Elsevier

Neurobiology of Aging

Volume 59, November 2017, Pages 107-120
Neurobiology of Aging

Regular article
Aged chimpanzees exhibit pathologic hallmarks of Alzheimer's disease

https://doi.org/10.1016/j.neurobiolaging.2017.07.006Get rights and content

Abstract

Alzheimer's disease (AD) is a uniquely human brain disorder characterized by the accumulation of amyloid-beta protein (Aβ) into extracellular plaques, neurofibrillary tangles (NFT) made from intracellular, abnormally phosphorylated tau, and selective neuronal loss. We analyzed a large group of aged chimpanzees (n = 20, age 37–62 years) for evidence of Aβ and tau lesions in brain regions affected by AD in humans. Aβ was observed in plaques and blood vessels, and tau lesions were found in the form of pretangles, NFT, and tau-immunoreactive neuritic clusters. Aβ deposition was higher in vessels than in plaques and correlated with increases in tau lesions, suggesting that amyloid build-up in the brain's microvasculature precedes plaque formation in chimpanzees. Age was correlated to greater volumes of Aβ plaques and vessels. Tangle pathology was observed in individuals that exhibited plaques and moderate or severe cerebral amyloid angiopathy, a condition in which amyloid accumulates in the brain's vasculature. Amyloid and tau pathology in aged chimpanzees suggests these AD lesions are not specific to the human brain.

Introduction

Alzheimer's disease (AD) is a progressive, irreversible proteinopathy, resulting in cognitive impairment, and characterized by pathologic aggregations of the amyloid-beta (Aβ) and tau proteins in the form of plaques and neurofibrillary tangles (NFTs) and selective neuronal loss (Bussière et al., 2003a, Bussière et al., 2003b; Montine et al., 2012). It has been suggested that humans are uniquely susceptible to AD, potentially due to genetic differences, changes in cerebral structure and function during evolution, and an increased life span (Hof et al., 2002, Rapoport and Nelson, 2011, Sherwood et al., 2011, Walker and Cork, 1999). Although Aβ and tau pathology have been identified in nonhuman primates, these animals typically present with only amyloid plaque pathology (Head et al., 2001, Heuer et al., 2012, Hof et al., 2002, Oikawa et al., 2010, Walker and Cork, 1999). In addition, aged primates demonstrate cognitive deficits in working memory, learning tasks, long-term retention, and cognitive flexibility similar to elderly humans (Joly et al., 2014, Lacreuse et al., 2014, Nagahara et al., 2010, Picq et al., 2015). Previous studies in aged monkeys and great apes confirmed the presence of diffuse and neuritic amyloid plaques as well as vascular amyloid (Gearing et al., 1994, Gearing et al., 1996, Gearing et al., 1997, Geula et al., 2002, Heuer et al., 2012, Kimura et al., 2003, Lemere et al., 2004, Lemere et al., 2008, Martin et al., 1991, Mufson et al., 1994, Oikawa et al., 2010, Perez et al., 2013, Perez et al., 2016, Poduri et al., 1994, Rosen et al., 2008, Walker et al., 1987). The presence of abnormally phosphorylated tau has been reported in neurons and glia in rhesus monkeys (Macaca mulatta) and squirrel monkeys (Saimiri sciureus) as well as baboons (Papio anubis) (Elfenbein et al., 2007, Hartig et al., 2000, Oikawa et al., 2010, Schultz et al., 2000, Selkoe et al., 1987). In great apes, captive and wild gorillas (Gorilla gorilla gorilla and Gorilla beringei beringei) exhibited hyperphosphorylated tau-immunoreactive (ir) neurons in the neocortex in close association with Aβ plaques, but the perikarya lacked classic AD tau histopathologic abnormalities (Perez et al., 2013, Perez et al., 2016). Several pathologic examinations of the chimpanzee (Pan troglodytes) and orangutan (Pongo pygmaeus) brain revealed diffuse Aβ plaques and vascular amyloid, but not tau-positive NFT and neuropil threads (Gearing et al., 1994, Gearing et al., 1996, Gearing et al., 1997). The only evidence of NFT and Aβ pathology coexisting in a primate species other than humans was reported in a 41-year-old female chimpanzee that suffered a stroke before death and had a history of high cholesterol (Rosen et al., 2008). This ape presented with scarce diffuse senile plaques and vascular amyloid, NFT, neuropil threads, and clusters of tau-positive neurites in the neocortex, which may have been the result of a rare ischemic event, as risk of dementia after stroke is increased twofold in humans (Schneider and Bennett, 2010).

Chimpanzees share 100% sequence homology and all 6 tau isoforms with humans (Chimpanzee Sequencing and Analysis Consortium, 2005, Holzer et al., 2004, Nelson et al., 2009). Moreover, amyloid precursor protein 695 (APP695) is more than 99% identical in chimpanzees and humans and can be cleaved into Aβ 40/42 peptides with 100% sequence homology to humans (Cervenáková et al., 1994, Götz and Ittner, 2008, Heuer et al., 2012). As one of our closest living primate relatives, chimpanzees demonstrate greater behavioral complexity and longer life spans than nonprimate AD animal models (Hakeem et al., 1996, Hill et al., 2001). In captivity, the average life span for chimpanzees ranges from 30 to 35 years with the oldest recorded age of 62 years (Dyke et al., 1995). Therefore, chimpanzees are ideal candidates to investigate the pathobiology of AD from an evolutionary neurobiological perspective.

Brain samples from great apes, particularly aged individuals, are incredibly scarce. In the wild, chimpanzees have an average life expectancy of 33 years (maximum age noted was 53 years), and only 10% of captive male chimpanzees survive to age 45 and captive females to age 55 (Dyke et al., 1995, Hill et al., 2001, Wood et al., 2017). In addition, prior neuropathologic studies in apes were limited to small samples sizes. The present study evaluated 4 brain regions affected by AD pathology in humans—prefrontal cortex (PFC), middle temporal gyrus (MTG), and CA1 and CA3 hippocampal subfields—in a large group of aged chimpanzees (Table 1) for Aβ-positive plaques, vascular amyloid, and tau lesions (i.e., pretangles, NFT, and neuritic clusters). We also developed an overall pathology scoring system based on clinical staging parameters for Aβ and NFT deposition in humans (see Section 2.6 and Table 2) (Attems et al., 2007, Braak and Braak, 1991, Braak et al., 2006, Mirra et al., 1991, Montine et al., 2012, Thal et al., 2002, Vonsattel et al., 1991). Here, we show that pretangles, NFT, and clusters of tau-ir dystrophic neurites (NC) co-occur with amyloid-positive plaques and vasculature in aged chimpanzees. These findings indicate that AD-like pathology is not limited to the human brain.

Section snippets

Specimens and sample processing

Postmortem brain specimens from 20 aged chimpanzees (Table 1) were acquired from Association of Zoos and Aquariums–accredited zoos or American Association for Accreditation of Laboratory Animal Care–accredited research institutions and maintained in accordance with each institution's animal care guidelines. Chimpanzee brain specimens were provided by the National Chimpanzee Brain Resource (supported by NIH grant NS092988), and health information has been included when possible (Table S1). The

APP/Aβ and Aβ42 plaque volume

To evaluate Aβ protein aggregates in the aging chimpanzee brain, we measured volume occupied by plaques using immunostaining against 2 peptides, APP/Aβ and Aβ42. Of the 20 chimpanzees, 13 had APP/Aβ-ir plaques (Fig. 1A–B, E), whereas 5 of those individuals also presented with Aβ42-ir plaques (Fig. 1C–D, F). Of those with Aβ42 plaques, 4 were the oldest individuals (57–62 years). Aβ42 plaques were not found in the youngest subjects (39–44 years). Diffuse and dense-core plaques were identified in

Discussion

To date, fewer than 50 brains of aged apes have been examined for AD pathology across all prior studies (Finch and Austad, 2015). Here, we present an investigation of AD neuropathology in the largest cohort of any great ape species consisting of 20 chimpanzee brains aged 37–62 years. Our findings establish that aged chimpanzees can exhibit the 2 main histological markers found in the brain of humans with AD, Aβ plaques, and NFT.

APP/Aβ and Aβ42-ir diffuse and dense-core plaques were most

Conclusions

Tau and Aβ lesions were identified in the neocortex and hippocampus in a large cohort of aged chimpanzees. Surprisingly, AT8-ir pretangles and Aβ-positive vessels were exhibited in all 20 apes, whereas Aβ plaques were found in two-thirds of individuals. We also demonstrated thioflavin S-positive NFTs in aged chimpanzees, although electron microscopic analysis is planned to confirm filament structure. Neuritic plaques with an Aβ core were absent, but clusters of AT8-ir tau neurites were

Acknowledgements

The authors would like to thank Jennifer Chilton for her assistance with health records, Dr. Jason R. Richardson for his editorial revisions, Dr. Peter Davies for providing tau antibodies, Dr. Michael Model for his confocal microscopy expertise, and Cheryl Stimpson, Bridget Wicinski, and Emily Munger for their technical assistance. Supported by grants from the NSF (BCS-1316829 to MAR), NIH (NS042867, NS073134, and NS092988 to WDH and CCS; AG017802 to JJE; AG014308 to JME; AG005138 to PRH; and

References (102)

  • S.A. Gravina et al.

    Amyloid beta protein (A beta) in Alzheimer’s disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at A beta 40 or A beta 42(43)

    J. Biol. Chem.

    (1995)
  • E. Head et al.

    Neurobiological models of aging in the dog and other vertebrate species

  • J.G. Herndon et al.

    Patterns of cognitive decline in aged rhesus monkeys

    Behav. Brain Res.

    (1997)
  • K. Hill et al.

    Mortality rates among wild chimpanzees

    J. Hum. Evol.

    (2001)
  • M. Holzer et al.

    Tau gene (MAPT) sequence variation among primates

    Gene

    (2004)
  • S. Itagaki et al.

    Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease

    J. Neuroimmunol

    (1989)
  • N. Kimura et al.

    Age-related changes of Alzheimer’s disease-associated proteins in cynomolgus monkey brains

    Biochem. Biophys. Res. Commun.

    (2003)
  • A. Lacreuse et al.

    Cognitive and motor aging in female chimpanzees

    Neurobiol. Aging

    (2014)
  • C.A. Lemere et al.

    Alzheimer’s disease abeta vaccine reduces central nervous system abeta levels in a non-human primate, the Caribbean vervet

    Am. J. Pathol.

    (2004)
  • T.L. Moore et al.

    Impairment in abstraction and set shifting in aged rhesus monkeys

    Neurobiol. Aging

    (2003)
  • T.L. Moore et al.

    Executive system dysfunction occurs as early as middle-age in the rhesus monkey

    Neurobiol. Aging

    (2006)
  • M.B. Moss et al.

    Recognition memory span in rhesus monkeys of advanced age

    Neurobiol. Aging

    (1997)
  • E.J. Mufson et al.

    Apolipoprotein E-immunoreactivity in aged rhesus monkey cortex: colocalization with amyloid plaques

    Neurobiol. Aging

    (1994)
  • A.H. Nagahara et al.

    Age-related cognitive deficits in rhesus monkeys mirrors human deficits on an automated test battery

    Neurobiol. Aging

    (2010)
  • N. Oikawa et al.

    Alzheimer-type tau pathology in advanced aged nonhuman primate brains harboring substantial amyloid deposition

    Brain Res.

    (2010)
  • S.E. Perez et al.

    Early Alzheimer’s disease-type pathology in the frontal cortex of wild mountain gorillas (Gorilla beringei beringei)

    Neurobiol. Aging

    (2016)
  • S.K. Presty et al.

    Age differences in recognition memory of the rhesus monkey (Macaca mulatta)

    Neurobiol. Aging

    (1987)
  • M.A. Raghanti et al.

    Species-specific distributions of tyrosine hydroxylase-immunoreactive neurons in the prefrontal cortex of anthropoid primates

    Neuroscience

    (2009)
  • M.A. Raghanti et al.

    Cortical dopaminergic innervation among humans, chimpanzees, and macaque monkeys: a comparative study

    Neuroscience

    (2008)
  • S.I. Rapoport et al.

    Biomarkers and evolution in Alzheimer disease

    Prog. Neurobiol.

    (2011)
  • P.R. Rapp et al.

    Recognition memory deficits in a subpopulation of aged monkeys resemble the effects of medial temporal lobe damage

    Neurobiol. Aging

    (1991)
  • C. Schultz et al.

    Age-related progression of tau pathology in brains of baboons

    Neurobiol. Aging

    (2000)
  • C.C. Sherwood et al.

    Is humanlike cytoarchitectural asymmetry present in another species with complex social vocalization? A stereologic analysis of mustached bat auditory cortex

    Brain Res.

    (2005)
  • L. Slomianka et al.

    Estimators of the precision of stereological estimates: an example based on the CA1 pyramidal cell layer of rats

    Neuroscience

    (2005)
  • L.C. Walker et al.

    The exceptional Vulnerability of humans to Alzheimer’s disease

    Trends Mol. Med.

    (2017)
  • L.C. Walker et al.

    Senile plaques in aged squirrel monkeys

    Neurobiol. Aging

    (1987)
  • B.M. Wood et al.

    Favorable ecological circumstances promote life expectancy in chimpanzees similar to that of human hunter-gatherers

    J. Hum. Evol.

    (2017)
  • U.S. Anderson et al.

    Relative numerousness judgment and summation in young and old Western lowland gorillas

    J. Comp. Psychol.

    (2005)
  • P.V. Arriagada et al.

    Distribution of Alzheimer-type pathologic changes in nondemented elderly individuals matches the pattern in Alzheimer’s disease

    Neurology

    (1992)
  • R.T. Bartus et al.

    Aging in the rhesus monkey: debilitating effects on short-term memory

    J. Gerontol.

    (1978)
  • I. Bernstein

    Response variability and rigidity in the adult chimpanzee

    J. Gerontol.

    (1961)
  • H. Braak et al.

    Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry

    Acta Neuropathol.

    (2006)
  • H. Braak et al.

    Neuropathological stageing of Alzheimer-related changes

    Acta Neuropathol.

    (1991)
  • H. Braak et al.

    Stages of the pathologic process in Alzheimer's disease

    J. Neuropathol. Exp. Neurol.

    (2011)
  • T. Bussière et al.

    Progressive degeneration of nonphosphorylated neurofilament protein-enriched pyramidal neurons predicts cognitive impairment in Alzheimer's disease: stereologic analysis of prefrontal cortex area 9

    J. Comp. Neurol.

    (2003)
  • T. Bussière et al.

    Stereologic analysis of neurofibrillary tangle formation in prefrontal cortex area 9 in aging and Alzheimer’s disease

    Neuroscience

    (2003)
  • L. Cervenáková et al.

    Infectious amyloid precursor gene sequences in primates used for experimental transmission of human spongiform encephalopathy

    Proc. Natl. Acad. Sci. U. S. A.

    (1994)
  • Initial sequence of the chimpanzee genome and comparison with the human genome

    Nature

    (2005)
  • K.A. Dorph-Petersen et al.

    Tissue shrinkage and unbiased stereological estimation of particle number and size

    J. Microsc.

    (2001)
  • B. Dyke et al.

    Model life table for captive chimpanzees

    Am. J. Primatol

    (1995)
  • Cited by (89)

    View all citing articles on Scopus
    View full text