Elsevier

Neurobiology of Aging

Volume 8, Issue 4, July–August 1987, Pages 291-296
Neurobiology of Aging

Senile plaques in aged squirrel monkeys

https://doi.org/10.1016/0197-4580(87)90067-4Get rights and content

Abstract

Aged squirrel monkeys develop senile plaques in the brain that are similar to those occurring in aged rhesus monkeys and aged humans. These plaques consist of abnormal, swollen neurites around an amyloid core. In whole-hemisphere coronal sections through the level of the rostral temporal lobe, plaques are present in temporal cortex, amygdala, hippocampal formation and, occasionally, in other cortical regions. In more rostral sections through the frontal lobe, plaques are most common in orbitofrontal and frontal opercular cortical regions. In immunocytochemical preparations, some neurites show immunoreactivity with antibodies directed against phosphorylated neurofilaments and neuropeptide Y. Thus, plaques in these New World primates are similar in distribution and composition to those occurring in aged Old World primates.

References (39)

  • A. Alzheimer

    Über eine eigenartige Erkrankung der Hirnrinde

    Zentralbl Nervenheilkunde Psychiatrie

    (1907)
  • D.M. Bowden et al.

    Aging research in nonhuman primates

  • V. Chan-Palay et al.

    Cortical neurons immunoreactive with antisera against neuropeptide Y are altered in Alzheimer's-type dementia

    J Comp Neurol

    (1985)
  • V. Chan-Palay et al.

    Distribution of altered hippocampal neurons and axons immunoreactive with antisera against neuropeptide Y in Alzheimer's-type dementia

    J Comp Neurol

    (1986)
  • A.D. Dayan

    Comparative neuropathology of ageing

  • B. Dyke et al.

    A demographic analysis of the Wisconsin Regional Primate Center rhesus colony, 1962–1982

    Am J Primatol

    (1986)
  • O. Fischer

    Miliare Nekrosen mit drusigen Wucherungen der Neurofibrillen, eine regelmässige Veränderung der Hirnrinde bei seniler Demenz

    Monatsschr Psychiatry Neurol

    (1907)
  • R.L. Friede

    Enzyme histochemical studies of senile plaques

    J Neuropathol Exp Neurol

    (1965)
  • S. Hopf et al.

    Life span in captive squirrel monkeys (Saimiri) with pathological and reproductive records

    Primates

    (1979)
  • Cited by (74)

    • Aging African green monkeys manifest transcriptional, pathological, and cognitive hallmarks of human Alzheimer's disease

      2018, Neurobiology of Aging
      Citation Excerpt :

      Hyperphosphorylated tau is even less frequently observed in the brains of aged rhesus and cynomolgus macaque (Hartig et al., 2000; Oikawa et al., 2010), and no evidence of tangles have been published for these species. Similar findings have been reported in squirrel monkeys with regard to Aβ and tau pathologies (Selkoe et al., 1987; Walker et al., 1987, 1990). Thus, within old- and new-world monkeys, the AGM appears rather unique with respect to the high prevalence of dense-core plaques, as well as the occurrence, albeit still rather infrequent, of hyperphosphorylated tau tangle-like structures later in life (Lemere et al., 2004, 2006).

    • Aged chimpanzees exhibit pathologic hallmarks of Alzheimer's disease

      2017, Neurobiology of Aging
      Citation Excerpt :

      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, 1996, 1997; Geula et al., 2002; Heuer et al., 2012; Kimura et al., 2003; Lemere et al., 2004, 2008; Martin et al., 1991; Mufson et al., 1994; Oikawa et al., 2010; Perez et al., 2013, 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).

    • Comparative pathobiology of β-amyloid and the unique susceptibility of humans to Alzheimer's disease

      2016, Neurobiology of Aging
      Citation Excerpt :

      The paradoxical existence of extensive cerebral Aβ-amyloidosis without overt neurodegeneration and dementia in animal models (and possibly humans) might be reconciled by differences in the post-translational characteristics of Aβ, such as species-specific populations of Aβ isoforms or the formation of structurally and functionally distinct proteopathic “strains” (Fritschi et al., 2014; Hatami et al., 2014; Heilbronner et al., 2013; Levine and Walker, 2010; Lu et al., 2013; Mehta et al., 2013; Meyer-Luehmann et al., 2006; Petkova et al., 2005; Rosen et al., 2010a, 2011; Stohr et al., 2014; Watts et al., 2014). To gain insight into the comparative pathobiology of Aβ in a species proximal to humans, we analyzed the properties of Aβ in the brains of humans with AD and aged squirrel monkeys (Saimiri sciureus), a New World primate that develops senile plaques and CAA with advancing age (Elfenbein et al., 2007; Walker et al., 1987, 1990). We found that the populations of Aβ and its post-translationally variant isoforms are remarkably similar in humans and squirrel monkeys, and that Aβ-rich cortical extracts from aged squirrel monkeys effectively seed the deposition of Aβ in an APP-transgenic mouse model.

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

      2016, Neurobiology of Aging
      Citation Excerpt :

      Although neurofibrillary tangles (NFTs) are a major pathological feature in human neurodegenerative conditions (Bouras et al., 1994; Braak and Braak, 1991; Jackson and Lowe, 1996), mainly beta amyloid (Aβ) deposits (neuritic and diffuse plaques), and Aβ angiopathy have been reported in several nonhuman primate species. Most non-human primate studies have been carried out in captive rhesus monkeys (Gearing et al., 1996; Martin et al., 1991; Mufson et al., 1994; Norvin et al., 2015; Poduri et al., 1994), long-tailed macaques (Kimura et al., 2003), Caribbean vervets (Lemere et al., 2004), squirrel monkeys (Walker et al., 1987), and cotton-top tamarins (Lemere et al., 2008), and a few in great apes (Edler et al., 2015; Gearing et al., 1994, 1996, 1997; Kimura et al., 2001; Perez et al., 2013; Rosen et al., 2008). Great apes, which include common chimpanzees, bonobos, gorillas, and orangutans, are of significant interest in aging and AD research because they are the closest living relatives of humans and therefore could provide clues to the evolutionary changes underlying the onset of AD pathology.

    • Should Alzheimer's disease be equated with human brain ageing?: A maladaptive interaction between brain evolution and senescence

      2012, Ageing Research Reviews
      Citation Excerpt :

      Amyloid plaques similar to Alzheimer's type NP have been detected in the cortex of some aged non-human mammalian species (Struble et al., 1985; Wisniewski et al., 1970; Selkoe et al., 1987) however this generally occurs in the absence of NFT formation. It is highly significant that primate relatives such as orangutans (Gearing et al., 1997), rhesus monkeys (Poduri et al., 1994; Uno et al., 1996), squirrel monkeys (Walker et al., 1987) and our closest relative the chimpanzee (Gearing et al., 1994), whilst developing NP with age, do not develop NFT with age. This is despite having similar tau sequence and expression profiles to humans (Holzer et al., 2004; Janke et al., 1999).

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

      2010, Brain Research
      Citation Excerpt :

      Distinct from Tg mice, both the production of Aβ and expression of tau are likely within physiological ranges in nontransgenic animals; thus, the natural spatiotemporal profiles of Aβ and tau pathologies should be presented in the brains of these animals. To date, it has been reported that a lot of mammalian species exhibit cerebral Aβ-amyloidosis, such as parenchymal Aβ deposition and amyloid angiopathy, and intraneuronal tau accumulation (Bons et al., 2006; Braak et al., 1994; Cummings et al., 1996a,b; Gearing et al., 1994; Geula et al., 2002; Härtig et al., 2000; Lemere et al., 2004, 2008; Podlisny et al., 1991; Roertgen et al., 1996; Schultz et al., 2000a,b; Selkoe et al., 1987; Walker et al., 1987). Among those animals, we paid particular attention to cynomolgus monkey because of the similarity of Aβ pathology as follows.

    View all citing articles on Scopus
    View full text