Display options
Share it on

Hum Mol Genet. 2021 May 28;30(9):811-822. doi: 10.1093/hmg/ddab090.

Exome sequencing revealed PDE11A as a novel candidate gene for early-onset Alzheimer's disease.

Human molecular genetics

Wei Qin, Aihong Zhou, Xiumei Zuo, Longfei Jia, Fangyu Li, Qi Wang, Ying Li, Yiping Wei, Hongmei Jin, Carlos Cruchaga, Bruno A Benitez, Jianping Jia

Affiliations

  1. Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing 100053, China.
  2. Department of Psychiatry, Washington University, St. Louis, MO 63110, USA.
  3. NeuroGenomics and Informatics Center, Washington University, St. Louis, MO 63110, USA.
  4. Department of Genetics, Washington University, St. Louis, MO 63110, USA.
  5. Beijing Key Laboratory of Geriatric Cognitive Disorders, Capital Medical University, Beijing 10053, China.
  6. Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing 10053, China.
  7. Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 10053, China.

PMID: 33835157 PMCID: PMC8161517 DOI: 10.1093/hmg/ddab090

Abstract

To identify novel risk genes and better understand the molecular pathway underlying Alzheimer's disease (AD), whole-exome sequencing was performed in 215 early-onset AD (EOAD) patients and 255 unrelated healthy controls of Han Chinese ethnicity. Subsequent validation, computational annotation and in vitro functional studies were performed to evaluate the role of candidate variants in EOAD. We identified two rare missense variants in the phosphodiesterase 11A (PDE11A) gene in individuals with EOAD. Both variants are located in evolutionarily highly conserved amino acids, are predicted to alter the protein conformation and are classified as pathogenic. Furthermore, we found significantly decreased protein levels of PDE11A in brain samples of AD patients. Expression of PDE11A variants and knockdown experiments with specific short hairpin RNA (shRNA) for PDE11A both resulted in an increase of AD-associated Tau hyperphosphorylation at multiple epitopes in vitro. PDE11A variants or PDE11A shRNA also caused increased cyclic adenosine monophosphate (cAMP) levels, protein kinase A (PKA) activation and cAMP response element-binding protein phosphorylation. In addition, pretreatment with a PKA inhibitor (H89) suppressed PDE11A variant-induced Tau phosphorylation formation. This study offers insight into the involvement of Tau phosphorylation via the cAMP/PKA pathway in EOAD pathogenesis and provides a potential new target for intervention.

© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: [email protected].

References

  1. Acta Neuropathol. 2019 Aug;138(2):201-220 - PubMed
  2. J Neurosci Res. 2017 Jul;95(7):1503-1512 - PubMed
  3. Lancet Neurol. 2019 Jan;18(1):88-106 - PubMed
  4. Mol Neurobiol. 2018 Jan;55(1):696-706 - PubMed
  5. JAMA Neurol. 2019 Sep 1;76(9):1099-1108 - PubMed
  6. Biochim Biophys Acta. 2014 Dec;1842(12 Pt A):2517-27 - PubMed
  7. Adv Neurobiol. 2017;17:201-230 - PubMed
  8. Proc Natl Acad Sci U S A. 2006 Oct 10;103(41):15124-9 - PubMed
  9. Cell Signal. 2014 Feb;26(2):383-97 - PubMed
  10. Neurobiol Dis. 2017 Jul;103:163-173 - PubMed
  11. Neuropsychopharmacology. 2016 Nov;41(12):2920-2931 - PubMed
  12. Alzheimers Dement. 2011 May;7(3):263-9 - PubMed
  13. Brain Res. 1999 Nov 6;846(2):265-7 - PubMed
  14. Neurobiol Aging. 2017 Jan;49:215.e1-215.e8 - PubMed
  15. Curr Pharm Des. 2015;21(3):389-416 - PubMed
  16. Acta Neuropathol. 2020 Jan;139(1):45-61 - PubMed
  17. J Neuroimmune Pharmacol. 2018 Jun;13(2):254-264 - PubMed
  18. PLoS Med. 2017 Mar 28;14(3):e1002270 - PubMed
  19. Proc Natl Acad Sci U S A. 2010 May 4;107(18):8457-62 - PubMed
  20. Cell Signal. 2018 Jan;42:281-291 - PubMed
  21. Nat Neurosci. 2019 Dec;22(12):2087-2097 - PubMed
  22. Alzheimers Dement. 2007 Jul;3(3):186-91 - PubMed
  23. Mov Disord. 2020 Apr;35(4):606-615 - PubMed
  24. Brain Res. 1999 Feb 13;818(2):383-96 - PubMed
  25. Proc Natl Acad Sci U S A. 2000 Mar 28;97(7):3702-7 - PubMed
  26. Mol Psychiatry. 2017 Dec;22(12):1714-1724 - PubMed
  27. Nat Neurosci. 2020 Mar;23(3):311-322 - PubMed
  28. FEBS Lett. 2006 Nov 13;580(26):6269-74 - PubMed
  29. Genet Med. 2015 May;17(5):405-24 - PubMed
  30. Autophagy. 2020 Dec;16(12):2193-2205 - PubMed
  31. Alzheimers Dement. 2016 Jun;12(6):733-48 - PubMed
  32. Neurology. 1984 Jul;34(7):939-44 - PubMed
  33. J Clin Neurol. 2017 Apr;13(2):144-154 - PubMed
  34. Curr Biol. 2019 Jul 22;29(14):2307-2321.e5 - PubMed
  35. Front Neurol. 2017 Aug 15;8:399 - PubMed
  36. Proc Natl Acad Sci U S A. 2014 Apr 1;111(13):5036-41 - PubMed
  37. Cell Signal. 2020 Jun;70:109592 - PubMed
  38. Transl Psychiatry. 2013 Nov 19;3:e325 - PubMed
  39. Alzheimers Res Ther. 2012 Aug 20;4(4):34 - PubMed
  40. Biochem Soc Trans. 2019 Oct 31;47(5):1557-1565 - PubMed
  41. Alzheimers Dement. 2014 Nov;10(6):609-618.e11 - PubMed

Publication Types

Grant support