboratory of Veterinary Toxicology, Tokyo University of
Agriculture and Technology, Tokyo, Japan
| | - Yutaro NAKAYAMA
- Laboratory of Veterinary Pathology, The University of Tokyo,
Tokyo, Japan
| | - James K CHAMBERS
- Laboratory of Veterinary Pathology, The University of Tokyo,
Tokyo, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, The University of Tokyo,
Tokyo, Japan
| | | | | | | | - Tomoaki MURAKAMI
- Laboratory of Veterinary Toxicology, Tokyo University of
Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
6
|
Neiworth JJ, Thall ME, Liu S, Leon-Moffly E, Rankin M, LoRusso MA, Thandi S, Garay-Hernandez J. A recognition test in monkeys to differentiate recollection from familiarity memory. Sci Rep 2023; 13:17579. [PMID: 37845334 PMCID: PMC10579227 DOI: 10.1038/s41598-023-44804-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023] Open
Abstract
Episodic memory is memory for experiences within a specific temporal and spatial context. Episodic memories decline early in Alzheimer's Disease (AD). Recollection of episodic memories can fail with both AD and aging, but familiarity and recollection memory uniquely fail in AD. Finding a means to differentiate specific memory failures in animal models is critical for translational research. Four cotton top tamarins participated in an object recognition test. They were exposed to two unique objects placed in a consistent context for 5 daily sessions. Next a delay of 1 day or 1 week was imposed. Subjects' memory of the objects was tested by replacing one of the familiarized objects with a novel one. The tamarins looked longer at the novel object after both delays, an indication of remembering the familiar object. In other tests, the test pair was relocated to a new location or presented at a different time of day. With context changes, tamarins showed greater interest in the novel object after a 1-week delay but not after a 1-day delay. It seems that context changes disrupted their recollection of recent events. But the monkeys showed accurate familiarity memory across context changes with longer delays.
Collapse
|
7
|
Cooper CP, Shafer AT, Armstrong NM, An Y, Erus G, Davatzikos C, Ferrucci L, Rapp PR, Resnick SM. Associations of baseline and longitudinal change in cerebellum volume with age-related changes in verbal learning and memory. Neuroimage 2023; 272:120048. [PMID: 36958620 PMCID: PMC11924273 DOI: 10.1016/j.neuroimage.2023.120048] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023] Open
Abstract
The cerebellum is involved in higher-order cognitive functions, e.g., learning and memory, and is susceptible to age-related atrophy. Yet, the cerebellum's role in age-related cognitive decline remains largely unknown. We investigated cross-sectional and longitudinal associations between cerebellar volume and verbal learning and memory. Linear mixed effects models and partial correlations were used to examine the relationship between changes in cerebellum volumes (total cerebellum, cerebellum white matter [WM], cerebellum hemisphere gray matter [GM], and cerebellum vermis subregions) and changes in verbal learning and memory performance among 549 Baltimore Longitudinal Study of Aging participants (2,292 visits). All models were adjusted by baseline demographic characteristics (age, sex, race, education), and APOE e4 carrier status. In examining associations between change with change, we tested an additional model that included either hippocampal (HC), cuneus, or postcentral gyrus (PoCG) volumes to assess whether cerebellar volumes were uniquely associated with verbal learning and memory. Cross-sectionally, the association of baseline cerebellum GM and WM with baseline verbal learning and memory was age-dependent, with the oldest individuals showing the strongest association between volume and performance. Baseline volume was not significantly associated with change in learning and memory. However, analysis of associations between change in volumes and changes in verbal learning and memory showed that greater declines in verbal memory were associated with greater volume loss in cerebellum white matter, and preserved GM volume in cerebellum vermis lobules VI-VII. The association between decline in verbal memory and decline in cerebellar WM volume remained after adjustment for HC, cuneus, and PoCG volume. Our findings highlight that associations between cerebellum volume and verbal learning and memory are age-dependent and regionally specific.
Collapse
Affiliation(s)
- C'iana P Cooper
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, United States
| | - Andrea T Shafer
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, United States
| | - Nicole M Armstrong
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, United States; Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Yang An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, United States
| | - Guray Erus
- Section of Biomedical Image Analysis, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Christos Davatzikos
- Section of Biomedical Image Analysis, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Peter R Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, United States
| | - Susan M Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224-6825, United States.
| |
Collapse
|
8
|
Wen Y, Zhang L, Li N, Tong A, Zhao C. Nutritional assessment models for Alzheimer's disease: Advances and perspectives. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Affiliation(s)
- Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry Faculty of Sciences Ourense Spain
| | - Lizhu Zhang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Aijun Tong
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
9
|
Karal-ogly DD, Shumeev AN, Keburiya VV, Mintel MV, Rybtsov SA. Age-Related Changes in the Clustering of Blood Populations in Cynomolgus Monkeys Depend on Sex and Immune Status. Life (Basel) 2023; 13:life13020316. [PMID: 36836673 PMCID: PMC9965083 DOI: 10.3390/life13020316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Non-anthropoid primates cynomolgus monkeys (Macaca fascicularis), also known as crab-eating macaques, are increasingly used in biomedical and preclinical studies due to their evolutionary proximity to humans, sharing similar diets, infectious and senile diseases. Age-related changes and sexual dimorphism of the immune system of C. monkeys have not been sufficiently characterized in literature, though age and sex differences affect the course of diseases and sensitivity to medications. Aging in C. monkeys is accompanied by an increase in CD3+CD4+CD8+ (DP-T) cells, plasma B-cells, and a decrease in platelets. Erythromyeloid bias has also been noticed in older animals. There was an increase in eosinophils, haematocrit (HCT) and haemoglobin concentration (HGB). Senile decline in the function of the immune system had sex differences. An increase in the number of monocytes, cytotoxic lymphocytes (CTL) and a decrease in the T-helper population were more pronounced in older females. A significant reduction in the number of B-cells and activated T-cells was detected in males only. A moderate correlation with the regression model of aging was established for DP-T, HCT and HGB. The reduction in the B cells count in males and the increase in CTL level in females are moderately correlated with age. Other blood cell populations did not show significant correlations in the regression models due to their high sample variability. The novel cell population CD3-CD20loCD16/CD56+, presumably NK-cells subset, was revealed. This cell population demonstrated an increase trend with age in both sexes. Population-statistical age norms for different sexes for young and very old macaques were established. The blood population clusters associated with sex and immune status in older animals were also identified.
Collapse
Affiliation(s)
| | - Alexander N. Shumeev
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
| | | | - Marina V. Mintel
- The Research Institute of Medical Primatology, 354383 Sochi, Russia
| | - Stanislav A. Rybtsov
- Centre for Cell Technology and Immunology, Sirius University of Science and Technology, 354340 Sochi, Russia
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH8 9YL, UK
- Correspondence:
| |
Collapse
|
10
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|
11
|
Hamamoto Y, Yu Y, Asada R, Mizuno S, Hasegawa D. Age-related brain atrophy in cats without apparent neurological and behavioral signs using voxel-based morphometry. Front Vet Sci 2022; 9:1071002. [PMID: 36504872 PMCID: PMC9729775 DOI: 10.3389/fvets.2022.1071002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Brain atrophy is observed with aging and may cause cognitive decline or dysfunction. Aging cats may demonstrate behavioral changes related to cognitive dysfunction. In the clinical veterinary field, although the conventional region of interest method by manual or semiauto tracing on magnetic resonance imaging is used to detect atrophy of regional structures, such as the hippocampus, it is difficult to assess atrophy globally. Voxel-based morphometry (VBM) has been developed to detect global and regional abnormalities in humans. The purpose of the present study investigates whether the feline brain volume decreases with aging using VBM analysis. Materials A total of 65 cats, aged 17-200 months, without apparent neurological and behavioral signs were included in the statistical analysis. Results We observed that the gray matter in the bilateral parietal lobes was decreased significantly with aging. The regions that showed decreased volume included the right postcruciate, cingulate gyrus, rostral suprasylvian/ectosylvian gyri, and the left postcruciate gyrus. No significant reduction in white matter was observed. Together, our results show that age-related brain atrophy can be detected using VBM analysis. Discussion The age-related atrophy of the parietal cortex may not cause neurological and behavioral signs in cats. Therefore, veterinarians should consider age when assessing the relation between morphometric and functional abnormalities of the parietal cortex in cats.
Collapse
Affiliation(s)
- Yuji Hamamoto
- Veterinary Medical Teaching Hospital, Nippon Veterinary and Life Science University, Musashino, Japan,Laboratory of Veterinary Radiology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino, Japan,The Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Musashino, Japan,*Correspondence: Yuji Hamamoto
| | - Yoshihiko Yu
- Laboratory of Veterinary Radiology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino, Japan
| | - Rikako Asada
- Laboratory of Veterinary Radiology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino, Japan
| | - Satoshi Mizuno
- Laboratory of Veterinary Radiology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino, Japan
| | - Daisuke Hasegawa
- Laboratory of Veterinary Radiology, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino, Japan,The Research Center for Animal Life Science, Nippon Veterinary and Life Science University, Musashino, Japan
| |
Collapse
|
12
|
Képes Z, Barkóczi A, Szabó JP, Kálmán-Szabó I, Arató V, Jószai I, Deák Á, Kertész I, Hajdu I, Trencsényi G. In Vivo Preclinical Assessment of β-Amyloid-Affine [ 11C]C-PIB Accumulation in Aluminium-Induced Alzheimer's Disease-Resembling Hypercholesterinaemic Rat Model. Int J Mol Sci 2022; 23:ijms232213950. [PMID: 36430429 PMCID: PMC9695619 DOI: 10.3390/ijms232213950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Aluminum (Al) excess and hypercholesterinaemia are established risks of Alzheimer's disease (AD). The aim of this study was to establish an AD-resembling hypercholesterinaemic animal model-with the involvement of 8 week and 48 week-old Fischer-344 rats-by Al administration for the safe and rapid verification of β-amyloid-targeted positron emission tomography (PET) radiopharmaceuticals. Measurement of lipid parameters and β-amyloid-affine [11C]C-Pittsburgh Compound B ([11C]C-PIB) PET examinations were performed. Compared with the control, the significantly elevated cholesterol and LDL levels of the rats receiving the cholesterol-rich diet support the development of hypercholesterinaemia (p ≤ 0.01). In the older cohort, a notably increased age-related radiopharmaceutical accumulation was registered compared to in the young (p ≤ 0.05; p ≤ 0.01). A monotherapy-induced slight elevation of mean standardised uptake values (SUVmean) was statistically not significant; however, adult rats administered a combined diet expressed remarkable SUVmean increment compared to the adult control (SUVmean: from 0.78 ± 0.16 to 1.99 ± 0.28). One and two months after restoration to normal diet, the cerebral [11C]C-PIB accumulation of AD-mimicking animals decreased by half and a third, respectively, to the baseline value. The proposed in vivo Al-induced AD-resembling animal system seems to be adequate for the understanding of AD neuropathology and future drug testing and radiopharmaceutical development.
Collapse
Affiliation(s)
- Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Correspondence:
| | - Alexandra Barkóczi
- Department of Urology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Judit P. Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ibolya Kálmán-Szabó
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Viktória Arató
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Jószai
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Ádám Deák
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Department of Operative Techniques and Surgical Research, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Kertész
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - István Hajdu
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Doctoral School of Clinical Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| |
Collapse
|
13
|
Freire-Cobo C, Edler MK, Varghese M, Munger E, Laffey J, Raia S, In SS, Wicinski B, Medalla M, Perez SE, Mufson EJ, Erwin JM, Guevara EE, Sherwood CC, Luebke JI, Lacreuse A, Raghanti MA, Hof PR. Comparative neuropathology in aging primates: A perspective. Am J Primatol 2021; 83:e23299. [PMID: 34255875 PMCID: PMC8551009 DOI: 10.1002/ajp.23299] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/27/2022]
Abstract
While humans exhibit a significant degree of neuropathological changes associated with deficits in cognitive and memory functions during aging, non-human primates (NHP) present with more variable expressions of pathological alterations among individuals and species. As such, NHP with long life expectancy in captivity offer an opportunity to study brain senescence in the absence of the typical cellular pathology caused by age-related neurodegenerative illnesses commonly seen in humans. Age-related changes at neuronal population, single cell, and synaptic levels have been well documented in macaques and marmosets, while age-related and Alzheimer's disease-like neuropathology has been characterized in additional species including lemurs as well as great apes. We present a comparative overview of existing neuropathologic observations across the primate order, including classic age-related changes such as cell loss, amyloid deposition, amyloid angiopathy, and tau accumulation. We also review existing cellular and ultrastructural data on neuronal changes, such as dendritic attrition and spine alterations, synaptic loss and pathology, and axonal and myelin pathology, and discuss their repercussions on cellular and systems function and cognition.
Collapse
Affiliation(s)
- Carmen Freire-Cobo
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Melissa K Edler
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emily Munger
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Jessie Laffey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sophia Raia
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Selena S In
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bridget Wicinski
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maria Medalla
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Joseph M Erwin
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Elaine E Guevara
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, USA
| | - Chet C Sherwood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Jennifer I Luebke
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Agnès Lacreuse
- Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Mary A Raghanti
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
14
|
Cooper CP, Shafer AT, Armstrong NM, Rossi SL, Young J, Herold C, Gu H, Yang Y, Stein EA, Resnick SM, Rapp PR. Recognition Memory is Associated with Distinct Patterns of Regional Gray Matter Volumes in Young and Aged Monkeys. Cereb Cortex 2021; 32:933-948. [PMID: 34448810 DOI: 10.1093/cercor/bhab257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 11/13/2022] Open
Abstract
Cognitive aging varies tremendously across individuals and is often accompanied by regionally specific reductions in gray matter (GM) volume, even in the absence of disease. Rhesus monkeys provide a primate model unconfounded by advanced neurodegenerative disease, and the current study used a recognition memory test (delayed non-matching to sample; DNMS) in conjunction with structural imaging and voxel-based morphometry (VBM) to characterize age-related differences in GM volume and brain-behavior relationships. Consistent with expectations from a long history of neuropsychological research, DNMS performance in young animals prominently correlated with the volume of multiple structures in the medial temporal lobe memory system. Less anticipated correlations were also observed in the cingulate and cerebellum. In aged monkeys, significant volumetric correlations with DNMS performance were largely restricted to the prefrontal cortex and striatum. Importantly, interaction effects in an omnibus analysis directly confirmed that the associations between volume and task performance in the MTL and prefrontal cortex are age-dependent. These results demonstrate that the regional distribution of GM volumes coupled with DNMS performance changes across the lifespan, consistent with the perspective that the aged primate brain retains a substantial capacity for structural reorganization.
Collapse
Affiliation(s)
- C'iana P Cooper
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, United States
| | - Andrea T Shafer
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 02903, United States
| | - Nicole M Armstrong
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Sharyn L Rossi
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, United States
| | - Jennifer Young
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, United States
| | - Christa Herold
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, United States
| | - Hong Gu
- Magnetic Resonance Imaging and Spectroscopy Section, Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Yihong Yang
- Magnetic Resonance Imaging and Spectroscopy Section, Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Elliot A Stein
- Cognitive and Affective Neuroscience of Addiction Section, Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Susan M Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 02903, United States
| | - Peter R Rapp
- Neurocognitive Aging Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224, United States
| |
Collapse
|
15
|
Koinuma S, Shimozawa N, Yasutomi Y, Kimura N. Aging induces abnormal accumulation of Aβ in extracellular vesicle and/or intraluminal membrane vesicle-rich fractions in nonhuman primate brain. Neurobiol Aging 2021; 106:268-281. [PMID: 34329965 DOI: 10.1016/j.neurobiolaging.2021.06.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 06/10/2021] [Accepted: 06/28/2021] [Indexed: 12/30/2022]
Abstract
Aβ metabolism in the brain is mediated by endocytosis, one part of the intracellular membrane trafficking system. We previously showed that aging attenuates the interaction of dynein with dynactin, which disrupts the endosomal/lysosomal trafficking pathway involved in Aβ metabolism, resulting in intracellular accumulation of Aβ. Several studies have shown that in Alzheimer's disease (AD), intraneuronal accumulation of Aβ precedes extracellular Aβ depositions. However, it is unclear what accounts for this transition from intracellular to extracellular depositions. Accumulating evidence suggest that autophagy has an important role in AD pathology, and we observed that autophagy-related protein levels began to decrease before amyloid plaque formation in cynomolgus monkey brains. Surprisingly, experimental induction of autophagosome formation in Neuro2a cells significantly increased intracellular Aβ and decreased extracellular release of Aβ, accompanied by the prominent reduction of extracellular vesicle (EV) secretion. RNAi study confirmed that EV secretion affected intracellular and extracellular Aβ levels, and siRNA-induced downregulation of autophagosome formation enhanced EV secretion to ameliorate intracellular Aβ accumulation induced by dynein knockdown. In aged cynomolgus monkeys, Aβ levels in EV/intraluminal membrane vesicle (ILV)-rich fractions isolated from temporal lobe parenchyma were drastically increased. Moreover, EV/ILV marker proteins overlapped spatially with amyloid plaques. These findings suggest that EV would be an important carrier of Aβ in brain and abnormal accumulation of Aβ in EVs/ILVs may be involved in the transition of age-dependent Aβ pathology.
Collapse
Affiliation(s)
- Shingo Koinuma
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, Obu, Aichi, Japan; Division of Biosignaling, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan
| | - Nobuyuki Kimura
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, Obu, Aichi, Japan; Laboratory of Experimental Animals, Research and Development Management Center, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan; Tsukuba Primate Research Center, National Institutes of Biomedical Innovation, Health and Nutrition, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
16
|
Iwaide S, Yanai T, Murakami T. Proteopathic lesions in the brain of a super-aged chimpanzee (Pan troglodytes). J Med Primatol 2021; 50:222-224. [PMID: 34096618 DOI: 10.1111/jmp.12530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/01/2021] [Accepted: 05/17/2021] [Indexed: 11/29/2022]
Abstract
The brain of a chimpanzee estimated to be 68 years old, the oldest reported so far, has been examined. Pathological analyses revealed the formation of mild tau-positive neuritic clusters and cytoplasmic α-synuclein aggregates, in addition to severe cerebral amyloid angiopathy and diffuse plaques, but no tangle lesions were observed.
Collapse
Affiliation(s)
- Susumu Iwaide
- Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Tokuma Yanai
- Hiwa Museum for Natural History, Hiroshima, Japan
| | - Tomoaki Murakami
- Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| |
Collapse
|
17
|
Edler MK, Mhatre-Winters I, Richardson JR. Microglia in Aging and Alzheimer's Disease: A Comparative Species Review. Cells 2021; 10:1138. [PMID: 34066847 PMCID: PMC8150617 DOI: 10.3390/cells10051138] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are the primary immune cells of the central nervous system that help nourish and support neurons, clear debris, and respond to foreign stimuli. Greatly impacted by their environment, microglia go through rapid changes in cell shape, gene expression, and functional behavior during states of infection, trauma, and neurodegeneration. Aging also has a profound effect on microglia, leading to chronic inflammation and an increase in the brain's susceptibility to neurodegenerative processes that occur in Alzheimer's disease. Despite the scientific community's growing knowledge in the field of neuroinflammation, the overall success rate of drug treatment for age-related and neurodegenerative diseases remains incredibly low. Potential reasons for the lack of translation from animal models to the clinic include the use of a single species model, an assumption of similarity in humans, and ignoring contradictory data or information from other species. To aid in the selection of validated and predictive animal models and to bridge the translational gap, this review evaluates similarities and differences among species in microglial activation and density, morphology and phenotype, cytokine expression, phagocytosis, and production of oxidative species in aging and Alzheimer's disease.
Collapse
Affiliation(s)
- Melissa K. Edler
- Department of Anthropology, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH 44240, USA;
| | - Isha Mhatre-Winters
- School of Biomedical Sciences, College of Arts and Sciences, Kent State University, Kent, OH 44240, USA;
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| | - Jason R. Richardson
- Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
18
|
Li ZH, He XP, Li H, He RQ, Hu XT. Age-associated changes in amyloid-β and formaldehyde concentrations in cerebrospinal fluid of rhesus monkeys. Zool Res 2020; 41:444-448. [PMID: 32543791 PMCID: PMC7340526 DOI: 10.24272/j.issn.2095-8137.2020.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Zhen-Hui Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,Kunming College of Life Science, University of the Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Xia-Ping He
- School of Basic Medical Sciences, Dali University, Dali, Yunnan 671000, China
| | - Hao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Rong-Qiao He
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China
| | - Xin-Tian Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.,CAS Center for Excellence in Brain Science, Chinese Academy of Sciences, Shanghai 200031, China.,Kunming Primate Research Center and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Chinese Academy of Sciences, Kunming, Yunnan 650223, China. E-mail:
| |
Collapse
|
19
|
Zeiss CJ. Utility of spontaneous animal models of Alzheimer’s disease in preclinical efficacy studies. Cell Tissue Res 2020; 380:273-286. [DOI: 10.1007/s00441-020-03198-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
|
20
|
Li H, Zhang L, Qin C. Current state of research on non-human primate models of Alzheimer's disease. Animal Model Exp Med 2019; 2:227-238. [PMID: 31942555 PMCID: PMC6930996 DOI: 10.1002/ame2.12092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
With the increasingly serious aging of the global population, dementia has already become a severe clinical challenge on a global scale. Dementia caused by Alzheimer's disease (AD) is the most common form of dementia observed in the elderly, but its pathogenetic mechanism has still not been fully elucidated. Furthermore, no effective treatment strategy has been developed to date, despite considerable efforts. This can be mainly attributed to the paucity of animal models of AD that are sufficiently similar to humans. Among the presently established animal models, non-human primates share the closest relationship with humans, and their neural anatomy and neurobiology share highly similar characteristics with those of humans. Thus, there is no doubt that these play an irreplaceable role in AD research. Considering this, the present literature on non-human primate models of AD was reviewed to provide a theoretical basis for future research.
Collapse
Affiliation(s)
- Hong‐Wei Li
- NHC Key Laboratory of Human Disease Comparative MedicinePeking Union Medical College (PUMC)BeijingChina
- Key Laboratory of Human Diseases Animal ModelState Administration of Traditional Chinese MedicinePeking Union Medical College (PUMC)BeijingChina
- The Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)Peking Union Medical College (PUMC)BeijingChina
- Ministry of HealthComparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative MedicinePeking Union Medical College (PUMC)BeijingChina
- Key Laboratory of Human Diseases Animal ModelState Administration of Traditional Chinese MedicinePeking Union Medical College (PUMC)BeijingChina
- The Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)Peking Union Medical College (PUMC)BeijingChina
- Ministry of HealthComparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative MedicinePeking Union Medical College (PUMC)BeijingChina
- Key Laboratory of Human Diseases Animal ModelState Administration of Traditional Chinese MedicinePeking Union Medical College (PUMC)BeijingChina
- The Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)Peking Union Medical College (PUMC)BeijingChina
- Ministry of HealthComparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| |
Collapse
|
21
|
Ouyang F, Chen X, Chen Y, Liang J, Chen Y, Lu T, Huang W, Zeng J. Neuronal loss without amyloid-β deposits in the thalamus and hippocampus in the late period after middle cerebral artery occlusion in cynomolgus monkeys. Brain Pathol 2019; 30:165-178. [PMID: 31278793 DOI: 10.1111/bpa.12764] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/01/2019] [Indexed: 11/30/2022] Open
Abstract
Conflicting evidence exists regarding whether focal cerebral infarction contributes to cerebral amyloid-β (Aβ) deposition, as observed in Alzheimer's disease. In this study, we aimed to evaluate the presence of Aβ deposits in the ipsilateral thalamus and hippocampus 12 months post-stroke in non-human primates, whose brains are structurally and functionally similar to that of humans. Four young male cynomolgus monkeys were subjected to unilateral permanent middle cerebral artery occlusion (MCAO), and another four sham-operated monkeys served as controls. All monkeys underwent magnetic resonance imaging examination on post-operative day 7 to assess the location and size of the infarction. The numbers of neurons, astrocytes, microglia and the Aβ load in the non-affected thalamus and hippocampus ipsilaterally remote from infarct foci were examined immunohistochemically at sacrifice 12 months after operation. Thioflavin S and Congo Red stainings were used to identify amyloid deposits. Multiple Aβ antibodies recognizing both the N-terminal and C-terminal epitopes of Aβ peptides were used to avoid antibody cross-reactivity. Aβ levels in cerebrospinal fluid (CSF) and plasma were examined using enzyme-linked immunosorbent assay. The initial infarct was restricted to the left temporal, parietal, insular cortex and the subcortical white matter, while the thalamus and hippocampus remained intact. Of note, there were fewer neurons and more glia in the ipsilateral thalamus and hippocampus in the MCAO group at 12 months post-stroke compared to the control group (all P < 0.05). However, there was no sign of extracellular Aβ plaques in the thalamus or hippocampus. No statistically significant difference was found in CSF or plasma levels of Aβ40 , Aβ42 or the Aβ40 /Aβ42 ratio between the two groups (P > 0.05). These results suggest that significant secondary neuronal loss and reactive gliosis occur in the non-affected thalamus and hippocampus without Aβ deposits in the late period after MCAO in non-human primates.
Collapse
Affiliation(s)
- Fubing Ouyang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinran Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yonghong Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Liang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yicong Chen
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Lu
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weixian Huang
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinsheng Zeng
- Department of Neurology and Stroke Center, National Key Clinical Department and Key Discipline of Neurology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
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
|
23
|
Edler MK, Sherwood CC, Meindl RS, Hopkins WD, Ely JJ, Erwin JM, Mufson EJ, Hof PR, Raghanti MA. Aged chimpanzees exhibit pathologic hallmarks of Alzheimer's disease. Neurobiol Aging 2017; 59:107-120. [PMID: 28888720 PMCID: PMC6343147 DOI: 10.1016/j.neurobiolaging.2017.07.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a uniquely human brain disorder characterized by the accumulation of amyloid-beta protein (Aβ) into extracellular plaques, neurofibrillary tangles (NFT) made from intracellular, abnormally phosphorylated tau, and selective neuronal loss. We analyzed a large group of aged chimpanzees (n = 20, age 37-62 years) for evidence of Aβ and tau lesions in brain regions affected by AD in humans. Aβ was observed in plaques and blood vessels, and tau lesions were found in the form of pretangles, NFT, and tau-immunoreactive neuritic clusters. Aβ deposition was higher in vessels than in plaques and correlated with increases in tau lesions, suggesting that amyloid build-up in the brain's microvasculature precedes plaque formation in chimpanzees. Age was correlated to greater volumes of Aβ plaques and vessels. Tangle pathology was observed in individuals that exhibited plaques and moderate or severe cerebral amyloid angiopathy, a condition in which amyloid accumulates in the brain's vasculature. Amyloid and tau pathology in aged chimpanzees suggests these AD lesions are not specific to the human brain.
Collapse
Affiliation(s)
- Melissa K Edler
- School of Biomedical Sciences, Kent State University, Kent, OH, USA; Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Richard S Meindl
- Department of Anthropology, Kent State University, Kent, OH, USA
| | - William D Hopkins
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA; Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | - Joseph M Erwin
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Elliott J Mufson
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; New York Consortium for Evolutionary Primatology, New York, NY, USA
| | - Mary Ann Raghanti
- School of Biomedical Sciences, Kent State University, Kent, OH, USA; Department of Anthropology, Kent State University, Kent, OH, USA
| |
Collapse
|
24
|
Toufexis D, King SB, Michopoulos V. Socially Housed Female Macaques: a Translational Model for the Interaction of Chronic Stress and Estrogen in Aging. Curr Psychiatry Rep 2017; 19:78. [PMID: 28905316 DOI: 10.1007/s11920-017-0833-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Estrogen's role in cognitive aging remains unclear. Despite evidence implicating stress in pathological aging, the interaction of stress with estrogen on cognition in older women has received little attention, and few animal models exist with which to examine this interaction. RECENT FINDINGS We present evidence that aging socially subordinate female macaques that experience chronic psychosocial stress constitute a suitable model to investigate this. First, we review studies showing that estrogen modulates cognition in animal models, as well as studies demonstrating that estrogen's action on certain types of cognition is impaired by stress. Next, we discuss data showing that middle-aged socially subordinate female macaques exhibit distinct stress-induced phenotypes, and review our investigations indicating that estrogen modulates behavior and physiology differently in subordinate female monkeys. We conclude that socially housed female macaques represent a translational animal model for investigating the interplay of chronic stress and estrogen on cognitive aging in women.
Collapse
Affiliation(s)
- Donna Toufexis
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA.,Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - S Bradley King
- Department of Psychological Science, The University of Vermont, Burlington, VT, USA
| | - Vasiliki Michopoulos
- Division of Development and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA, USA. .,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
25
|
Arendt T, Stieler J, Ueberham U. Is sporadic Alzheimer's disease a developmental disorder? J Neurochem 2017; 143:396-408. [PMID: 28397252 DOI: 10.1111/jnc.14036] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 03/29/2017] [Accepted: 04/06/2017] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of higher age that specifically occurs in human. Its clinical phase, characterized by a decline in physiological, psychological, and social functioning, is preceded by a long clinically silent phase of at least several decades that might perhaps even start very early in life. Overall, key functional abilities in AD patients decline in reverse order of the development of these abilities during normal childhood and adolescence. Early symptoms of AD, thus, typically affect mental functions that have been acquired only during very recent hominid evolution and as such are specific to human. Neurofibrillar degeneration, a typical neuropathological lesion of the disease and one of the most robust pathological correlates of cognitive impairment, is rarely seen in non-primate mammals and even non-human primates hardly develop a pathology comparable to those seen in AD patients. Neurofibrillar degeneration is not randomly distributed throughout the AD brain. It preferentially affects brain areas that become increasingly predominant during the evolutionary process of encephalization. During progression of the disease, it affects cortical areas in a stereotypic sequence that inversely recapitulates ontogenetic brain development. The specific distribution of cortical pathology in AD, moreover, appears to be determined by the modular organization of the cerebral cortex which basically is a structural reflection of its ontogeny. Here, we summarize recent evidence that phylogenetic and ontogenetic dimensions of brain structure and function provide the key to our understanding of AD. More recent molecular biological studies of the potential pathogenetic role of a genomic mosaic in the brains of patients with AD might even provide arguments for a developmental origin of AD. This article is part of a series "Beyond Amyloid".
Collapse
Affiliation(s)
- Thomas Arendt
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Jens Stieler
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| | - Uwe Ueberham
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
26
|
Quan Q, Feng J, Lui LT, Shi T, Chu IK. Phosphoproteome of crab-eating macaque cerebral cortex characterized through multidimensional reversed-phase liquid chromatography/mass spectrometry with tandem anion/cation exchange columns. J Chromatogr A 2017; 1498:196-206. [DOI: 10.1016/j.chroma.2017.01.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/13/2017] [Accepted: 01/21/2017] [Indexed: 02/06/2023]
|
27
|
Crimins JL, Wang ACJ, Yuk F, Puri R, Janssen WGM, Hara Y, Rapp PR, Morrison JH. Diverse Synaptic Distributions of G Protein-coupled Estrogen Receptor 1 in Monkey Prefrontal Cortex with Aging and Menopause. Cereb Cortex 2017; 27:2022-2033. [PMID: 26941383 PMCID: PMC5909633 DOI: 10.1093/cercor/bhw050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Age- and menopause-related impairment in working memory mediated by the dorsolateral prefrontal cortex (dlPFC) occurs in humans and nonhuman primates. Long-term cyclic 17β-estradiol treatment rescues cognitive deficits in aged ovariectomized rhesus monkeys while restoring highly plastic synapses. Here we tested whether distributions of G protein-coupled estrogen receptor 1 (GPER1) within monkey layer III dlPFC synapses are sensitive to age and estradiol, and coupled to cognitive function. Ovariectomized young and aged monkeys administered vehicle or estradiol were first tested on a delayed response test of working memory. Then, quantitative serial section immunoelectron microscopy was used to determine the distributions of synaptic GPER1. GPER1-containing nonperforated axospinous synapse density was reduced with age, and partially restored with estrogen treatment. The majority of synapses expressed GPER1, which was predominately localized to presynaptic cytoplasm and mitochondria. GPER1 was also abundant at plasmalemmas, and within cytoplasmic and postsynaptic density (PSD) domains of dendritic spines. GPER1 levels did not differ with age or treatment, and none of the variables examined were tightly associated with cognitive function. However, greater representation of GPER1 subjacent to the PSD accompanied higher synapse density. These data suggest that GPER1 is positioned to support diverse functions key to synaptic plasticity in monkey dlPFC.
Collapse
Affiliation(s)
| | - Athena Ching-Jung Wang
- Linda Crnic Institute for Down Syndrome, University of Colorado School of Medicine, CO 80045, USA
| | - Frank Yuk
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | - Rishi Puri
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | | | - Yuko Hara
- Fishberg Department of Neuroscience and Friedman Brain Institute
| | - Peter R Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224, USA
| | - John H Morrison
- Fishberg Department of Neuroscience and Friedman Brain Institute
- Department of Geriatrics and Palliative Medicine
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- California National Primate Research Center, Davis, CA 95616, USA
- Department of Neurology, School of Medicine, University of California Davis, Davis 95616, USA
| |
Collapse
|
28
|
Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C. Evidence of Tau Hyperphosphorylation and Dystrophic Microglia in the Common Marmoset. Front Aging Neurosci 2016; 8:315. [PMID: 28066237 PMCID: PMC5177639 DOI: 10.3389/fnagi.2016.00315] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/08/2016] [Indexed: 01/22/2023] Open
Abstract
Common marmosets (Callithrix jacchus) have recently gained popularity in biomedical research as models of aging research. Basically, they confer advantages from other non-human primates due to their shorter lifespan with onset of appearance of aging at 8 years. Old marmosets present some markers linked to neurodegeneration in the brain such as amyloid beta (Aβ)1-42 and Aβ1-40. However, there are no studies exploring other cellular markers associated with neurodegenerative diseases in this non-human primate. Using immunohistochemistry, we analyzed brains of male adolescent, adult, old, and aged marmosets. We observed accumulation of Aβ1-40 and Aβ1-42 in the cortex of aged subjects. Tau hyperphosphorylation was already detected in the brain of adolescent animals and increased with aging in a more fibrillary form. Microglia activation was also observed in the aging process, while a dystrophic phenotype accumulates in aged subjects. Interestingly, dystrophic microglia contained hyperphosphorylated tau, but active microglia did not. These results support previous findings regarding microglia dysfunctionality in aging and neurodegenerative diseases as Alzheimer's disease. Further studies should explore the functional consequences of these findings to position this non-human primate as animal model of aging and neurodegeneration.
Collapse
Affiliation(s)
- Juan D Rodriguez-Callejas
- Laboratory of Neuroplasticity and Neurodegeneration, Department of Pharmacology, Center for Research and Advanced Studies (CINVESTAV) Mexico City, Mexico
| | - Eberhard Fuchs
- Clinical Neurobiology Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen Germany
| | - Claudia Perez-Cruz
- Laboratory of Neuroplasticity and Neurodegeneration, Department of Pharmacology, Center for Research and Advanced Studies (CINVESTAV) Mexico City, Mexico
| |
Collapse
|
29
|
Hara H, Ono F, Nakamura S, Matsumoto SE, Jin H, Hattori N, Tabira T. An Oral Aβ Vaccine Using a Recombinant Adeno-Associated Virus Vector in Aged Monkeys: Reduction in Plaque Amyloid and Increase in Aβ Oligomers. J Alzheimers Dis 2016; 54:1047-1059. [DOI: 10.3233/jad-160514] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hideo Hara
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Division of Neurology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Fumiko Ono
- The Corporation for Production and Research of Laboratory Primates, Tsukuba, Ibaraki, Japan
- Faculty of Risk and Crisis Management, Chiba Institute of Science, Shiomi, Choshi, Chiba, Japan
| | - Shinichiro Nakamura
- The Corporation for Production and Research of Laboratory Primates, Tsukuba, Ibaraki, Japan
- Shiga University of Medical Science, Research Center for Animal Life Science, Seta-Tsukinowa, Otsu, Shiga, Japan
| | - Shin-ei Matsumoto
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Haifeng Jin
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Takeshi Tabira
- National Institute for Longevity Sciences, National Center for Geriatrics and Gerontology, Morioka, Obu, Aichi, Japan
- Department of Diagnosis, Prevention and Treatment of Dementia, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
30
|
Reference values of hematology, biochemistry, and blood type in cynomolgus monkeys from cambodia origin. Lab Anim Res 2016; 32:46-55. [PMID: 27051442 PMCID: PMC4816996 DOI: 10.5625/lar.2016.32.1.46] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/14/2015] [Accepted: 03/03/2016] [Indexed: 11/21/2022] Open
Abstract
Cynomolgus monkeys as nonhuman primates are valuable animal models because they have a high level of human gene homology. There are many reference values for hematology and biochemistry of Cynomolgus monkeys that are needed for proper clinical diagnosis and biomedical research conduct. The body weight information and blood type are also key success factors in allogeneic or xenogeneic models. Moreover, the biological parameters could be different according to the origin of the Cynomolgus monkey. However, there are limited references provided, especially of Cambodia origin. In this study, we measured average body weight of 2,518 Cynomolgus monkeys and analyzed hematology and serum biochemistry using 119 males, and determined blood types in 642 monkeys with Cambodia origin. The average body weight of male Cynomolgus monkeys were 2.56±0.345 kg and female group was 2.43±0.330 kg at the age from 2 to 3 years. The male group showed relatively sharp increased average body weight from the 3 to 4 age period compared to the female group. In hematology and biochemistry, it was found that most of the data was similar when compared to other references even though some results showed differences. The ABO blood type result showed that type A, B, AB, and O was approximately 15.6, 33.3, 44.2, and 6.9%, respectively. The main blood type in this facility was B and AB. These biological background references of Cambodia origin could be used to provide important information to researchers who are using them in their biomedical research.
Collapse
|
31
|
Perez SE, Sherwood CC, Cranfield MR, Erwin JM, Mudakikwa A, Hof PR, Mufson EJ. Early Alzheimer's disease-type pathology in the frontal cortex of wild mountain gorillas (Gorilla beringei beringei). Neurobiol Aging 2016; 39:195-201. [PMID: 26923416 PMCID: PMC6317356 DOI: 10.1016/j.neurobiolaging.2015.12.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/18/2015] [Accepted: 12/23/2015] [Indexed: 01/04/2023]
Abstract
Amyloid beta (Aβ) and tau pathology have been described in the brains of captive aged great apes, but the natural progression of these age-related pathologies from wild great apes, including the gorilla, is unknown. In our previous study of Western lowland gorillas (Gorilla gorilla gorilla) who were housed in American Zoos and Aquariums-accredited facilities, we found an age-related increase in Aβ-positive plaques and vasculature, tau-positive astrocytes, oligodendrocyte coiled bodies, and neuritic clusters in the neocortex as well as hippocampus in older animals. Here, we demonstrate that aged wild mountain gorillas (Gorilla beringei beringei), who spent their entire lives in their natural habitat, also display an age-related increase in amyloid precursor protein (APP) and/or Aβ-immunoreactive blood vessels and plaques, but very limited tau pathology, in the frontal cortex. These results indicate that Aβ and tau lesions are age-related events that occur in the brain of gorillas living in captivity and in the wild.
Collapse
Affiliation(s)
- Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA.
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Michael R Cranfield
- Mountain Gorilla Veterinary Project, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Joseph M Erwin
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC, USA
| | - Antoine Mudakikwa
- Rwanda Development Board, Department of Tourism and Conservation, Kigali, Rwanda
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
32
|
Hara Y, Waters EM, McEwen BS, Morrison JH. Estrogen Effects on Cognitive and Synaptic Health Over the Lifecourse. Physiol Rev 2015; 95:785-807. [PMID: 26109339 PMCID: PMC4491541 DOI: 10.1152/physrev.00036.2014] [Citation(s) in RCA: 284] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Estrogen facilitates higher cognitive functions by exerting effects on brain regions such as the prefrontal cortex and hippocampus. Estrogen induces spinogenesis and synaptogenesis in these two brain regions and also initiates a complex set of signal transduction pathways via estrogen receptors (ERs). Along with the classical genomic effects mediated by activation of ER α and ER β, there are membrane-bound ER α, ER β, and G protein-coupled estrogen receptor 1 (GPER1) that can mediate rapid nongenomic effects. All key ERs present throughout the body are also present in synapses of the hippocampus and prefrontal cortex. This review summarizes estrogen actions in the brain from the standpoint of their effects on synapse structure and function, noting also the synergistic role of progesterone. We first begin with a review of ER subtypes in the brain and how their abundance and distributions are altered with aging and estrogen loss (e.g., ovariectomy or menopause) in the rodent, monkey, and human brain. As there is much evidence that estrogen loss induced by menopause can exacerbate the effects of aging on cognitive functions, we then review the clinical trials of hormone replacement therapies and their effectiveness on cognitive symptoms experienced by women. Finally, we summarize studies carried out in nonhuman primate models of age- and menopause-related cognitive decline that are highly relevant for developing effective interventions for menopausal women. Together, we highlight a new understanding of how estrogen affects higher cognitive functions and synaptic health that go well beyond its effects on reproduction.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Elizabeth M Waters
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - Bruce S McEwen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| | - John H Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Friedman Brain Institute, Department of Geriatrics and Palliative Medicine, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; and Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, New York
| |
Collapse
|
33
|
Okabayashi S, Shimozawa N, Yasutomi Y, Yanagisawa K, Kimura N. Diabetes mellitus accelerates Aβ pathology in brain accompanied by enhanced GAβ generation in nonhuman primates. PLoS One 2015; 10:e0117362. [PMID: 25675436 PMCID: PMC4326359 DOI: 10.1371/journal.pone.0117362] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 12/21/2014] [Indexed: 11/18/2022] Open
Abstract
Growing evidence suggests that diabetes mellitus (DM) is one of the strongest risk factors for developing Alzheimer’s disease (AD). However, it remains unclear why DM accelerates AD pathology. In cynomolgus monkeys older than 25 years, senile plaques (SPs) are spontaneously and consistently observed in their brains, and neurofibrillary tangles are present at 32 years of age and older. In laboratory-housed monkeys, obesity is occasionally observed and frequently leads to development of type 2 DM. In the present study, we performed histopathological and biochemical analyses of brain tissue in cynomolgus monkeys with type 2 DM to clarify the relationship between DM and AD pathology. Here, we provide the evidence that DM accelerates Aβ pathology in vivo in nonhuman primates who had not undergone any genetic manipulation. In DM-affected monkey brains, SPs were observed in frontal and temporal lobe cortices, even in monkeys younger than 20 years. Biochemical analyses of brain revealed that the amount of GM1-ganglioside-bound Aβ (GAβ)—the endogenous seed for Aβ fibril formation in the brain—was clearly elevated in DM-affected monkeys. Furthermore, the level of Rab GTPases was also significantly increased in the brains of adult monkeys with DM, almost to the same levels as in aged monkeys. Intraneuronal accumulation of enlarged endosomes was also observed in DM-affected monkeys, suggesting that exacerbated endocytic disturbance may underlie the acceleration of Aβ pathology due to DM.
Collapse
Affiliation(s)
- Sachi Okabayashi
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1–1 Hachimandai, Tsukuba-shi, Ibaraki, 305–0843, Japan
- The Corporation for Production and Research of Laboratory Primates, 1–1 Hachimandai, Tsukuba-shi, Ibaraki, 305–0843, Japan
| | - Nobuhiro Shimozawa
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1–1 Hachimandai, Tsukuba-shi, Ibaraki, 305–0843, Japan
| | - Yasuhiro Yasutomi
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1–1 Hachimandai, Tsukuba-shi, Ibaraki, 305–0843, Japan
| | - Katsuhiko Yanagisawa
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology (NCGG), Gengo 35, Moriika, Obu, Aichi, 474–8511, Japan
| | - Nobuyuki Kimura
- Tsukuba Primate Research Center, National Institute of Biomedical Innovation, 1–1 Hachimandai, Tsukuba-shi, Ibaraki, 305–0843, Japan
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology (NCGG), Gengo 35, Moriika, Obu, Aichi, 474–8511, Japan
- * E-mail:
| |
Collapse
|
34
|
Quantitative expression analysis of APP pathway and tau phosphorylation-related genes in the ICV STZ-induced non-human primate model of sporadic Alzheimer's disease. Int J Mol Sci 2015; 16:2386-402. [PMID: 25622254 PMCID: PMC4346842 DOI: 10.3390/ijms16022386] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 11/17/2022] Open
Abstract
The accumulation and aggregation of misfolded proteins in the brain, such as amyloid-β (Aβ) and hyperphosphorylated tau, is a neuropathological hallmark of Alzheimer's disease (AD). Previously, we developed and validated a novel non-human primate model for sporadic AD (sAD) research using intracerebroventricular administration of streptozotocin (icv STZ). To date, no characterization of AD-related genes in different brain regions has been performed. Therefore, in the current study, the expression of seven amyloid precursor protein (APP) pathway-related and five tau phosphorylation-related genes was investigated by quantitative real-time PCR experiments, using two matched-pair brain samples from control and icv STZ-treated cynomolgus monkeys. The genes showed similar expression patterns within the control and icv STZ-treated groups; however, marked differences in gene expression patterns were observed between the control and icv STZ-treated groups. Remarkably, other than β-secretase (BACE1) and cyclin-dependent kinase 5 (CDK5), all the genes tested showed similar expression patterns in AD models compared to controls, with increased levels in the precuneus and occipital cortex. However, significant changes in gene expression patterns were not detected in the frontal cortex, hippocampus, or posterior cingulate. Based on these results, we conclude that APP may be cleaved via the general metabolic mechanisms of increased α- and γ-secretase levels, and that hyperphosphorylation of tau could be mediated by elevated levels of tau protein kinase, specifically in the precuneus and occipital cortex.
Collapse
|
35
|
Autrey MM, Reamer LA, Mareno MC, Sherwood CC, Herndon JG, Preuss T, Schapiro SJ, Hopkins WD. Age-related effects in the neocortical organization of chimpanzees: gray and white matter volume, cortical thickness, and gyrification. Neuroimage 2014; 101:59-67. [PMID: 24983715 PMCID: PMC4165649 DOI: 10.1016/j.neuroimage.2014.06.053] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 06/03/2014] [Accepted: 06/23/2014] [Indexed: 12/15/2022] Open
Abstract
Among primates, humans exhibit the most profound degree of age-related brain volumetric decline in particular regions, such as the hippocampus and the frontal lobe. Recent studies have shown that our closest living relatives, the chimpanzees, experience little to no volumetric decline in gray and white matter over the adult lifespan. However, these previous studies were limited with a small sample of chimpanzees of the most advanced ages. In the present study, we sought to further test for potential age-related decline in cortical organization in chimpanzees by expanding the sample size of aged chimpanzees. We used the BrainVisa software to measure total brain volume, gray and white matter volumes, gray matter thickness, and gyrification index in a cross-sectional sample of 219 captive chimpanzees (8-53 years old), with 38 subjects being 40 or more years of age. Mean depth and cortical fold opening of 11 major sulci of the chimpanzee brains were also measured. We found that chimpanzees showed increased gyrification with age and a cubic relationship between age and white matter volume. For the association between age and sulcus depth and width, the results were mostly non-significant with the exception of one negative correlation between age and the fronto-orbital sulcus. In short, results showed that chimpanzees exhibit few age-related changes in global cortical organization, sulcus folding and sulcus width. These findings support previous studies and the theory that the age-related changes in the human brain is due to an extended lifespan.
Collapse
Affiliation(s)
- Michelle M Autrey
- Department of Psychology, Agnes Scott College, Decatur, GA 30030, USA
| | - Lisa A Reamer
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Mary Catherine Mareno
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington DC 20052, USA
| | - James G Herndon
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Todd Preuss
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Steve J Schapiro
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - William D Hopkins
- Neuroscience Institute and Language Research Center, Georgia State University, Atlanta, GA 30302, USA; Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| |
Collapse
|
36
|
Rachalski A, Authier S, Bassett L, Pouliot M, Tremblay G, Mongrain V. Sleep electroencephalographic characteristics of the Cynomolgus monkey measured by telemetry. J Sleep Res 2014; 23:619-627. [PMID: 25109588 DOI: 10.1111/jsr.12189] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 05/24/2014] [Indexed: 12/24/2022]
Abstract
Cynomolgus monkeys are widely used as models of diseases and in pre-clinical studies to assess the impact of new pharmacotherapies on brain function and behaviour. However, the time course of electroencephalographic delta activity during sleep, which represents the main marker of sleep intensity associated with recovery during sleep, has never been described in this non-human primate. In this study, telemetry implants were used to record one spontaneous 24-h sleep-wake cycle in four freely-moving Cynomolgus monkeys, and to quantify the time course of electroencephalographic activity during sleep using spectral analysis. Animals presented a diurnal activity pattern interrupted by short naps. During the dark period, most of the time was spent in sleep with non-rapid eye movement sleep/rapid eye movement sleep alternations and sleep consolidation profiles intermediate between rodents and humans. Deep non-rapid eye movement sleep showed a typical predominance at the beginning of the night with decreased propensity in the course of the night, which was accompanied by a progressive increase in rapid eye movement sleep duration. Spectral profiles showed characteristic changes between vigilance states as reported in other mammalian species. Importantly, delta activity also followed the expected time course of variation, showing a build-up with wakefulness duration and dissipation across the night. Thus, Cynomolgus monkeys present typical characteristics of sleep architecture and spectral structure as those observed in other mammalian species including humans, validating the use of telemetry in this non-human primate model for translational sleep studies.
Collapse
Affiliation(s)
- Adeline Rachalski
- Department of Psychiatry, Université de Montréal, Montreal, QC, Canada.,Research Center, Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada
| | - Simon Authier
- CiToxLAB North America, 445 Armand Frappier, Laval, QC, Canada
| | - Leanne Bassett
- CiToxLAB North America, 445 Armand Frappier, Laval, QC, Canada
| | - Mylène Pouliot
- CiToxLAB North America, 445 Armand Frappier, Laval, QC, Canada
| | - Gaétan Tremblay
- Research Center, Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada
| | - Valérie Mongrain
- Research Center, Hôpital du Sacré-Coeur de Montréal, Montreal, QC, Canada.,Department of Neurosciences, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
37
|
Hara Y, Yuk F, Puri R, Janssen WGM, Rapp PR, Morrison JH. Presynaptic mitochondrial morphology in monkey prefrontal cortex correlates with working memory and is improved with estrogen treatment. Proc Natl Acad Sci U S A 2014; 111:486-91. [PMID: 24297907 PMCID: PMC3890848 DOI: 10.1073/pnas.1311310110] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Humans and nonhuman primates are vulnerable to age- and menopause-related decline in working memory, a cognitive function reliant on the energy-demanding recurrent excitation of neurons within Brodmann's Area 46 of the dorsolateral prefrontal cortex (dlPFC). Here, we tested the hypothesis that the number and morphology (straight, curved, or donut-shaped) of mitochondria in dlPFC presynaptic boutons are altered with aging and menopause in rhesus monkeys (Macaca mulatta) and that these metrics correlate with delayed response (DR) accuracy, a well-characterized measure of dlPFC-dependent working memory. Although presynaptic bouton density or size was not significantly different across groups distinguished by age or menses status, DR accuracy correlated positively with the number of total and straight mitochondria per dlPFC bouton. In contrast, DR accuracy correlated inversely with the frequency of boutons containing donut-shaped mitochondria, which exhibited smaller active zone areas and fewer docked synaptic vesicles than those with straight or curved mitochondria. We then examined the effects of estrogen administration to test whether a treatment known to improve working memory influences mitochondrial morphology. Aged ovariectomized monkeys treated with vehicle displayed significant working memory impairment and a concomitant 44% increase in presynaptic donut-shaped mitochondria, both of which were reversed with cyclic estradiol treatment. Together, our data suggest that hormone replacement therapy may benefit cognitive aging, in part by promoting mitochondrial and synaptic health in the dlPFC.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Frank Yuk
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Rishi Puri
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - William G. M. Janssen
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
| | - Peter R. Rapp
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD 21224
| | - John H. Morrison
- Fishberg Department of Neuroscience
- Kastor Neurobiology of Aging Laboratories
- Friedman Brain Institute
- Department of Geriatrics and Palliative Medicine, and
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; and
| |
Collapse
|
38
|
Toledano A, Álvarez M, López-Rodríguez A, Toledano-Díaz A, Fernández-Verdecia C. Does Alzheimer disease exist in all primates? Alzheimer pathology in non-human primates and its pathophysiological implications (II). NEUROLOGÍA (ENGLISH EDITION) 2014. [DOI: 10.1016/j.nrleng.2011.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
39
|
Perez SE, Raghanti MA, Hof PR, Kramer L, Ikonomovic MD, Lacor PN, Erwin JM, Sherwood CC, Mufson EJ. Alzheimer's disease pathology in the neocortex and hippocampus of the western lowland gorilla (Gorilla gorilla gorilla). J Comp Neurol 2013; 521:4318-38. [PMID: 23881733 PMCID: PMC6317365 DOI: 10.1002/cne.23428] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 06/28/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022]
Abstract
The two major histopathologic hallmarks of Alzheimer's disease (AD) are amyloid beta protein (Aβ) plaques and neurofibrillary tangles (NFT). Aβ pathology is a common feature in the aged nonhuman primate brain, whereas NFT are found almost exclusively in humans. Few studies have examined AD-related pathology in great apes, which are the closest phylogenetic relatives of humans. In the present study, we examined Aβ and tau-like lesions in the neocortex and hippocampus of aged male and female western lowland gorillas using immunohistochemistry and histochemistry. Analysis revealed an age-related increase in Aβ-immunoreactive plaques and vasculature in the gorilla brain. Aβ plaques were more abundant in the neocortex and hippocampus of females, whereas Aβ-positive blood vessels were more widespread in male gorillas. Plaques were also Aβ40-, Aβ42-, and Aβ oligomer-immunoreactive, but only weakly thioflavine S- or 6-CN-PiB-positive in both sexes, indicative of the less fibrillar (diffuse) nature of Aβ plaques in gorillas. Although phosphorylated neurofilament immunostaining revealed a few dystrophic neurites and neurons, choline acetyltransferase-immunoreactive fibers were not dystrophic. Neurons stained for the tau marker Alz50 were found in the neocortex and hippocampus of gorillas at all ages. Occasional Alz50-, MC1-, and AT8-immunoreactive astrocyte and oligodendrocyte coiled bodies and neuritic clusters were seen in the neocortex and hippocampus of the oldest gorillas. This study demonstrates the spontaneous presence of both Aβ plaques and tau-like lesions in the neocortex and hippocampus in old male and female western lowland gorillas, placing this species at relevance in the context of AD research.
Collapse
Affiliation(s)
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, Ohio 44242
- Cleveland Metroparks Zoo, Cleveland, Ohio 44109
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | | | - Milos D. Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pennsylvania 15213
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pennsylvania 15213
| | - Pascale N. Lacor
- Neurobiology Department and Cognitive Neurology and Alzheimer’s Disease Center, Northwestern University, Evanston, Illinois 60208
| | - Joseph M. Erwin
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | - Chet C. Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052
| | | |
Collapse
|
40
|
Delayed response task performance as a function of age in cynomolgus monkeys (Macaca fascicularis). Primates 2013; 55:259-67. [PMID: 24248474 PMCID: PMC3973945 DOI: 10.1007/s10329-013-0397-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/25/2013] [Indexed: 11/05/2022]
Abstract
We compared delayed response task performance in young, middle-aged, and old cynomolgus monkeys using three memory tests that have been used with non-human primates. Eighteen cynomolgus monkeys—6 young (4–9 years), 6 middle-aged (10–19 years), and 6 old (above 20 years)—were tested. In general, the old monkeys scored significantly worse than did the animals in the two other age groups. Longer delays between stimulus presentation and response increased the performance differences between the old and younger monkeys. The old monkeys in particular showed signs of impaired visuo-spatial memory and deteriorated memory consolidation and executive functioning. These results add to the body of evidence supporting the utility of Macaca fascicularis in studies of cognition and as a potential translational model for age-associated memory impairment/dementia-related disorders.
Collapse
|
41
|
Aerobic glycolysis in the primate brain: reconsidering the implications for growth and maintenance. Brain Struct Funct 2013; 219:1149-67. [DOI: 10.1007/s00429-013-0662-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 10/15/2013] [Indexed: 12/15/2022]
|
42
|
Kuwajima M, Spacek J, Harris KM. Beyond counts and shapes: studying pathology of dendritic spines in the context of the surrounding neuropil through serial section electron microscopy. Neuroscience 2013; 251:75-89. [PMID: 22561733 PMCID: PMC3535574 DOI: 10.1016/j.neuroscience.2012.04.061] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 04/16/2012] [Accepted: 04/20/2012] [Indexed: 02/06/2023]
Abstract
Because dendritic spines are the sites of excitatory synapses, pathological changes in spine morphology should be considered as part of pathological changes in neuronal circuitry in the forms of synaptic connections and connectivity strength. In the past, spine pathology has usually been measured by changes in their number or shape. A more complete understanding of spine pathology requires visualization at the nanometer level to analyze how the changes in number and size affect their presynaptic partners and associated astrocytic processes, as well as organelles and other intracellular structures. Currently, serial section electron microscopy (ssEM) offers the best approach to address this issue because of its ability to image the volume of brain tissue at the nanometer resolution. Renewed interest in ssEM has led to recent technological advances in imaging techniques and improvements in computational tools indispensable for three-dimensional analyses of brain tissue volumes. Here we consider the small but growing literature that has used ssEM analysis to unravel ultrastructural changes in neuropil including dendritic spines. These findings have implications in altered synaptic connectivity and cell biological processes involved in neuropathology, and serve as anatomical substrates for understanding changes in network activity that may underlie clinical symptoms.
Collapse
Affiliation(s)
- Masaaki Kuwajima
- Center for Learning and Memory, The University of Texas at Austin
| | - Josef Spacek
- Charles University Prague, Faculty of Medicine in Hradec Kralove, Czech Republic
| | - Kristen M. Harris
- Center for Learning and Memory, The University of Texas at Austin
- Section of Neurobiology, The University of Texas at Austin
| |
Collapse
|
43
|
Guilarte TR. Manganese neurotoxicity: new perspectives from behavioral, neuroimaging, and neuropathological studies in humans and non-human primates. Front Aging Neurosci 2013; 5:23. [PMID: 23805100 PMCID: PMC3690350 DOI: 10.3389/fnagi.2013.00023] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/05/2013] [Indexed: 01/10/2023] Open
Abstract
Manganese (Mn) is an essential metal and has important physiological functions for human health. However, exposure to excess levels of Mn in occupational settings or from environmental sources has been associated with a neurological syndrome comprising cognitive deficits, neuropsychological abnormalities and parkinsonism. Historically, studies on the effects of Mn in humans and experimental animals have been concerned with effects on the basal ganglia and the dopaminergic system as it relates to movement abnormalities. However, emerging studies are beginning to provide significant evidence of Mn effects on cortical structures and cognitive function at lower levels than previously recognized. This review advances new knowledge of putative mechanisms by which exposure to excess levels of Mn alters neurobiological systems and produces neurological deficits not only in the basal ganglia but also in the cerebral cortex. The emerging evidence suggests that working memory is significantly affected by chronic Mn exposure and this may be mediated by alterations in brain structures associated with the working memory network including the caudate nucleus in the striatum, frontal cortex and parietal cortex. Dysregulation of the dopaminergic system may play an important role in both the movement abnormalities as well as the neuropsychiatric and cognitive function deficits that have been described in humans and non-human primates exposed to Mn.
Collapse
Affiliation(s)
- Tomás R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University New York, NY, USA
| |
Collapse
|
44
|
Age- and sex-based hematological and biochemical parameters for Macaca fascicularis. PLoS One 2013; 8:e64892. [PMID: 23762263 PMCID: PMC3677909 DOI: 10.1371/journal.pone.0064892] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/19/2013] [Indexed: 11/30/2022] Open
Abstract
Background The cynomolgus monkey (Macaca fascicularis) has been increasingly used in biomedical research, making knowledge of its blood-based parameters essential to support the selection of healthy subjects and its use in preclinical research. As age and sex affect these blood-based parameters, it is important to establish baseline indices for these parameters on an age and sex basis and determine the effects of age and sex on these indices. Methods A total of 917 cynomolgus monkeys (374 males and 543 females) were selected and segregated by age (five groups) and sex. A total of 30 hematological and 22 biochemical parameters were measured, and the effects of age and sex were analyzed. Results Baseline indices for hematological and biochemical parameters were separately established by age and sex. Significant effects by age, sex, and age-sex interaction were observed in a number of blood parameters. In the 49–60 months and 61–72 months age groups, red blood cell count, hemoglobulin, and hematocrit showed significantly lower values (P<0.01) in females than males. Serum alkaline phosphatase varied with age in both sexes (P<0.01) and was significantly higher in females than males (P<0.05) in the groups aged 13–24 months and 25–36 months; however, in the three groups aged over 25–36 months, serum alkaline phosphatase was significantly lower in females than males (P<0.01). Creatinine concentration increased with age (P<0.01) in all age groups; specifically in the groups aged 49–60 months and 61–72 months, creatinine was significantly higher (P<0.01) in males than females. Total protein and globulin both increased with age (P<0.01). Conclusion The baseline values of hematological and biochemical parameters reported herein establish reference indices of blood-based parameters in the cynomolgus monkey by age and sex, thereby aiding researchers in selecting healthy subjects and evaluating preclinical studies using this species.
Collapse
|
45
|
Darusman H, Sajuthi D, Kalliokoski O, Jacobsen K, Call J, Schapiro S, Gjedde A, Abelson K, Hau J. Correlations between serum levels of beta amyloid, cerebrospinal levels of tau and phospho tau, and delayed response tasks in young and aged cynomolgus monkeys (Macaca fascicularis
). J Med Primatol 2013; 42:137-46. [DOI: 10.1111/jmp.12044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2013] [Indexed: 12/20/2022]
Affiliation(s)
- H.S. Darusman
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
- Department of Anatomy; Physiology and Pharmacology; Faculty of Veterinary Medicine; Bogor Agricultural University; Bogor Indonesia
| | - D. Sajuthi
- Primate Research Center; Bogor Agricultural University; Bogor Indonesia
| | - O. Kalliokoski
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
| | - K.R. Jacobsen
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
| | - J. Call
- Max Planck Institute of Evolutionary Anthropology; Leipzig Germany
| | - S.J. Schapiro
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
- Department of Veterinary Sciences; The University of Texas MD Andersson Cancer Center; Bastrop TX USA
| | - A. Gjedde
- Department of Neuroscience and Pharmacology; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
- Center for Functionally Integrative Neuroscience; University of Aarhus; Aarhus Denmark
- Department of Radiology and Radiological Science; Johns Hopkins University; Baltimore MD USA
| | - K.S.P. Abelson
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
| | - J. Hau
- Department of Experimental Medicine; Faculty of Health Science; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
46
|
Hara Y, Rapp PR, Morrison JH. Neuronal and morphological bases of cognitive decline in aged rhesus monkeys. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1051-73. [PMID: 21710198 PMCID: PMC3448991 DOI: 10.1007/s11357-011-9278-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 06/03/2011] [Indexed: 05/13/2023]
Abstract
Rhesus monkeys provide a valuable model for studying the basis of cognitive aging because they are vulnerable to age-related decline in executive function and memory in a manner similar to humans. Some of the behavioral tasks sensitive to the effects of aging are the delayed response working memory test, recognition memory tests including the delayed nonmatching-to-sample and the delayed recognition span task, and tests of executive function including reversal learning and conceptual set-shifting task. Much effort has been directed toward discovering the neurobiological parameters that are coupled to individual differences in age-related cognitive decline. Area 46 of the dorsolateral prefrontal cortex (dlPFC) has been extensively studied for its critical role in executive function while the hippocampus and related cortical regions have been a major target of research for memory function. Some of the key age-related changes in area 46 include decreases in volume, microcolumn strength, synapse density, and α1- and α2-adrenergic receptor binding densities. All of these measures significantly correlate with cognitive scores. Interestingly, the critical synaptic subtypes associated with cognitive function appear to be different between the dlPFC and the hippocampus. For example, the dendritic spine subtype most critical to task acquisition and vulnerable to aging in area 46 is the thin spine, whereas in the dentate gyrus, the density of large mushroom spines with perforated synapses correlates with memory performance. This review summarizes age-related changes in anatomical, neuronal, and synaptic parameters within brain areas implicated in cognition and whether these changes are associated with cognitive decline.
Collapse
Affiliation(s)
- Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Mount Sinai School of Medicine, New York, NY 10029 USA
- Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1065, New York, NY 10029 USA
| | - Peter R. Rapp
- Laboratory of Experimental Gerontology, National Institute on Aging, Baltimore, MD 21224 USA
| | - John H. Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories, Mount Sinai School of Medicine, New York, NY 10029 USA
- Friedman Brain Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1065, New York, NY 10029 USA
- Department of Geriatrics and Palliative Medicine, Mount Sinai School of Medicine, New York, NY 10029 USA
- Computational Neurobiology and Imaging Center, Mount Sinai School of Medicine, New York, NY 10029 USA
- Department of Neuroscience, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1065, New York, NY 10029 USA
| |
Collapse
|
47
|
Kohama SG, Rosene DL, Sherman LS. Age-related changes in human and non-human primate white matter: from myelination disturbances to cognitive decline. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1093-110. [PMID: 22203458 PMCID: PMC3448998 DOI: 10.1007/s11357-011-9357-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2011] [Accepted: 12/01/2011] [Indexed: 05/04/2023]
Abstract
The cognitive decline associated with normal aging was long believed to be due primarily to decreased synaptic density and neuron loss. Recent studies in both humans and non-human primates have challenged this idea, pointing instead to disturbances in white matter (WM) including myelin damage. Here, we review both cross-sectional and longitudinal studies in humans and non-human primates that collectively support the hypothesis that WM disturbances increase with age starting at middle age in humans, that these disturbances contribute to age-related cognitive decline, and that age-related WM changes may occur as a result of free radical damage, degenerative changes in cells in the oligodendrocyte lineage, and changes in microenvironments within WM.
Collapse
Affiliation(s)
- Steven G. Kohama
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR USA
| | | | - Larry S. Sherman
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR USA
- Division of Neuroscience, Oregon National Primate Research Center, 505 NW 185th Ave, Beaverton, OR 97006 USA
| |
Collapse
|
48
|
Nishimura M, Nakamura SI, Kimura N, Liu L, Suzuki T, Tooyama I. Age-related modulation of γ-secretase activity in non-human primate brains. J Neurochem 2012; 123:21-8. [DOI: 10.1111/j.1471-4159.2012.07884.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
49
|
¿Existe la enfermedad de Alzheimer en todos los primates? Afección de Alzheimer en primates no humanos y sus implicaciones fisiopatológicas (I). Neurologia 2012; 27:354-69. [DOI: 10.1016/j.nrl.2011.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 05/19/2011] [Indexed: 11/17/2022] Open
|
50
|
Toledano A, Álvarez M, López-Rodríguez A, Toledano-Díaz A, Fernández-Verdecia C. Does Alzheimer's disease exist in all primates? Alzheimer pathology in non-human primates and its pathophysiological implications (I). NEUROLOGÍA (ENGLISH EDITION) 2012. [DOI: 10.1016/j.nrleng.2012.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|