Skip to main content

Advertisement

Log in

Primary neuron and astrocyte cultures from postnatal Callithrix jacchus: a non-human primate in vitro model for research in neuroscience, nervous system aging, and neurological diseases of aging

  • Methods Paper
  • Published:
GeroScience Aims and scope Submit manuscript

A Correction to this article was published on 02 February 2022

This article has been updated

Abstract

The ability to generate in vitro cultures of neuronal cells has been instrumental in advancing our understanding of the nervous system. Rodent models have been the principal source of brain cells used in primary cultures for over a century, providing insights that are widely applicable to human diseases. However, therapeutic agents that showed benefit in rodent models, particularly those pertaining to aging and age-associated dementias, have frequently failed in clinical trials. This discrepancy established a potential “translational gap” between human and rodent studies that may at least partially be explained by the phylogenetic distance between rodent and primate species. Several non-human primate (NHP) species, including the common marmoset (Callithrix jacchus), have been used extensively in neuroscience research, but in contrast to rodent models, practical approaches to the generation of primary cell culture systems amenable to molecular studies that can inform in vivo studies are lacking. Marmosets are a powerful model in biomedical research and particularly in studies of aging and age-associated diseases because they exhibit an aging phenotype similar to humans. Here, we report a practical method to culture primary marmoset neurons and astrocytes from brains of medically euthanized postnatal day 0 (P0) marmoset newborns that yield highly pure primary neuron and astrocyte cultures. Primary marmoset neuron and astrocyte cultures can be generated reliably to provide a powerful NHP in vitro model in neuroscience research that may enable mechanistic studies of nervous system aging and of age-related neurodegenerative disorders. Because neuron and astrocyte cultures can be used in combination with in vivo approaches in marmosets, primary marmoset neuron and astrocyte cultures may help bridge the current translational gap between basic and clinical studies in nervous system aging and age-associated neurological diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Change history

References

  1. Bracken MB. Why animal studies are often poor predictors of human reactions to exposure. J R Soc Med. 2009;102(3):120–2.

    PubMed  PubMed Central  Google Scholar 

  2. Zeiss CJ, Allore HG, Beck AP. Established patterns of animal study design undermine translation of disease-modifying therapies for Parkinson’s disease. PLoS One. 2017;12(2):e0171790.

    PubMed  PubMed Central  Google Scholar 

  3. Abbott DH, Barnett DK, Colman RJ, Yamamoto ME, Schultz-Darken NJ. Aspects of common marmoset basic biology and life history important for biomedical research. Comp Med. 2003;53(4):339–50.

    CAS  PubMed  Google Scholar 

  4. Kishi N, Sato K, Sasaki E, Okano H. Common marmoset as a new model animal for neuroscience research and genome editing technology. Develop Growth Differ. 2014;56(1):53–62.

    CAS  Google Scholar 

  5. Ross C, Salmon AB. Aging research using the common marmoset: focus on aging interventions. J Nutr Health Aging. 2019;5(2):97–109.

    Google Scholar 

  6. Munger EL, Takemoto A, Raghanti MA, Nakamura K. Visual discrimination and reversal learning in aged common marmosets (Callithrix jacchus). Neurosci Res. 2017;124:57–62.

    PubMed  Google Scholar 

  7. Phillips KA, Watson CM, Bearman A, Knippenberg AR, Adams J, Ross C, et al. Age-related changes in myelin of axons of the corpus callosum and cognitive decline in common marmosets. Am J Primatol. 2019;81(2):e22949.

    PubMed  PubMed Central  Google Scholar 

  8. Ross CN, Adams J, Gonzalez O, Dick E, Giavedoni L, Hodara VL, et al. Cross-sectional comparison of health-span phenotypes in young versus geriatric marmosets. Am J Primatol. 2019;81(2):e22952.

    PubMed  PubMed Central  Google Scholar 

  9. Sadoun A, Rosito M, Fonta C, Girard P. Key periods of cognitive decline in a nonhuman primate model of cognitive aging, the common marmoset (Callithrix jacchus). Neurobiol Aging. 2019;74:1–14.

    PubMed  Google Scholar 

  10. Tardif SD, Mansfield KG, Ratnam R, Ross CN, Ziegler TE. The marmoset as a model of aging and age-related diseases. ILAR J. 2011;52(1):54–65.

    CAS  PubMed  Google Scholar 

  11. Miller CT, Freiwald WA, Leopold DA, Mitchell JF, Silva AC, Wang X. Marmosets: a neuroscientific model of human social behavior. Neuron. 2016;90(2):219–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Mashiko H, Yoshida AC, Kikuchi SS, Niimi K, Takahashi E, Aruga J, et al. Comparative anatomy of marmoset and mouse cortex from genomic expression. J Neurosci. 2012;32(15):5039–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Tardif SD. Marmosets as a translational aging model-introduction. Am J Primatol. 2019;81(2):e22912.

    PubMed  PubMed Central  Google Scholar 

  14. Finch CE, Austad SN. Primate aging in the mammalian scheme: the puzzle of extreme variation in brain aging. Age (Dordr). 2012;34(5):1075–91.

    CAS  Google Scholar 

  15. Choudhury GR, Daadi MM. Charting the onset of Parkinson-like motor and non-motor symptoms in nonhuman primate model of Parkinson’s disease. PLoS One. 2018;13(8):e0202770.

    PubMed  PubMed Central  Google Scholar 

  16. Van Skike CE, et al. Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer’s disease and vascular cognitive impairment. Am J Physiol Heart Circ Physiol. 2018;314(4):H693–703.

    PubMed  Google Scholar 

  17. Pomilio C, Gorojod RM, Riudavets M, Vinuesa A, Presa J, Gregosa A, et al. Microglial autophagy is impaired by prolonged exposure to beta-amyloid peptides: evidence from experimental models and Alzheimer’s disease patients. Geroscience. 2020;42(2):613–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Schafer MJ, LeBrasseur NK. The influence of GDF11 on brain fate and function. Geroscience. 2019;41(1):1–11.

    PubMed  PubMed Central  Google Scholar 

  19. Qiu G, Wan R, Hu J, Mattson MP, Spangler E, Liu S, et al. Adiponectin protects rat hippocampal neurons against excitotoxicity. Age (Dordr). 2011;33(2):155–65.

    CAS  Google Scholar 

  20. Tokuno H, Moriya-Ito K, Tanaka I. Experimental techniques for neuroscience research using common marmosets. Exp Anim. 2012;61(4):389–97.

    CAS  PubMed  Google Scholar 

  21. Hashikawa T, Nakatomi R, Iriki A. Current models of the marmoset brain. Neurosci Res. 2015;93:116–27.

    PubMed  Google Scholar 

  22. Mertens J, Reid D, Lau S, Kim Y, Gage FH. Aging in a dish: iPSC-derived and directly induced neurons for studying brain aging and age-related neurodegenerative diseases. Annu Rev Genet. 2018;52:271–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Chang CY, Ting HC, Liu CA, Su HL, Chiou TW, Harn HJ, Lin SZ. Induced Pluripotent Stem Cells: A Powerful Neurodegenerative Disease Modeling Tool for Mechanism Study and Drug Discovery. Cell Transplant. 2018;27(11):1588–602.

  24. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, et al. Epigenetic memory in induced pluripotent stem cells. Nature. 2010;467(7313):285–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kim K, Zhao R, Doi A, Ng K, Unternaehrer J, Cahan P, et al. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nat Biotechnol. 2011;29(12):1117–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Tan KY, et al. Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat Biotechnol. 2010;28(8):848–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Bar-Nur O, Russ HA, Efrat S, Benvenisty N. Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet beta cells. Cell Stem Cell. 2011;9(1):17–23.

    CAS  PubMed  Google Scholar 

  28. Ghosh Z, Wilson KD, Wu Y, Hu S, Quertermous T, Wu JC. Persistent donor cell gene expression among human induced pluripotent stem cells contributes to differences with human embryonic stem cells. PLoS One. 2010;5(2):e8975.

    PubMed  PubMed Central  Google Scholar 

  29. Doss MX, Sachinidis A. Current Challenges of iPSC-Based Disease Modeling and Therapeutic Implications. Cells. 2019;8(5):403.

  30. Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer’s disease with iPSC-derived brain cells. Mol Psychiatry. 2020;25(1):148–67.

    PubMed  Google Scholar 

  31. Wang W, Jin K, Mao XO, Close N, Greenberg DA, Xiong ZG. Electrophysiological properties of mouse cortical neuron progenitors differentiated in vitro and in vivo. Int J Clin Exp Med. 2008;1(2):145–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Evans MS, Collings MA, Brewer GJ. Electrophysiology of embryonic, adult and aged rat hippocampal neurons in serum-free culture. J Neurosci Methods. 1998;79(1):37–46.

    CAS  PubMed  Google Scholar 

  33. Harris RA, Tardif SD, Vinar T, Wildman DE, Rutherford JN, Rogers J, et al. Evolutionary genetics and implications of small size and twinning in callitrichine primates. Proc Natl Acad Sci U S A. 2014;111(4):1467–72.

    CAS  PubMed  Google Scholar 

  34. Chen Y, Holstein DM, Aime S, Bollo M, Lechleiter JD. Calcineurin beta protects brain after injury by activating the unfolded protein response. Neurobiol Dis. 2016;94:139–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lin DT, Wu J, Holstein D, Upadhyay G, Rourk W, Muller E, et al. Ca2+ signaling, mitochondria and sensitivity to oxidative stress in aging astrocytes. Neurobiol Aging. 2007;28(1):99–111.

    PubMed  Google Scholar 

  36. Nelson GM, Guynn JM, Chorley BN. Procedure and Key Optimization Strategies for an Automated Capillary Electrophoretic-based Immunoassay Method. J Vis Exp. 2017;127:55911. https://doi.org/10.3791/55911.

    Article  CAS  Google Scholar 

  37. Harris VM. Protein detection by simple Western analysis. Methods Mol Biol. 2015;1312:465–8.

    PubMed  Google Scholar 

  38. LeBlanc A. Increased production of 4 kDa amyloid beta peptide in serum deprived human primary neuron cultures: possible involvement of apoptosis. J Neurosci. 1995;15(12):7837–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Schildge S, Bohrer C, Beck K, Schachtrup C. Isolation and culture of mouse cortical astrocytes. J Vis Exp. 2013;71:50079.

  40. Sun X, Hu X, Wang D, Yuan Y, Qin S, Tan Z, et al. Establishment and characterization of primary astrocyte culture from adult mouse brain. Brain Res Bull. 2017;132:10–9.

    CAS  PubMed  Google Scholar 

  41. Gradisnik L, Maver U, Bosnjak R, Velnar T. Optimised isolation and characterisation of adult human astrocytes from neurotrauma patients. J Neurosci Methods. 2020;341:108796.

    CAS  PubMed  Google Scholar 

  42. Homman-Ludiye J, Merson TD, Bourne JA. The early postnatal nonhuman primate neocortex contains self-renewing multipotent neural progenitor cells. PLoS One. 2012;7(3):e34383.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Mor E, Cabilly Y, Goldshmit Y, Zalts H, Modai S, Edry L, et al. Species-specific microRNA roles elucidated following astrocyte activation. Nucleic Acids Res. 2011;39(9):3710–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Hui CW, Zhang Y, Herrup K. Non-neuronal cells are required to mediate the effects of neuroinflammation: results from a neuron-enriched culture system. PLoS One. 2016;11(1):e0147134.

    PubMed  PubMed Central  Google Scholar 

  45. Ferrer I, García MA, González IL, Lucena DD, Villalonga AR, Tech MC, et al. Aging-related tau astrogliopathy (ARTAG): not only tau phosphorylation in astrocytes. Brain Pathol. 2018;28(6):965–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Correale J, Farez MF. The role of astrocytes in multiple sclerosis progression. Front Neurol. 2015;6:180.

    PubMed  PubMed Central  Google Scholar 

  47. Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang HY. Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res. 2003;971(2):197–209.

    CAS  PubMed  Google Scholar 

  48. Spanic E, et al. Role of microglial cells in Alzheimer’s disease tau propagation. Front Aging Neurosci. 2019;11:271.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Oberheim NA, Takano T, Han X, He W, Lin JHC, Wang F, et al. Uniquely hominid features of adult human astrocytes. J Neurosci. 2009;29(10):3276–87.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Boghdadi AG, Teo L, Bourne JA. The neuroprotective role of reactive astrocytes after central nervous system injury. J Neurotrauma. 2020;37(5):681–91.

    PubMed  Google Scholar 

  51. Fernandez E, Ross C, Liang H, Javors M, Tardif S, Salmon AB. Evaluation of the pharmacokinetics of metformin and acarbose in the common marmoset. Pathobiol Aging Age Relat Dis. 2019;9(1):1657756.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lee HJ, Gonzalez O, Dick EJ Jr, Donati A, Feliers D, Choudhury GG, et al. Marmoset as a model to study kidney changes associated with aging. J Gerontol A Biol Sci Med Sci. 2019;74(3):315–24.

    CAS  PubMed  Google Scholar 

  53. Reveles KR, Patel S, Forney L, Ross CN. Age-related changes in the marmoset gut microbiome. Am J Primatol. 2019;81(2):e22960.

    PubMed  PubMed Central  Google Scholar 

  54. Yun JW, Ahn JB, Kang BC. Modeling Parkinson’s disease in the common marmoset (Callithrix jacchus): overview of models, methods, and animal care. Lab Anim Res. 2015;31(4):155–65.

    PubMed  PubMed Central  Google Scholar 

  55. Sharma G, Huo A, Kimura T, Shiozawa S, Kobayashi R, Sahara N, et al. Tau isoform expression and phosphorylation in marmoset brains. J Biol Chem. 2019;294(30):11433–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Philippens IH, Ormel PR, Baarends G, Johansson M, Remarque EJ, Doverskog M. Acceleration of amyloidosis by inflammation in the amyloid-beta marmoset monkey model of Alzheimer’s disease. J Alzheimers Dis. 2017;55(1):101–13.

    CAS  PubMed  Google Scholar 

  57. Rodriguez-Callejas JD, Fuchs E, Perez-Cruz C. Evidence of tau hyperphosphorylation and dystrophic microglia in the common marmoset. Front Aging Neurosci. 2016;8:315.

    PubMed  PubMed Central  Google Scholar 

Download references

Funding

These studies were supported by Merit Review Award 2I0 1BX002211-05A1 from the US Department of Veterans Affairs Biomedical Laboratory Research and Development Service, NIH/NIA R01AG057964-01, the Robert L. Bailey and daughter Lisa K. Bailey Alzheimer’s Fund in memory of Jo Nell Bailey to VG, a William & Ella Owens Medical Research Foundation Grant, the San Antonio Medical Foundation, and the JMR Barker Foundation to VG. These studies were also supported by an award to VG through the NCATS/NIH Clinical and Translational Science Award grant UL1TR002645. SAH was supported by a Career Development (1 IK2 BX003798-01A1) award from the US Department of Veterans Affairs Biomedical Laboratory Research and Development Service. AOD was supported by NIA Training Grant T32AG021890. ABS was supported by NIH/NIA AG050797, the San Antonio Claude D. Pepper Older Americans Independence Center award (NIH/NIA P30 AG044271), and the San Antonio Nathan Shock Center of Excellence in the Biology of Aging (NIH/NIA P30 AG013319).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Veronica Galvan.

Ethics declarations

The UTHSCSA Institutional Animal Care and Use Committee (IACUC) regularly monitored marmoset housing and animal conditions to ensure all guidelines for the health and safety of the animals were met. This research was reviewed and approved by the UTHSCSA IACUC and experiments were conducted in compliance with the US Public Health Service’s Policy on Humane Care and Use of Laboratory Animals and the Guide for the Care and Use of Laboratory Animals and adhered to the American Society of Primatologists (ASP) principles for the ethical treatment of non-human primates.

Disclaimer

The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dorigatti, A.O., Hussong, S.A., Hernandez, S.F. et al. Primary neuron and astrocyte cultures from postnatal Callithrix jacchus: a non-human primate in vitro model for research in neuroscience, nervous system aging, and neurological diseases of aging. GeroScience 43, 115–124 (2021). https://doi.org/10.1007/s11357-020-00284-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11357-020-00284-z

Keywords

Navigation