1
|
Huhe H, Shapley SM, Duong DM, Wu F, Ha SK, Choi SH, Kofler J, Mou Y, Guimaraes TR, Thathiah A, Watson CM, Schaeffer LKH, Carter GW, Seyfried NT, Silva AC, Sukoff Rizzo SJ. Marmosets as model systems for the study of Alzheimer's disease and related dementias: Substantiation of physiological tau 3R and 4R isoform expression and phosphorylation. Alzheimers Dement 2024. [PMID: 39559898 DOI: 10.1002/alz.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Marmosets spontaneously develop pathological hallmarks of Alzheimer's disease (AD) including amyloid beta plaques. However, tau expression in the marmoset brain has been understudied. METHODS Isoforms of tau were examined by western blot, mass spectrometry, immunofluorescence, and immunohistochemical staining. RESULTS 3R and 4R tau isoforms are expressed in marmoset brains at both the transcript and protein levels across ages. Mass spectrometry analysis revealed that tau peptides in marmoset corresponded to the 3R and 4R peptides in human brain, with 3R predominating at birth and an ≈40%:60% 3R:4R ratios in adolescents and adults; tau was distributed widely in neurons, with localization in the soma and synaptic regions. Phosphorylation residues were observed on Threonine (Thr) Thr181, Thr217, Thr231, Serine (Ser) Ser202/Thr205, and Ser396/Ser404. DISCUSSION Our results confirm both 3R and 4R tau isoform expression and phosphorylation residues in the marmoset brain, and emphasize the significance of marmosets with natural expression of AD-related hallmarks as important translational models for AD. Highlights We report comprehensive characterization of tau isoform expression in marmoset brains across the lifespan. 3R and 4R tau isoforms are expressed in marmoset brains at both the transcript and protein levels across ages. These data emphasize the significance of marmosets with natural expression of primate-specific traits that are important for the study of Alzheimer's disease.
Collapse
Affiliation(s)
- Hasi Huhe
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah M Shapley
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fang Wu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Seung-Kwon Ha
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sang-Ho Choi
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yongshan Mou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thais Rafael Guimaraes
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amantha Thathiah
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Caroline M Watson
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lauren K H Schaeffer
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Afonso C Silva
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stacey J Sukoff Rizzo
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
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
|
Varma C, Luo E, Bostrom G, Bathini P, Berdnik D, Wyss‐Coray T, Zhao T, Dong X, Ervin FR, Beierschmitt A, Palmour RM, Lemere CA. Plasma and CSF biomarkers of aging and cognitive decline in Caribbean vervets. Alzheimers Dement 2024; 20:5460-5480. [PMID: 38946666 PMCID: PMC11350037 DOI: 10.1002/alz.14038] [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: 02/22/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 07/02/2024]
Abstract
INTRODUCTION Vervets are non-human primates that share high genetic homology with humans and develop amyloid beta (Aβ) pathology with aging. We expand current knowledge by examining Aβ pathology, aging, cognition, and biomarker proteomics. METHODS Amyloid immunoreactivity in the frontal cortex and temporal cortex/hippocampal regions from archived vervet brain samples ranging from young adulthood to old age was quantified. We also obtained cognitive scores, plasma samples, and cerebrospinal fluid (CSF) samples in additional animals. Plasma and CSF proteins were quantified with platforms utilizing human antibodies. RESULTS We found age-related increases in Aβ deposition in both brain regions. Bioinformatic analyses assessed associations between biomarkers and age, sex, cognition, and CSF Aβ levels, revealing changes in proteins related to immune-related inflammation, metabolism, and cellular processes. DISCUSSION Vervets are an effective model of aging and early-stage Alzheimer's disease, and we provide translational biomarker data that both align with previous results in humans and provide a basis for future investigations. HIGHLIGHTS We found changes in immune and metabolic plasma biomarkers associated with age and cognition. Cerebrospinal fluid (CSF) biomarkers revealed changes in cell signaling indicative of adaptative processes. TNFRSF19 (TROY) and Artemin co-localize with Alzheimer's disease pathology. Vervets are a relevant model for translational studies of early-stage Alzheimer's disease.
Collapse
Affiliation(s)
- Curran Varma
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
| | - Eva Luo
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
| | - Gustaf Bostrom
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Department of Public Health and Caring SciencesGeriatrics, Uppsala UniversityUppsalaSweden
- Centre for Clinical ResearchUppsala UniversityVästmanland County HospitalVästeråsSweden
| | - Praveen Bathini
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Daniela Berdnik
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
| | - Tingting Zhao
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Xianjun Dong
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
- Genomics and Bioinformatics HubBrigham and Women's HospitalBostonMassachusettsUSA
| | - Frank R. Ervin
- Behavioral Sciences FoundationSaint Kitts, Eastern CaribbeanMontrealCanada
- Faculty of Medicine and Health SciencesMcGill UniversityMontrealCanada
| | - Amy Beierschmitt
- Behavioral Sciences FoundationSaint Kitts, Eastern CaribbeanMontrealCanada
- Department of Biomedical SciencesRoss University School of Veterinary MedicineSt KittsUK
| | - Roberta M. Palmour
- Behavioral Sciences FoundationSaint Kitts, Eastern CaribbeanMontrealCanada
- Faculty of Medicine and Health SciencesMcGill UniversityMontrealCanada
| | - Cynthia A. Lemere
- Department of NeurologyAnn Romney Center for Neurologic DiseasesBrigham and Women's HospitalBostonMassachusettsUSA
- Department of NeurologyHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
5
|
Balamayooran G, Tooze JA, Gardin JF, Long MC, Caudell DL, Cline JM, Kock ND, Paitsel M, Moore S, Jorgensen MJ. Age and sex associated organ weight differences in vervets/African green monkeys (Chlorocebus aethiops sabaeus). J Med Primatol 2024; 53:e12721. [PMID: 39048121 PMCID: PMC11378953 DOI: 10.1111/jmp.12721] [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: 04/24/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024]
Abstract
AbstractBackgroundAfrican green monkeys (AGMs, also known as vervets, Cholorocebus aethiops sabaeus) have been used in a variety of biomedical research studies. The aim of this study was to generate a reference for normal organ weights and percentage organ weights in AGMs of different age categories and sex.MethodsThe organ weights were compiled from 479 AGMs (285 females and 194 males) from 2004 to 2021. Age and sex differences of absolute and relative organ weights were analyzed using analysis of variance.ResultsThe findings demonstrate that males had higher body and organ weights than age‐matched females, but relative organ weights did not differ between males and females. At maturity, adrenal gland, brain, kidney, liver, thymus, and thyroid gland weights as a percentage of body weight declined, but relative weights of prostate gland, testes, and uterus were higher.ConclusionThese data should be beneficial to biomedical researchers and pathologists working with AGMs.
Collapse
Affiliation(s)
- Gayathriy Balamayooran
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Janet A Tooze
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jean F Gardin
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Margaret C Long
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - David L Caudell
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nancy D Kock
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Monica Paitsel
- Animal Resources Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Stacy Moore
- Animal Resources Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
6
|
Huhe H, Shapley SM, Duong D, Wu F, Ha SK, Choi SH, Kofler J, Mou Y, Guimaraes TR, Thathiah A, Schaeffer LKH, Carter GW, Seyfried NT, Silva AC, Sukoff Rizzo SJ. Marmosets as model systems for the study of Alzheimer's disease and related dementias: substantiation of physiological Tau 3R and 4R isoform expression and phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590453. [PMID: 38746277 PMCID: PMC11092449 DOI: 10.1101/2024.04.26.590453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Marmosets have been shown to spontaneously develop pathological hallmarks of Alzheimer's disease (AD) during advanced age, including amyloid-beta plaques, positioning them as a model system to overcome the rodent-to-human translational gap for AD. However, Tau expression in the marmoset brain has been understudied. METHODS To comprehensively investigate Tau isoform expression in marmosets, brain tissue from eight unrelated marmosets across various ages was evaluated and compared to human postmortem AD tissue. Microtubule-associated protein tau ( MAPT ) mRNA expression and splicing were confirmed by RT-PCR. Tau isoforms in the marmoset brain were examined by western blot, mass spectrometry, immunofluorescence, and immunohistochemical staining. Synaptic Tau expression was analyzed from crude synaptosome extractions. RESULTS 3R and 4R Tau isoforms are expressed in marmoset brains at both transcript and protein levels across ages. Results from western blot analysis were confirmed by mass spectrometry, which revealed that Tau peptides in marmoset corresponded to the 3R and 4R peptides in the human AD brain. 3R Tau was primarily enriched in neonate brains, and 4R enhanced in adult and aged brains. Tau was widely distributed in neurons with localization in the soma and synaptic regions. Phosphorylation residues were observed on Thr-181, Thr-217, and Thr-231, Ser202/Thr205, Ser396/Ser404. Paired helical filament (PHF)-like aggregates were also detected in aged marmosets. DISCUSSION Our results confirm the expression of both 3R and 4R Tau isoforms and important phosphorylation residues in the marmoset brain. These data emphasize the significance of marmosets with natural expression of AD-related hallmarks as important translational models for the study of AD.
Collapse
|
7
|
Lee GY, Ham S, Sohn J, Kwon HC, Lee SJV. Meta-analysis of the transcriptome identifies aberrant RNA processing as common feature of aging in multiple species. Mol Cells 2024; 47:100047. [PMID: 38508494 PMCID: PMC11026732 DOI: 10.1016/j.mocell.2024.100047] [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: 02/07/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
Aging is accompanied by the gradual deregulation of the transcriptome. However, whether age-dependent changes in the transcriptome are evolutionarily conserved or diverged remains largely unexplored. Here, we performed a meta-analysis examining the age-dependent changes in the transcriptome using publicly available datasets of 11 representative metazoans, ranging from Caenorhabditis elegans to humans. To identify the transcriptomic changes associated with aging, we analyzed various aspects of the transcriptome, including genome composition, RNA processing, and functional consequences. The use of introns and novel splice sites tended to increase with age, particularly in the brain. In addition, our analysis suggests that the age-dependent accumulation of premature termination codon-containing transcripts is a common feature of aging across multiple animal species. Using C. elegans as a test model, we showed that several splicing factors that are evolutionarily conserved and age-dependently downregulated were required to maintain a normal lifespan. Thus, aberrant RNA processing appears to be associated with aging and a short lifespan in various species.
Collapse
Affiliation(s)
- Gee-Yoon Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Seokjin Ham
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Jooyeon Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Hyunwoo C Kwon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea
| | - Seung-Jae V Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| |
Collapse
|
8
|
Polis B, Samson AO. Addressing the Discrepancies Between Animal Models and Human Alzheimer's Disease Pathology: Implications for Translational Research. J Alzheimers Dis 2024; 98:1199-1218. [PMID: 38517793 DOI: 10.3233/jad-240058] [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] [Indexed: 03/24/2024]
Abstract
Animal models, particularly transgenic mice, are extensively used in Alzheimer's disease (AD) research to emulate key disease hallmarks, such as amyloid plaques and neurofibrillary tangles formation. Although these models have contributed to our understanding of AD pathogenesis and can be helpful in testing potential therapeutic interventions, their reliability is dubious. While preclinical studies have shown promise, clinical trials often yield disappointing results, highlighting a notable gap and disparity between animal models and human AD pathology. Existing models frequently overlook early-stage human pathologies and other key AD characteristics, thereby limiting their application in identifying optimal therapeutic interventions. Enhancing model reliability necessitates rigorous study design, comprehensive behavioral evaluations, and biomarker utilization. Overall, a nuanced understanding of each model's neuropathology, its fidelity to human AD, and its limitations is essential for accurate interpretation and successful translation of findings. This article analyzes the discrepancies between animal models and human AD pathology that complicate the translation of findings from preclinical studies to clinical applications. We also delve into AD pathogenesis and attributes to propose a new perspective on this pathology and deliberate over the primary limitations of key experimental models. Additionally, we discuss several fundamental problems that may explain the translational failures and suggest some possible directions for more effective preclinical studies.
Collapse
Affiliation(s)
- Baruh Polis
- Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel
| | | |
Collapse
|
9
|
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
|
10
|
Rothwell ES, Carp SB, Bliss-Moreau E. The importance of social behavior in nonhuman primate studies of aging: A mini-review. Neurosci Biobehav Rev 2023; 154:105422. [PMID: 37806369 PMCID: PMC10716830 DOI: 10.1016/j.neubiorev.2023.105422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 08/30/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
Social behavior plays an important role in supporting both psychological and physical health across the lifespan. People's social lives change as they age, and the nature of these changes differ based on whether people are on healthy aging trajectories or are experiencing neurodegenerative diseases that cause dementia, such as Alzheimer's disease and Parkinson's disease. Nonhuman primate models of aging have provided a base of knowledge comparing aging trajectories in health and disease, but these studies rarely emphasize social behavior changes as a consequence of the aging process. What data exist hold particular value, as negative effects of disease and aging on social behavior are likely to have disproportionate impacts on quality of life. In this mini review, we examine the literature on nonhuman primate models of aging with a focus on social behavior, in the context of both health and disease. We propose that adopting a greater focus on social behavior outcomes in nonhuman primates will improve our understanding of the intersection of health, aging and sociality in humans.
Collapse
Affiliation(s)
- Emily S Rothwell
- Department of Neurobiology, School of Medicine University of Pittsburgh, 3501 Fifth Avenue, Biomedical Science Tower 3, Pittsburgh, PA 15213, USA.
| | - Sarah B Carp
- Neuroscience & Behavior Unit, California National Primate Research Center, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA
| | - Eliza Bliss-Moreau
- Neuroscience & Behavior Unit, California National Primate Research Center, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA; Department of Psychology, University of California Davis, County Road 98 at Hutchinson Drive, Davis, CA 95616, USA
| |
Collapse
|
11
|
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
|
12
|
de Sousa AA, Rigby Dames BA, Graff EC, Mohamedelhassan R, Vassilopoulos T, Charvet CJ. Going beyond established model systems of Alzheimer's disease: companion animals provide novel insights into the neurobiology of aging. Commun Biol 2023; 6:655. [PMID: 37344566 PMCID: PMC10284893 DOI: 10.1038/s42003-023-05034-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/12/2023] [Indexed: 06/23/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by brain plaques, tangles, and cognitive impairment. AD is one of the most common age-related dementias in humans. Progress in characterizing AD and other age-related disorders is hindered by a perceived dearth of animal models that naturally reproduce diseases observed in humans. Mice and nonhuman primates are model systems used to understand human diseases. Still, these model systems lack many of the biological characteristics of Alzheimer-like diseases (e.g., plaques, tangles) as they grow older. In contrast, companion animal models (cats and dogs) age in ways that resemble humans. Both companion animal models and humans show evidence of brain atrophy, plaques, and tangles, as well as cognitive decline with age. We embrace a One Health perspective, which recognizes that the health of humans is connected to those of animals, and we illustrate how such a perspective can work synergistically to enhance human and animal health. A comparative biology perspective is ideally suited to integrate insights across veterinary and human medical disciplines and solve long-standing problems in aging.
Collapse
Affiliation(s)
- Alexandra A de Sousa
- Centre for Health and Cognition, Bath Spa University, Bath, UK
- Department of Psychology, University of Bath, Bath, UK
| | - Brier A Rigby Dames
- Department of Psychology, University of Bath, Bath, UK
- Department of Computer Science, University of Bath, Bath, UK
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Emily C Graff
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Rania Mohamedelhassan
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Tatianna Vassilopoulos
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA.
| |
Collapse
|
13
|
Hopkins WD, Li X, Roberts N, Mulholland MM, Sherwood CC, Edler MK, Raghanti MA, Schapiro SJ. Age differences in cortical thickness and their association with cognition in chimpanzee (Pan troglodytes). Neurobiol Aging 2023; 126:91-102. [PMID: 36958104 PMCID: PMC10106435 DOI: 10.1016/j.neurobiolaging.2023.02.008] [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: 10/20/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Humans and chimpanzees are genetically similar and share a number of life history, behavioral, cognitive and neuroanatomical similarities. Notwithstanding, our understanding of age-related changes in cognitive and motor functions in chimpanzees remains largely unstudied despite recent evident demonstrating that chimpanzees exhibit many of the same neuropathological features of Alzheimer's disease observed in human postmortem brains. Here, we examined age-related differences in cognition and cortical thickness measured from magnetic resonance images in a sample of 215 chimpanzees ranging in age between 9 and 54 years. We found that chimpanzees showed global and region-specific thinning of cortex with increasing age. Further, within the elderly cohort, chimpanzees that performed better than average had thicker cortex in frontal, temporal and parietal regions compared to chimpanzees that performed worse than average. Independent of age, we also found sex differences in cortical thickness in 4 brain regions. Males had higher adjusted cortical thickness scores for the caudal anterior cingulate, rostral anterior cingulate, and medial orbital frontal while females had higher values for the inferior parietal cortex. We found no evidence that increasing age nor sex was associated with asymmetries in cortical thickness. Moreover, age-related differences in cognitive function were only weakly associated with asymmetries in cortical thickness. In summary, as has been reported in humans and other primates, elderly chimpanzees show thinner cortex and variation in cortical thickness is associated with general cognitive functions.
Collapse
Affiliation(s)
- William D Hopkins
- National Center for Chimpanzee Care, Department of Comparative Medicine, The University of Texas MD Anderson Cancer Center, Bastrop, TX.
| | - Xiang Li
- School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Neil Roberts
- School of Clinical Sciences, University of Edinburgh, Edinburgh, UK
| | - Michele M Mulholland
- National Center for Chimpanzee Care, Department of Comparative Medicine, The University of Texas MD Anderson Cancer Center, Bastrop, TX
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC
| | - Melissa K Edler
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, OH
| | - Mary Ann Raghanti
- Department of Anthropology, School of Biomedical Sciences, and Brain Health Research Institute, Kent State University, Kent, OH
| | - Steven J Schapiro
- National Center for Chimpanzee Care, Department of Comparative Medicine, The University of Texas MD Anderson Cancer Center, Bastrop, TX; Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Wang S, Ning H, Wang X, Chen L, Hua L, Ren F, Hu D, Li R, Ma Z, Ge Y, Yin Z. Exposure to bisphenol A induces neurotoxicity associated with synaptic and cytoskeletal dysfunction in neuro-2a cells. Toxicol Ind Health 2023; 39:325-335. [PMID: 37122122 DOI: 10.1177/07482337231172827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Bisphenol A (BPA) has been reported to injure the developing and adult brain. However, the underlying mechanism still remains elusive. This study used neuro-2a cells as a cellular model to investigate the neurotoxic effects of BPA. Microtubule-associated protein 2 (MAP2) and tau protein maintain microtubule normal function and promote the normal development of the nervous system. Synaptophysin (SYP) and drebrin (Dbn) proteins are involved in regulating synaptic plasticity. Cells were exposed to the minimum essential medium (MEM), 0.01% (v/v) DMSO, and 150 μM BPA for 12, 24, or 36 h. Morphological analysis revealed that the cells in the BPA-treated groups shrank and collapsed compared with those in the control groups. CCK-8 and lactate dehydrogenase assay (LDH) assays showed that the mortality of neuro-2a cells increased as the BPA treatment time was prolonged. Ultrastructural analysis further revealed that cells demonstrated nucleolar swelling, dissolution of nuclear and mitochondrial membranes, and partial mitochondrial condensation following exposure to BPA. BPA also decreased the relative protein expression levels of MAP2, tau, and Dbn. Interestingly, the relative protein expression levels of SYP increased. These results indicated that BPA inhibited the proliferation and disrupted cytoskeleton and synaptic integrity of neuro-2a cells.
Collapse
Affiliation(s)
- Siting Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Hongmei Ning
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Xinrui Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Lingli Chen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Liushuai Hua
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Fei Ren
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Dongfang Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Rongbo Li
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Zhisheng Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Yaming Ge
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| | - Zhihong Yin
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, P. R. China
| |
Collapse
|
15
|
Rigby Dames BA, Kilili H, Charvet CJ, Díaz-Barba K, Proulx MJ, de Sousa AA, Urrutia AO. Evolutionary and genomic perspectives of brain aging and neurodegenerative diseases. PROGRESS IN BRAIN RESEARCH 2023; 275:165-215. [PMID: 36841568 PMCID: PMC11191546 DOI: 10.1016/bs.pbr.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This chapter utilizes genomic concepts and evolutionary perspectives to further understand the possible links between typical brain aging and neurodegenerative diseases, focusing on the two most prevalent of these: Alzheimer's disease and Parkinson's disease. Aging is the major risk factor for these neurodegenerative diseases. Researching the evolutionary and molecular underpinnings of aging helps to reveal elements of the typical aging process that leave individuals more vulnerable to neurodegenerative pathologies. Very little is known about the prevalence and susceptibility of neurodegenerative diseases in nonhuman species, as only a few individuals have been observed with these neuropathologies. However, several studies have investigated the evolution of lifespan, which is closely connected with brain size in mammals, and insights can be drawn from these to enrich our understanding of neurodegeneration. This chapter explores the relationship between the typical aging process and the events in neurodegeneration. First, we examined how age-related processes can increase susceptibility to neurodegenerative diseases. Second, we assessed to what extent neurodegeneration is an accelerated form of aging. We found that while at the phenotypic level both neurodegenerative diseases and the typical aging process share some characteristics, at the molecular level they show some distinctions in their profiles, such as variation in genes and gene expression. Furthermore, neurodegeneration of the brain is associated with an earlier onset of cellular, molecular, and structural age-related changes. In conclusion, a more integrative view of the aging process, both from a molecular and an evolutionary perspective, may increase our understanding of neurodegenerative diseases.
Collapse
Affiliation(s)
- Brier A Rigby Dames
- Department of Computer Science, University of Bath, Bath, United Kingdom; Department of Psychology, University of Bath, Bath, United Kingdom.
| | - Huseyin Kilili
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Karina Díaz-Barba
- Licenciatura en Ciencias Genómicas, UNAM, CP62210, Cuernavaca, México; Instituto de Ecología, UNAM, Ciudad Universitaria, CP04510, Ciudad de México, México
| | - Michael J Proulx
- Department of Psychology, University of Bath, Bath, United Kingdom
| | | | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom; Licenciatura en Ciencias Genómicas, UNAM, CP62210, Cuernavaca, México; Instituto de Ecología, UNAM, Ciudad Universitaria, CP04510, Ciudad de México, México.
| |
Collapse
|
16
|
Gupta A, Vardalakis N, Wagner FB. Neuroprosthetics: from sensorimotor to cognitive disorders. Commun Biol 2023; 6:14. [PMID: 36609559 PMCID: PMC9823108 DOI: 10.1038/s42003-022-04390-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Neuroprosthetics is a multidisciplinary field at the interface between neurosciences and biomedical engineering, which aims at replacing or modulating parts of the nervous system that get disrupted in neurological disorders or after injury. Although neuroprostheses have steadily evolved over the past 60 years in the field of sensory and motor disorders, their application to higher-order cognitive functions is still at a relatively preliminary stage. Nevertheless, a recent series of proof-of-concept studies suggest that electrical neuromodulation strategies might also be useful in alleviating some cognitive and memory deficits, in particular in the context of dementia. Here, we review the evolution of neuroprosthetics from sensorimotor to cognitive disorders, highlighting important common principles such as the need for neuroprosthetic systems that enable multisite bidirectional interactions with the nervous system.
Collapse
Affiliation(s)
- Ankur Gupta
- grid.462010.1Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | | | - Fabien B. Wagner
- grid.462010.1Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
17
|
Park J, Won J, Jeon CY, Lim KS, Choi WS, Park SH, Seo J, Cho J, Seong JB, Yeo HG, Kim K, Kim YG, Kim M, Yi KS, Lee Y. XperCT-guided Intra-cisterna Magna Injection of Streptozotocin for Establishing an Alzheimer's Disease Model Using the Cynomolgus Monkey ( Macaca fascicularis). Exp Neurobiol 2022; 31:409-418. [PMID: 36631849 PMCID: PMC9841743 DOI: 10.5607/en22027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/13/2023] Open
Abstract
Till date, researchers have been developing animal models of Alzheimer's disease (AD) in various species to understand the pathological characterization and molecular mechanistic pathways associated with this condition in humans to identify potential therapeutic treatments. A widely recognized AD model that mimics the pathology of human AD involves the intracerebroventricular (ICV) injection with streptozotocin (STZ). However, ICV injection as an invasive approach has several limitations related to complicated surgical procedures. Therefore, in the present study, we created a customized stereotaxic frame using the XperCT-guided system for injecting STZ in cynomolgus monkeys, aiming to establish an AD model. The anatomical structures surrounding the cisterna magna (CM) were confirmed using CT/MRI fusion images of monkey brain with XperCT, the c-arm cone beam computed tomography. XperCT was used to determine the appropriate direction in which the needle tip should be inserted within the CM region. Cerebrospinal fluid (CSF) was collected to confirm the accurate target site when STZ was injected into the CM. Cynomolgus monkeys were administered STZ dissolved in artificial CSF once every week for 4 weeks via intracisterna magna (ICM) injection using XperCT-guided stereotactic system. The molecular mechanisms underlying the progression of STZ-induced AD pathology were analyzed two weeks after the final injection. The monkeys subjected to XperCT-based STZ injection via the ICM route showed features of AD pathology, including markedly enhanced neuronal loss, synaptic impairment, and tau phosphorylation in the hippocampus. These findings suggest a new approach for the construction of neurodegenerative disease models and development of therapeutic strategies.
Collapse
Affiliation(s)
- Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Sung-hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jiyeon Cho
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jung Bae Seong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Hyeon-Gu Yeo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Yu Gyeong Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Minji Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Kyung Sik Yi
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea,To whom correspondence should be addressed. TEL: 82-43-240-6316, FAX: 82-43-240-6309, e-mail:
| |
Collapse
|
18
|
Wakeman DR, Weed MR, Perez SE, Cline EN, Viola KL, Wilcox KC, Moddrelle DS, Nisbett EZ, Kurian AM, Bell AF, Pike R, Jacobson PB, Klein WL, Mufson EJ, Lawrence MS, Elsworth JD. Intrathecal amyloid-beta oligomer administration increases tau phosphorylation in the medial temporal lobe in the African green monkey: A nonhuman primate model of Alzheimer's disease. Neuropathol Appl Neurobiol 2022; 48:e12800. [PMID: 35156715 PMCID: PMC10902791 DOI: 10.1111/nan.12800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 11/26/2022]
Abstract
AIMS An obstacle to developing new treatment strategies for Alzheimer's disease (AD) has been the inadequate translation of findings in current AD transgenic rodent models to the prediction of clinical outcomes. By contrast, nonhuman primates (NHPs) share a close neurobiology with humans in virtually all aspects relevant to developing a translational AD model. The present investigation used African green monkeys (AGMs) to refine an inducible NHP model of AD based on the administration of amyloid-beta oligomers (AβOs), a key upstream initiator of AD pathology. METHODS AβOs or vehicle were repeatedly delivered over 4 weeks to age-matched young adult AGMs by intracerebroventricular (ICV) or intrathecal (IT) injections. Induction of AD-like pathology was assessed in subregions of the medial temporal lobe (MTL) by quantitative immunohistochemistry (IHC) using the AT8 antibody to detect hyperphosphorylated tau. Hippocampal volume was measured by magnetic resonance imaging (MRI) scans prior to, and after, intrathecal injections. RESULTS IT administration of AβOs in young adult AGMs revealed an elevation of tau phosphorylation in the MTL cortical memory circuit compared with controls. The largest increases were detected in the entorhinal cortex that persisted for at least 12 weeks after dosing. MRI scans showed a reduction in hippocampal volume following AβO injections. CONCLUSIONS Repeated IT delivery of AβOs in young adult AGMs led to an accelerated AD-like neuropathology in MTL, similar to human AD, supporting the value of this translational model to de-risk the clinical trial of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Sylvia E Perez
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Erika N Cline
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kirsten L Viola
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kyle C Wilcox
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - David S Moddrelle
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ernell Z Nisbett
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - Amanda F Bell
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ricaldo Pike
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - William L Klein
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Elliott J Mufson
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
19
|
Robertson EL, Boehnke SE, Lyra e Silva NDM, Armitage‐Brown B, Winterborn A, Cook DJ, De Felice FG, Munoz DP. Characterization of cerebrospinal fluid biomarkers associated with neurodegenerative diseases in healthy cynomolgus and rhesus macaque monkeys. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12289. [PMID: 35415210 PMCID: PMC8984079 DOI: 10.1002/trc2.12289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
Monkeys are becoming important translational models of neurodegenerative disease. To facilitate model development, we measured cerebrospinal fluid (CSF) concentrations of key biomarkers in healthy male and female cynomolgus and rhesus macaques. Amyloid beta (Aβ40, Aβ42), tau (total tau [t-tau], phosphorylated tau [pThr181]), and neurofilament light (NfL) concentrations were measured in CSF of 82 laboratory-housed, experimentally naïve cynomolgus (n = 33) and rhesus (n = 49) macaques. Aβ40 and Aβ42 were significantly higher in rhesus, and female rhesus were higher than males. NfL and t-tau were higher in males, and NfL was higher in rhesus macaques. p-tau was not affected by species or sex. We also examined whether sample location (lumbar or cisterna puncture) affected concentrations. Sample acquisition site only affected NfL, which was higher in CSF from lumbar puncture compared to cisterna magna puncture. Establishing normative biomarker values for laboratory-housed macaque monkeys provides an important resource by which to compare to monkey models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Emma L. Robertson
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
| | - Susan E. Boehnke
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Natalia de M. Lyra e Silva
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| | - Brittney Armitage‐Brown
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Animal Care ServicesQueen's UniversityKingstonOntarioCanada
| | | | - Douglas J. Cook
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of SurgeryKingston Health Sciences CentreKingstonOntarioCanada
| | - Fernanda G. De Felice
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de Janeiro, Cidade Universitaria – Rio de JaneiroRio de JaneiroBrazil
- D'OR Institute for Research and EducationRio de JaneiroBrazil
- Department of PsychiatryProvidence Care HospitalKingstonOntarioCanada
| | - Douglas P. Munoz
- Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of Biomedical and Molecular SciencesQueen's UniversityKingstonOntarioCanada
| |
Collapse
|
20
|
Jester HM, Gosrani SP, Ding H, Zhou X, Ko MC, Ma T. Characterization of Early Alzheimer's Disease-Like Pathological Alterations in Non-Human Primates with Aging: A Pilot Study. J Alzheimers Dis 2022; 88:957-970. [PMID: 35723096 PMCID: PMC9378582 DOI: 10.3233/jad-215303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Sporadic or late onset Alzheimer's disease (LOAD) is a multifactorial neurodegenerative disease with aging the most known risk factor. Non-human primates (NHPs) may serve as an excellent model to study LOAD because of their close similarity to humans in many aspects including neuroanatomy and neurodevelopment. Recent studies reveal AD-like pathology in old NHPs. OBJECTIVE In this pilot study, we took advantage of brain samples from 6 Cynomolgus macaques that were divided into two groups: middle aged (average age 14.81 years) and older (average age 19.33 years). We investigated whether AD-like brain pathologies are present in the NHPs. METHODS We used immunohistochemical method to examine brain Aβ pathology and neuron density. We applied biochemical assays to measure tau phosphorylation and multiple signaling pathways indicated in AD. We performed electron microscopy experiments to study alterations of postsynaptic density and mitochondrial morphology in the brain of NHPs. RESULTS We found multiple AD-like pathological alteration in the prefrontal cortex (but not in the hippocampus) of the older NHPs including tau hyperphosphorylation, increased activity of AMP-activated protein kinase (AMPK), decreased expression of protein phosphatase 2A (PP2A), impairments in mitochondrial morphology, and postsynaptic densities formation. CONCLUSION These findings may provide insights into the factors contributing to the development of LOAD, particularly during the early stage transitioning from middle to old age. Future endeavors are warranted to elucidate mechanisms underlying the regional (and perhaps cellular) vulnerability with aging and the functional correlation of such pathological changes in NHPs.
Collapse
Affiliation(s)
- Hannah M. Jester
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Saahj P. Gosrani
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Xueyan Zhou
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Tao Ma
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
21
|
Gilberto DB, Michener MS, Smith BE, Szczerba PJ, Holahan MA, Gray TL, Motzel SL. Chronic Collection of Cerebrospinal Fluid from Rhesus Macaques (Macaca mulatta) with Cisterna Magna Ports: Update on Refinements. Comp Med 2021; 72:45-49. [PMID: 34903315 DOI: 10.30802/aalas-cm-21-000060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
More than 20 y ago, we developed an animal model for chronic and continuous collection of cerebrospinal fluid (CSF) from conscious rhesus macaques. Since our previous publication in 2003, we have successfully implanted 168 rhesus macaquesusing this approach. Our experience enables us to provide up-to-date information regarding the model, including refinementsto our implant design, reductions in maintenance, and new procedures for dealing with contamination. The results of our experiences have reduced the number of surgeries required and helped to increase the longevity of the implant, with some functioning for more than 18 y. Building on our success in rhesus macaques, we attempted to develop similar animal models in the African green monkeys and dogs but have been unable to develop reliable chronic models for CSF collection in these species.
Collapse
|
22
|
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: 9] [Impact Index Per Article: 2.3] [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
|
23
|
Frye BM, Craft S, Latimer CS, Keene CD, Montine TJ, Register TC, Orr ME, Kavanagh K, Macauley SL, Shively CA. Aging-related Alzheimer's disease-like neuropathology and functional decline in captive vervet monkeys (Chlorocebus aethiops sabaeus). Am J Primatol 2021; 83:e23260. [PMID: 33818801 PMCID: PMC8626867 DOI: 10.1002/ajp.23260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Age-related neurodegeneration characteristic of late-onset Alzheimer's disease (LOAD) begins in middle age, well before symptoms. Translational models to identify modifiable risk factors are needed to understand etiology and identify therapeutic targets. Here, we outline the evidence supporting the vervet monkey (Chlorocebus aethiops sabaeus) as a model of aging-related AD-like neuropathology and associated phenotypes including cognitive function, physical function, glucose handling, intestinal physiology, and CSF, blood, and neuroimaging biomarkers. This review provides the most comprehensive multisystem description of aging in vervets to date. This review synthesizes a large body of evidence that suggests that aging vervets exhibit a coordinated suite of traits consistent with early AD and provide a powerful, naturally occurring model for LOAD. Notably, relationships are identified between AD-like neuropathology and modifiable risk factors. Gaps in knowledge and key limitations are provided to shape future studies to illuminate mechanisms underlying divergent neurocognitive aging trajectories and to develop interventions that increase resilience to aging-associated chronic disease, particularly, LOAD.
Collapse
Affiliation(s)
- Brett M. Frye
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | | | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Miranda E. Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Kylie Kavanagh
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
| | - Shannon L. Macauley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| | - Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| |
Collapse
|
24
|
Shively CA, Lacreuse A, Frye BM, Rothwell ES, Moro M. Nonhuman primates at the intersection of aging biology, chronic disease, and health: An introduction to the American Journal of Primatology Special Issue on aging, cognitive decline, and neuropathology in nonhuman primates. Am J Primatol 2021; 83:e23309. [PMID: 34403529 PMCID: PMC8935964 DOI: 10.1002/ajp.23309] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/05/2021] [Indexed: 11/06/2022]
Abstract
Aging across the Primate Order is poorly understood because ages of individuals are often unknown, there is a dearth of aged animals available for study, and because aging is best characterized by longitudinal studies which are difficult to carry out in long-lived species. The human population is aging rapidly, and advanced age is a primary risk factor for several chronic diseases and conditions that impact healthspan. As lifespan has increased, diseases and disorders of the central nervous system (CNS) have become more prevalent, and Alzheimer's disease and related dementias have become epidemic. Nonhuman primate (NHP) models are key to understanding the aging primate CNS. This Special Issue presents a review of current knowledge about NHP CNS aging across the Primate Order. Similarities and differences to human aging, and their implications for the validity of NHP models of aging are considered. Topics include aging-related brain structure and function, neuropathologies, cognitive performance, social behavior and social network characteristics, and physical, sensory, and motor function. Challenges to primate CNS aging research are discussed. Together, this collection of articles demonstrates the value of studying aging in a breadth of NHP models to advance our understanding of human and nonhuman primate aging and healthspan.
Collapse
Affiliation(s)
- Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
- Alzheimer’s Disease Research Center, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Agnès Lacreuse
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Brett M. Frye
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Emily S. Rothwell
- Department of Psychological and Brain Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Manuel Moro
- Division of Aging Biology, National Institute on Aging, National Institutes of Health, Maryland, USA
| |
Collapse
|
25
|
Scott JT, Bourne JA. Modelling behaviors relevant to brain disorders in the nonhuman primate: Are we there yet? Prog Neurobiol 2021; 208:102183. [PMID: 34728308 DOI: 10.1016/j.pneurobio.2021.102183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 12/30/2022]
Abstract
Recent years have seen a profound resurgence of activity with nonhuman primates (NHPs) to model human brain disorders. From marmosets to macaques, the study of NHP species offers a unique window into the function of primate-specific neural circuits that are impossible to examine in other models. Examining how these circuits manifest into the complex behaviors of primates, such as advanced cognitive and social functions, has provided enormous insights to date into the mechanisms underlying symptoms of numerous neurological and neuropsychiatric illnesses. With the recent optimization of modern techniques to manipulate and measure neural activity in vivo, such as optogenetics and calcium imaging, NHP research is more well-equipped than ever to probe the neural mechanisms underlying pathological behavior. However, methods for behavioral experimentation and analysis in NHPs have noticeably failed to keep pace with these advances. As behavior ultimately lies at the junction between preclinical findings and its translation to clinical outcomes for brain disorders, approaches to improve the integrity, reproducibility, and translatability of behavioral experiments in NHPs requires critical evaluation. In this review, we provide a unifying account of existing brain disorder models using NHPs, and provide insights into the present and emerging contributions of behavioral studies to the field.
Collapse
Affiliation(s)
- Jack T Scott
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
26
|
Yue F, Feng S, Lu C, Zhang T, Tao G, Liu J, Yue C, Jing N. Synthetic amyloid-β oligomers drive early pathological progression of Alzheimer's disease in nonhuman primates. iScience 2021; 24:103207. [PMID: 34704001 PMCID: PMC8524197 DOI: 10.1016/j.isci.2021.103207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/04/2021] [Accepted: 09/28/2021] [Indexed: 11/21/2022] Open
Abstract
As an insidious and slowly progressive neurodegenerative disorder, Alzheimer’s disease (AD) uniquely develops in humans but fails in other species. Therefore, it has been challenged to rebuild human AD in animals, including in non-human primates. Here, we bilaterally delivered synthetic Aβ oligomers (AβOs) into the cerebral parenchyma of cynomolgus monkeys, which rapidly drove the formation of massive Aβ plaques and concomitant neurofibrillary tangles in the cynomolgus brain. The amyloid and tau pathology as well as their co-occurrence in AβO-monkeys were reminiscent of those in patients with AD. In addition, the activated astrocytes and microglia surrounding Aβ plaques indicated the triggered neuroinflammation. The degenerative neurons and synapses around Aβ plaques also emerged in cynomolgus brain. Together, soluble AβOs caused the cascade of pathologic events associated with AD in monkeys as occurred in patients at the early phase, which could facilitate the development of a promising animal model for human AD in non-human primates. The Aβ oligomers (AβOs) drive to develop massive Aβ plaque in the monkey brain Neurofibrillary tangles form in multiple brain regions of AβO-monkeys The co-occurrence of amyloid and tau pathology in AβO-monkeys as in patients with AD The neuroinflammation and neurodegeneration are triggered in AβO-monkeys
Collapse
Affiliation(s)
- Feng Yue
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.,Department of Neurobiology, Beijing Institute of Geriatrics, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Su Feng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Chunling Lu
- Wincon TheraCells Biotechnologies Co, LTD, Nanning, 530000, China
| | - Ting Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.,National Clinical Research Center for Ophthalmic Diseases, Shanghai, 200080, China
| | - Guoxian Tao
- Wincon TheraCells Biotechnologies Co, LTD, Nanning, 530000, China
| | - Jing Liu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Chunmei Yue
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China.,Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, 215000, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.,CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
27
|
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: 64] [Impact Index Per Article: 16.0] [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
|
28
|
Vitek MP, Araujo JA, Fossel M, Greenberg BD, Howell GR, Rizzo SJS, Seyfried NT, Tenner AJ, Territo PR, Windisch M, Bain LJ, Ross A, Carrillo MC, Lamb BT, Edelmayer RM. Translational animal models for Alzheimer's disease: An Alzheimer's Association Business Consortium Think Tank. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 6:e12114. [PMID: 33457489 PMCID: PMC7798310 DOI: 10.1002/trc2.12114] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022]
Abstract
Over 5 million Americans and 50 million individuals worldwide are living with Alzheimer's disease (AD). The progressive dementia associated with AD currently has no cure. Although clinical trials in patients are ultimately required to find safe and effective drugs, animal models of AD permit the integration of brain pathologies with learning and memory deficits that are the first step in developing these new drugs. The purpose of the Alzheimer's Association Business Consortium Think Tank meeting was to address the unmet need to improve the discovery and successful development of Alzheimer's therapies. We hypothesize that positive responses to new therapies observed in validated models of AD will provide predictive evidence for positive responses to these same therapies in AD patients. To achieve this goal, we convened a meeting of experts to explore the current state of AD animal models, identify knowledge gaps, and recommend actions for development of next-generation models with better predictability. Among our findings, we all recognize that models reflecting only single aspects of AD pathogenesis do not mimic AD. Models or combinations of new models are needed that incorporate genetics with environmental interactions, timing of disease development, heterogeneous mechanisms and pathways, comorbidities, and other pathologies that lead to AD and related dementias. Selection of the best models requires us to address the following: (1) which animal species, strains, and genetic backgrounds are most appropriate; (2) which models permit efficient use throughout the drug development pipeline; (3) the translatability of behavioral-cognitive assays from animals to patients; and (4) how to match potential AD therapeutics with particular models. Best practice guidelines to improve reproducibility also need to be developed for consistent use of these models in different research settings. To enhance translational predictability, we discuss a multi-model evaluation strategy to de-risk the successful transition of pre-clinical drug assets to the clinic.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nicholas T. Seyfried
- Departments of Biochemistry and NeurologyEmory School of MedicineAtlantaGeorgiaUSA
| | - Andrea J. Tenner
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCaliforniaUSA
| | | | | | - Lisa J. Bain
- Independent Science and Medical WriterElversonPennsylvaniaUSA
| | - April Ross
- Former Alzheimer's Association EmployeeChicagoIllinoisUSA
| | | | - Bruce T. Lamb
- Indiana University School of MedicineStark Neurosciences Research InstituteIndianapolisIndianaUSA
| | | |
Collapse
|
29
|
Plant M, Armstrong C, Ruggiero A, Sherrill C, Uberseder B, Jeffries R, Nevarez J, Jorgensen MJ, Kavanagh K, Quinn MA. Advanced maternal age impacts physiologic adaptations to pregnancy in vervet monkeys. GeroScience 2020; 42:1649-1661. [PMID: 32588342 PMCID: PMC7732933 DOI: 10.1007/s11357-020-00219-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
The trend to delay pregnancy in the USA has resulted in the number of advanced maternal age (AMA) pregnancies to also increase. In humans, AMA is associated with a variety of pregnancy-related pathologies such as preeclampsia (PE). While AMA is known to be a factor which contributes to the development of pregnancy-induced diseases, the molecular and cellular mechanisms giving rise to this phenomenon are still very limited. This is due in part to lack of a preclinical model which has physiologic relevance to human pregnancy while also allowing control of environmental and genetic variability inherent in human studies. To determine potential physiologic relevance of the vervet/African green monkey (Chlorocebus aethiops sabaeus) as a preclinical model to study the effects of AMA on adaptations to pregnancy, thirteen age-diverse pregnant vervet monkeys (3-16 years old) were utilized to measure third trimester blood pressure (BP), complete blood count, iron measurements, and hormone levels. Significant associations were observed between third trimester diastolic BP and maternal age. Furthermore, the presence of leukocytosis with enhanced circulating neutrophils was observed in AMA mothers compared to younger mothers. Moreover, we observed a negative relationship between maternal age and estradiol, progesterone, and cortisol levels. Finally, offspring born to AMA mothers displayed a postnatal growth retardation phenotype. These studies demonstrate physiologic impairment in the adaptation to pregnancy in AMA vervet/African green monkeys. Our data indicate that the vervet/African green monkey may serve as a useful preclinical model and tool for deciphering pathological mediators of maternal disease in AMA pregnancy.
Collapse
Affiliation(s)
- Maren Plant
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Cecilia Armstrong
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Alistaire Ruggiero
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Chrissy Sherrill
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Beth Uberseder
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Rachel Jeffries
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Justin Nevarez
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Matthew A Quinn
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
30
|
Edler MK, Munger EL, Meindl RS, Hopkins WD, Ely JJ, Erwin JM, Mufson EJ, Hof PR, Sherwood CC, Raghanti MA. Neuron loss associated with age but not Alzheimer's disease pathology in the chimpanzee brain. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190619. [PMID: 32951541 PMCID: PMC7540958 DOI: 10.1098/rstb.2019.0619] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2020] [Indexed: 12/25/2022] Open
Abstract
In the absence of disease, ageing in the human brain is accompanied by mild cognitive dysfunction, gradual volumetric atrophy, a lack of significant cell loss, moderate neuroinflammation, and an increase in the amyloid beta (Aβ) and tau proteins. Conversely, pathologic age-related conditions, particularly Alzheimer's disease (AD), result in extensive neocortical and hippocampal atrophy, neuron death, substantial Aβ plaque and tau-associated neurofibrillary tangle pathologies, glial activation and severe cognitive decline. Humans are considered uniquely susceptible to neurodegenerative disorders, although recent studies have revealed Aβ and tau pathology in non-human primate brains. Here, we investigate the effect of age and AD-like pathology on cell density in a large sample of postmortem chimpanzee brains (n = 28, ages 12-62 years). Using a stereologic, unbiased design, we quantified neuron density, glia density and glia:neuron ratio in the dorsolateral prefrontal cortex, middle temporal gyrus, and CA1 and CA3 hippocampal subfields. Ageing was associated with decreased CA1 and CA3 neuron densities, while AD pathologies were not correlated with changes in neuron or glia densities. Differing from cerebral ageing and AD in humans, these data indicate that chimpanzees exhibit regional neuron loss with ageing but appear protected from the severe cell death found in AD. This article is part of the theme issue 'Evolution of the primate ageing process'.
Collapse
Affiliation(s)
- Melissa K. Edler
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
- Department of Anthropology, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Emily L. Munger
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
- Department of Anthropology, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| | - Richard S. Meindl
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
- Department of Anthropology, Kent State University, Kent, OH 44242, USA
| | - William D. Hopkins
- Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Joseph M. Erwin
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Elliott J. Mufson
- Departments of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- New York Consortium for Evolutionary Primatology, New York, NY 10468, USA
| | - Chet C. Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, DC 20052, USA
| | - Mary Ann Raghanti
- School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
- Department of Anthropology, Kent State University, Kent, OH 44242, USA
- Brain Health Research Institute, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
31
|
Takemura H, Palomero-Gallagher N, Axer M, Gräßel D, Jorgensen MJ, Woods R, Zilles K. Anatomy of nerve fiber bundles at micrometer-resolution in the vervet monkey visual system. eLife 2020; 9:e55444. [PMID: 32844747 PMCID: PMC7532002 DOI: 10.7554/elife.55444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022] Open
Abstract
Although the primate visual system has been extensively studied, detailed spatial organization of white matter fiber tracts carrying visual information between areas has not been fully established. This is mainly due to the large gap between tracer studies and diffusion-weighted MRI studies, which focus on specific axonal connections and macroscale organization of fiber tracts, respectively. Here we used 3D polarization light imaging (3D-PLI), which enables direct visualization of fiber tracts at micrometer resolution, to identify and visualize fiber tracts of the visual system, such as stratum sagittale, inferior longitudinal fascicle, vertical occipital fascicle, tapetum and dorsal occipital bundle in vervet monkey brains. Moreover, 3D-PLI data provide detailed information on cortical projections of these tracts, distinction between neighboring tracts, and novel short-range pathways. This work provides essential information for interpretation of functional and diffusion-weighted MRI data, as well as revision of wiring diagrams based upon observations in the vervet visual system.
Collapse
Affiliation(s)
- Hiromasa Takemura
- Center for Information and Neural Networks (CiNet), National Institute of Information and Communications Technology, and Osaka UniversityOsakaJapan
- Graduate School of Frontier Biosciences, Osaka UniversityOsakaJapan
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine INM-1, Research Centre JülichJülichGermany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH AachenAachenGermany
- C. & O. Vogt Institute for Brain Research, Heinrich-Heine-UniversityDüsseldorfGermany
| | - Markus Axer
- Institute of Neuroscience and Medicine INM-1, Research Centre JülichJülichGermany
| | - David Gräßel
- Institute of Neuroscience and Medicine INM-1, Research Centre JülichJülichGermany
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Roger Woods
- Ahmanson-Lovelace Brain Mapping Center, Departments of Neurology and of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, UCLALos AngelesUnited States
| | - Karl Zilles
- Institute of Neuroscience and Medicine INM-1, Research Centre JülichJülichGermany
- JARA - Translational Brain MedicineAachenGermany
| |
Collapse
|
32
|
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]
|
33
|
Wang X, Daley C, Gakhar V, Lange HS, Vardigan JD, Pearson M, Zhou X, Warren L, Miller CO, Belden M, Harvey AJ, Grishin AA, Coles CJ, O'Connor SM, Thomson F, Duffy JL, Bell IM, Uslaner JM. Pharmacological Characterization of the Novel and Selective α7 Nicotinic Acetylcholine Receptor-Positive Allosteric Modulator BNC375. J Pharmacol Exp Ther 2020; 373:311-324. [PMID: 32094294 DOI: 10.1124/jpet.119.263483] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/17/2020] [Indexed: 12/28/2022] Open
Abstract
Treatments for cognitive deficits associated with central nervous system (CNS) disorders such as Alzheimer disease and schizophrenia remain significant unmet medical needs that incur substantial pressure on the health care system. The α7 nicotinic acetylcholine receptor (nAChR) has garnered substantial attention as a target for cognitive deficits based on receptor localization, robust preclinical effects, genetics implicating its involvement in cognitive disorders, and encouraging, albeit mixed, clinical data with α7 nAChR orthosteric agonists. Importantly, previous orthosteric agonists at this receptor suffered from off-target activity, receptor desensitization, and an inverted U-shaped dose-effect curve in preclinical assays that limit their clinical utility. To overcome the challenges with orthosteric agonists, we have identified a novel selective α7 positive allosteric modulator (PAM), BNC375. This compound is selective over related receptors and potentiates acetylcholine-evoked α7 currents with only marginal effect on the receptor desensitization kinetics. In addition, BNC375 enhances long-term potentiation of electrically evoked synaptic responses in rat hippocampal slices and in vivo. Systemic administration of BNC375 reverses scopolamine-induced cognitive deficits in rat novel object recognition and rhesus monkey object retrieval detour (ORD) task over a wide range of exposures, showing no evidence of an inverted U-shaped dose-effect curve. The compound also improves performance in the ORD task in aged African green monkeys. Moreover, ex vivo 13C-NMR analysis indicates that BNC375 treatment can enhance neurotransmitter release in rat medial prefrontal cortex. These findings suggest that α7 nAChR PAMs have multiple advantages over orthosteric α7 nAChR agonists for the treatment of cognitive dysfunction associated with CNS diseases. SIGNIFICANCE STATEMENT: BNC375 is a novel and selective α7 nicotinic acetylcholine receptor (nAChR) positive allosteric modulator (PAM) that potentiates acetylcholine-evoked α7 currents in in vitro assays with little to no effect on the desensitization kinetics. In vivo, BNC375 demonstrated robust procognitive effects in multiple preclinical models across a wide exposure range. These results suggest that α7 nAChR PAMs have therapeutic potential in central nervous system diseases with cognitive impairments.
Collapse
Affiliation(s)
- Xiaohai Wang
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Christopher Daley
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Vanita Gakhar
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Henry S Lange
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Joshua D Vardigan
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Michelle Pearson
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Xiaoping Zhou
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Lee Warren
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Corin O Miller
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Michelle Belden
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Andrew J Harvey
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Anton A Grishin
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Carolyn J Coles
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Susan M O'Connor
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Fiona Thomson
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Joseph L Duffy
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Ian M Bell
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| | - Jason M Uslaner
- Merck Research Laboratories, Merck & Co., Inc., Kenilworth, New Jersey (X.W., C.D., V.G., H.S.L., J.D.V., M.P., X.Z., L.W., C.O.M., M.B., F.T., J.L.D., I.M.B., J.M.U.) and Bionomics Limited, Thebarton, Australia (A.J.H., A.A.G., C.J.C., S.M.O.)
| |
Collapse
|
34
|
Arnsten AFT, Datta D, Leslie S, Yang ST, Wang M, Nairn AC. Alzheimer's-like pathology in aging rhesus macaques: Unique opportunity to study the etiology and treatment of Alzheimer's disease. Proc Natl Acad Sci U S A 2019; 116:26230-26238. [PMID: 31871209 PMCID: PMC6936707 DOI: 10.1073/pnas.1903671116] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although mouse models of Alzheimer's disease (AD) have provided tremendous breakthroughs, the etiology of later onset AD remains unknown. In particular, tau pathology in the association cortex is poorly replicated in mouse models. Aging rhesus monkeys naturally develop cognitive deficits, amyloid plaques, and the same qualitative pattern and sequence of tau pathology as humans, with tangles in the oldest animals. Thus, aging rhesus monkeys can play a key role in AD research. For example, aging monkeys can help reveal how synapses in the prefrontal association cortex are uniquely regulated compared to the primary sensory cortex in ways that render them vulnerable to calcium dysregulation and tau phosphorylation, resulting in the selective localization of tau pathology observed in AD. The ability to assay early tau phosphorylation states and perform high-quality immunoelectron microscopy in monkeys is a great advantage, as one can capture early-stage degeneration as it naturally occurs in situ. Our immunoelectron microscopy studies show that phosphorylated tau can induce an "endosomal traffic jam" that drives amyloid precursor protein cleavage to amyloid-β in endosomes. As amyloid-β increases tau phosphorylation, this creates a vicious cycle where varied precipitating factors all lead to a similar phenotype. These data may help explain why circuits with aggressive tau pathology (e.g., entorhinal cortex) may degenerate prior to producing significant amyloid pathology. Aging monkeys therefore can play an important role in identifying and testing potential therapeutics to protect the association cortex, including preventive therapies that are challenging to test in humans.
Collapse
Affiliation(s)
- Amy F. T. Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
| | - Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Shannon Leslie
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
| | - Sheng-Tao Yang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510
| | - Angus C. Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06510
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
35
|
Oligomeric Aβ in the monkey brain impacts synaptic integrity and induces accelerated cortical aging. Proc Natl Acad Sci U S A 2019; 116:26239-26246. [PMID: 31871145 DOI: 10.1073/pnas.1902301116] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
As the average age of the population continues to rise, the number of individuals affected with age-related cognitive decline and Alzheimer's disease (AD) has increased and is projected to cost more than $290 billion in the United States in 2019. Despite significant investment in research over the last decades, there is no effective treatment to prevent or delay AD progression. There is a translational gap in AD research, with promising drugs based on work in rodent models failing in clinical trials. Aging is the leading risk factor for developing AD and understanding neurobiological changes that affect synaptic integrity with aging will help clarify why the aged brain is vulnerable to AD. We describe here the development of a rhesus monkey model of AD using soluble oligomers of the amyloid beta (Aβ) peptide (AβOs). AβOs infused into the monkey brain target a specific population of spines in the prefrontal cortex, induce neuroinflammation, and increase AD biomarkers in the cerebrospinal fluid to similar levels observed in patients with AD. Importantly, AβOs lead to similar dendritic spine loss to that observed in normal aging in monkeys, but so far without detection of amyloid plaques or tau pathology. Understanding the basis of synaptic impairment is the most effective route to early intervention and prevention or postponement of age-related cognitive decline and transition to AD. These initial findings support the use of monkeys as a platform to understand age-related vulnerabilities of the primate brain and may help develop effective disease-modifying therapies for treatment of AD and related dementias.
Collapse
|
36
|
Devinsky O, Boesch JM, Cerda-Gonzalez S, Coffey B, Davis K, Friedman D, Hainline B, Houpt K, Lieberman D, Perry P, Prüss H, Samuels MA, Small GW, Volk H, Summerfield A, Vite C, Wisniewski T, Natterson-Horowitz B. A cross-species approach to disorders affecting brain and behaviour. Nat Rev Neurol 2019; 14:677-686. [PMID: 30287906 DOI: 10.1038/s41582-018-0074-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Structural and functional elements of biological systems are highly conserved across vertebrates. Many neurological and psychiatric conditions affect both humans and animals. A cross-species approach to the study of brain and behaviour can advance our understanding of human disorders via the identification of unrecognized natural models of spontaneous disorders, thus revealing novel factors that increase vulnerability or resilience, and via the assessment of potential therapies. Moreover, diagnostic and therapeutic advances in human neurology and psychiatry can often be adapted for veterinary patients. However, clinical and research collaborations between physicians and veterinarians remain limited, leaving this wealth of comparative information largely untapped. Here, we review pain, cognitive decline syndromes, epilepsy, anxiety and compulsions, autoimmune and infectious encephalitides and mismatch disorders across a range of animal species, looking for novel insights with translational potential. This comparative perspective can help generate novel hypotheses, expand and improve clinical trials and identify natural animal models of disease resistance and vulnerability.
Collapse
Affiliation(s)
- Orrin Devinsky
- Department of Neurology, New York University (NYU) Langone Medical Center and NYU School of Medicine, New York, NY, USA.
| | - Jordyn M Boesch
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | - Barbara Coffey
- Department of Child and Adolescent Psychiatry, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kathryn Davis
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Friedman
- Department of Neurology, New York University (NYU) Langone Medical Center and NYU School of Medicine, New York, NY, USA
| | - Brian Hainline
- Department of Neurology, New York University (NYU) Langone Medical Center and NYU School of Medicine, New York, NY, USA
| | - Katherine Houpt
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Daniel Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Pamela Perry
- College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Harald Prüss
- Department of Neurology with Experimental Neurology, Charité University Medicine Berlin, Berlin, Germany, and German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | | | - Gary W Small
- University of California-Los Angeles (UCLA) Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Holger Volk
- Veterinary Neurology and Neurosurgery, The Royal Veterinary College, University of London, London, UK
| | - Artur Summerfield
- Institute of Virology and Immunology and Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Charles Vite
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Wisniewski
- Department of Neurology, New York University (NYU) Langone Medical Center and NYU School of Medicine, New York, NY, USA
| | - Barbara Natterson-Horowitz
- Department of Ecology and Evolutionary Biology, Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
37
|
Lyra E Silva NDM, Gonçalves RA, Boehnke SE, Forny-Germano L, Munoz DP, De Felice FG. Understanding the link between insulin resistance and Alzheimer's disease: Insights from animal models. Exp Neurol 2019; 316:1-11. [PMID: 30930096 DOI: 10.1016/j.expneurol.2019.03.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease affecting millions of people worldwide. AD is characterized by a profound impairment of higher cognitive functions and still lacks any effective disease-modifying treatment. Defective insulin signaling has been implicated in AD pathophysiology, but the mechanisms underlying this process are not fully understood. Here, we review the molecular mechanisms underlying defective brain insulin signaling in rodent models of AD, and in a non-human primate (NHP) model of the disease that recapitulates features observed in AD brains. We further highlight similarities between the NHP and human brains and discuss why NHP models of AD are important to understand disease mechanisms and to improve the translation of effective therapies to humans. We discuss how studies using different animal models have contributed to elucidate the link between insulin resistance and AD.
Collapse
Affiliation(s)
| | | | - Susan E Boehnke
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada.
| | - Fernanda G De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada; Department of Psychiatry, Queen's University, Kingston, ON, Canada; Institute of Medical Biochemistry Leopoldo De Meis, Federal University of Rio de Janeiro, Brazil.
| |
Collapse
|
38
|
Crimins JL, Puri R, Calakos KC, Yuk F, Janssen WGM, Hara Y, Rapp PR, Morrison JH. Synaptic distributions of pS214-tau in rhesus monkey prefrontal cortex are associated with spine density, but not with cognitive decline. J Comp Neurol 2019; 527:856-873. [PMID: 30408169 PMCID: PMC6333519 DOI: 10.1002/cne.24576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/18/2018] [Accepted: 10/21/2018] [Indexed: 12/31/2022]
Abstract
Female rhesus monkeys and women are subject to age- and menopause-related deficits in working memory, an executive function mediated by the dorsolateral prefrontal cortex (dlPFC). Long-term cyclic administration of 17β-estradiol improves working memory, and restores highly plastic axospinous synapses within layer III dlPFC of aged ovariectomized monkeys. In this study, we tested the hypothesis that synaptic distributions of tau protein phosphorylated at serine 214 (pS214-tau) are altered with age or estradiol treatment, and couple to working memory performance. First, ovariectormized young and aged monkeys received vehicle or estradiol treatment, and were tested on the delayed response (DR) test of working memory. Serial section electron microscopic immunocytochemistry was then performed to quantitatively assess the subcellular synaptic distributions of pS214-tau. Overall, the majority of synapses contained pS214-tau immunogold particles, which were predominantly localized to the cytoplasm of axon terminals. pS214-tau was also abundant within synaptic and cytoplasmic domains of dendritic spines. The density of pS214-tau immunogold within the active zone, cytoplasmic, and plasmalemmal domains of axon terminals, and subjacent to the postsynaptic density within the subsynaptic domains of dendritic spines, were each reduced with age. None of the variables examined were directly linked to cognitive status, but a high density of pS214-tau immunogold particles within presynaptic cytoplasmic and plasmalemmal domains, and within postsynaptic subsynaptic and plasmalemmal domains, accompanied high synapse density. Together, these data support a possible physiological, rather than pathological, role for pS214-tau in the modulation of synaptic morphology in monkey dlPFC.
Collapse
Affiliation(s)
- Johanna L. Crimins
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Rishi Puri
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Katina C. Calakos
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Frank Yuk
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - William G. M. Janssen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter R. Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224
| | - John H. Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- California National Primate Research Center, Davis, CA 95616
- Department of Neurology, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
39
|
Beckman D, Baxter MG, Morrison JH. Future directions in animal models of Alzheimer's disease. J Neurosci Res 2018; 96:1829-1830. [PMID: 30204256 DOI: 10.1002/jnr.24328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California
| | - Mark G Baxter
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John H Morrison
- California National Primate Research Center, UC Davis, Davis, California.,Department of Neurology, School of Medicine, UC Davis, Davis, California
| |
Collapse
|
40
|
Pérez MJ, Jara C, Quintanilla RA. Contribution of Tau Pathology to Mitochondrial Impairment in Neurodegeneration. Front Neurosci 2018; 12:441. [PMID: 30026680 PMCID: PMC6041396 DOI: 10.3389/fnins.2018.00441] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022] Open
Abstract
Tau is an essential protein that physiologically promotes the assembly and stabilization of microtubules, and participates in neuronal development, axonal transport, and neuronal polarity. However, in a number of neurodegenerative diseases, including Alzheimer’s disease (AD), tau undergoes pathological modifications in which soluble tau assembles into insoluble filaments, leading to synaptic failure and neurodegeneration. Mitochondria are responsible for energy supply, detoxification, and communication in brain cells, and important evidence suggests that mitochondrial failure could have a pivotal role in the pathogenesis of AD. In this context, our group and others investigated the negative effects of tau pathology on specific neuronal functions. In particular, we observed that the presence of these tau forms could affect mitochondrial function at three different levels: (i) mitochondrial transport, (ii) morphology, and (iii) bioenergetics. Therefore, mitochondrial dysfunction mediated by anomalous tau modifications represents a novel mechanism by which these forms contribute to the pathogenesis of AD. In this review, we will discuss the main results reported on pathological tau modifications and their effects on mitochondrial function and their importance for the synaptic communication and neurodegeneration.
Collapse
Affiliation(s)
- María J Pérez
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
| | - Claudia Jara
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Universidad Autónoma de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes (CIAA), Santiago, Chile
| |
Collapse
|