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

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, 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

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: journals.permissions@oup.com.

Figures

Figure 1
Figure 1
Workflow of the current study. Exome sequencing was performed in 215 Chinese individuals with EOAD and 255 controls. Subsequent direct sequencing was performed in an independent cohort to validate the selected rare variants. Then in vitro functional studies were performed to evaluate the role of candidate variants in EOAD and the underlying mechanisms.
Figure 2
Figure 2
Functional annotation of PDE11A variants. (A) Sanger sequencing confirmation of PDE11A variants. (B) Protein homologs in different species were aligned. The p.Arg202His and p.Leu756Gln are conserved in all PDE11A orthologs. The variants are highlighted in the red boxes. (C) Predicted domain maps of PDE11A protein with the identified missense variants marked: G: GAF domain (amino acids 217–380 and amino acids 402–568), P: HD/PDEase domain (amino acids 663–839), C: 3′5′-cyclic nucleotide phosphodiesterase, catalytic domain (amino acids 588–912). The p.Arg202His variant is located near the GAF domain, and p.Leu756Gln variant in the 3′5′-cyclic nucleotide phosphodiesterase, catalytic domain. (D) Predicted three-dimensional structure of wide type (WT) and mutant PDE11A protein. (E) Structure comparison between wild-type PDE11A and R202H variant. Critical hydrogen bonds with surrounding amino acids were predicted to be eliminated. Dashed lines indicate hydrogen bonds. (F) Structure comparison between wild-type PDE11A and L756Q variant. Q756 mutant was predicted to induce more hydrogen bonds and then affect helix structure.
Figure 3
Figure 3
The PDE11A expression in brain tissues of AD. (A and B) Based on single-nucleus RNA sequencing data, PDE11A gene was expressed almost in all kinds of cells. (C) Representative western blots illustrate the expression of PDE11A in postmortem brain tissues. (D) The histogram shows the quantification of PDE11A detected by immunoblot relative to control levels. The data are represented as the mean ± SEM, based on three unrelated measurements. **P < 0.01 by Student’s t-test.
Figure 4
Figure 4
The PDE11A variants led to significantly high Tau phosphorylation levels. The cells were co-infected with MAPT and PDE11A lentivirus (PDE11A WT, mutants, scramble or shRNA). A total of 72 h after infection, cell lysates were used to detect levels of p-Tau(T181), p-Tau(S404), p-Tau(S202), p-Tau(S416), p-Tau(S214), p-Tau(S396), p-Tau(AT8) and Tau. (A) Representative western blots illustrate the expressions of phosphorylated Tau. (B and C) The histograms show the quantification of phosphorylated Tau levels detected by immunoblot relative to control levels. The data are represented as the mean ± SEM, based on three unrelated measurements. *P < 0.05, **P < 0.01 by one-way ANOVA and Dunnett test or Student’s t-test.
Figure 5
Figure 5
The PDE11A variants affect cAMP/PKA signaling. ELISA analysis showed cAMP levels in cells after infection with WT PDE11A (with MAPT), decreased as expected; they increased following infection with PDE11A p.Arg202His, PDE11A p.Leu756Gln (A) or shRNA (with MAPT) (B). (C) Western blot of PKA signaling. The histograms show the quantification of p-PKA, PKA, p-CREB/CREB levels detected by immunoblot relative to control levels in cells infected with PDE11A mutants (D) or shRNA (with MAPT) (E). The data are represented as the mean ± SEM. *P < 0.05, **P < 0.01 by one-way ANOVA and Dunnett test or Student’s t-test.
Figure 6
Figure 6
The effects of PDE11A mutants on phosphorylation of Tau after treatment with PKA inhibitor. SH-SY5Y cells were treated with or without the PKA inhibitor H89 (10M). Then, cells were co-transfected with MAPT and PDE11A mutant or shRNA plasmids. (A) Cell lysates were collected to examine Tau phosphorylation levels using western blot. (B and C) The histograms show the quantification of phosphorylated Tau levels detected by immunoblot relative to control levels. The data are represented as the mean ± SEM. *P < 0.05, **P < 0.01 by one-way ANOVA and Dunnett test.

References

    1. GBD Dementia Collaborators (2019) Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol., 18, 88–106.
    1. Brookmeyer, R., Johnson, E., Ziegler-Graham, K. and Arrighi, H.M. (2007) Forecasting the global burden of Alzheimer’s disease. Alzheimers Dement., 3, 186–191.
    1. Barber, I.S., Braae, A., Clement, N., Patel, T., Guetta-Baranes, T., Brookes, K., Medway, C., Chappell, S., Guerreiro, R., Bras, J. et al. (2017) Mutation analysis of sporadic early-onset Alzheimer’s disease using the NeuroX array. Neurobiol. Aging, 49, 215.e211–215.e218.
    1. Cacace, R., Sleegers, K. and Van Broeckhoven, C. (2016) Molecular genetics of early-onset Alzheimer’s disease revisited. Alzheimers Dement., 12, 733–748.
    1. Sims, R., Hill, M. and Williams, J. (2020) The multiplex model of the genetics of Alzheimer’s disease. Nat. Neurosci., 23, 311–322.
    1. Lanoiselee, H.M., Nicolas, G., Wallon, D., Rovelet-Lecrux, A., Lacour, M., Rousseau, S., Richard, A.C., Pasquier, F., Rollin-Sillaire, A., Martinaud, O. et al. (2017) APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: a genetic screening study of familial and sporadic cases. PLoS Med., 14, e1002270.
    1. De Roeck, A., Van Broeckhoven, C. and Sleegers, K. (2019) The role of ABCA7 in Alzheimer’s disease: evidence from genomics, transcriptomics and methylomics. Acta Neuropathol., 138, 201–220.
    1. Ma, Y., Jun, G.R., Zhang, X., Chung, J., Naj, A.C., Chen, Y., Bellenguez, C., Hamilton-Nelson, K., Martin, E.R., Kunkle, B.W. et al. (2019) Analysis of whole-exome sequencing data for Alzheimer disease stratified by APOE genotype. JAMA Neurol., 76, 1099–1108.
    1. Jin, S.C., Pastor, P., Cooper, B., Cervantes, S., Benitez, B.A., Razquin, C., Goate, A., Ibero-American Alzheimer Disease Genetics Group, R and Cruchaga, C. (2012) Pooled-DNA sequencing identifies novel causative variants in PSEN1, GRN and MAPT in a clinical early-onset and familial Alzheimer’s disease Ibero-American cohort. Alzheimers Res. Ther., 4, 34.
    1. Richards, S., Aziz, N., Bale, S., Bick, D., Das, S., Gastier-Foster, J., Grody, W.W., Hegde, M., Lyon, E., Spector, E. et al. (2015) Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med., 17, 405–424.
    1. Kelly, M.P., Logue, S.F., Brennan, J., Day, J.P., Lakkaraju, S., Jiang, L., Zhong, X., Tam, M., Sukoff Rizzo, S.J., Platt, B.J. et al. (2010) Phosphodiesterase 11A in brain is enriched in ventral hippocampus and deletion causes psychiatric disease-related phenotypes. Proc. Natl. Acad. Sci. U. S. A., 107, 8457–8462.
    1. Grubman, A., Chew, G., Ouyang, J.F., Sun, G., Choo, X.Y., McLean, C., Simmons, R.K., Buckberry, S., Vargas-Landin, D.B., Poppe, D. et al. (2019) A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat. Neurosci., 22, 2087–2097.
    1. Fawcett, L., Baxendale, R., Stacey, P., McGrouther, C., Harrow, I., Soderling, S., Hetman, J., Beavo, J.A. and Phillips, S.C. (2000) Molecular cloning and characterization of a distinct human phosphodiesterase gene family: PDE11A. Proc. Natl. Acad. Sci. U. S. A., 97, 3702–3707.
    1. Kelly, M.P., Adamowicz, W., Bove, S., Hartman, A.J., Mariga, A., Pathak, G., Reinhart, V., Romegialli, A. and Kleiman, R.J. (2014) Select 3′,5′-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cell. Signal., 26, 383–397.
    1. Wong, M.L., Whelan, F., Deloukas, P., Whittaker, P., Delgado, M., Cantor, R.M., McCann, S.M. and Licinio, J. (2006) Phosphodiesterase genes are associated with susceptibility to major depression and antidepressant treatment response. Proc. Natl. Acad. Sci. U. S. A., 103, 15124–15129.
    1. Coon, H., Darlington, T., Pimentel, R., Smith, K.R., Huff, C.D., Hu, H., Jerominski, L., Hansen, J., Klein, M., Callor, W.B. et al. (2013) Genetic risk factors in two Utah pedigrees at high risk for suicide. Transl. Psychiatry, 3, e325.
    1. Pathak, G., Agostino, M.J., Bishara, K., Capell, W.R., Fisher, J.L., Hegde, S., Ibrahim, B.A., Pilarzyk, K., Sabin, C., Tuczkewycz, T. et al. (2017) PDE11A negatively regulates lithium responsivity. Mol. Psychiatry, 22, 1714–1724.
    1. Lee, P., Ryoo, H., Park, J., Jeong, Y. and Alzheimer’s Disease Neuroimaging Initiative (2017) Morphological and microstructural changes of the hippocampus in early MCI: a study utilizing the Alzheimer’s Disease Neuroimaging Initiative database. J. Clin. Neurol., 13, 144–154.
    1. Nie, X., Sun, Y., Wan, S., Zhao, H., Liu, R., Li, X., Wu, S., Nedelska, Z., Hort, J., Qing, Z. et al. (2017) Subregional structural alterations in hippocampus and nucleus accumbens correlate with the clinical impairment in patients with Alzheimer’s disease clinical spectrum: parallel combining volume and vertex-based approach. Front. Neurol., 8, 399.
    1. Hegde, S., Capell, W.R., Ibrahim, B.A., Klett, J., Patel, N.S., Sougiannis, A.T. and Kelly, M.P. (2016) Phosphodiesterase 11A (PDE11A), enriched in ventral hippocampus neurons, is required for consolidation of social but not nonsocial memories in mice. Neuropsychopharmacology, 41, 2920–2931.
    1. Pilarzyk, K., Klett, J., Pena, E.A., Porcher, L., Smith, A.J. and Kelly, M.P. (2019) Loss of function of phosphodiesterase 11A4 shows that recent and remote long-term memories can be uncoupled. Curr. Biol., 29, 2307–2321.e2305.
    1. Tibbo, A.J., Tejeda, G.S. and Baillie, G.S. (2019) Understanding PDE4’s function in Alzheimer's disease; a target for novel therapeutic approaches. Biochem. Soc. Trans., 47, 1557–1565.
    1. Cheng, J., Liao, Y., Dong, Y., Hu, H., Yang, N., Kong, X., Li, S., Li, X., Guo, J., Qin, L. et al. (2020) Microglial autophagy defect causes Parkinson disease-like symptoms by accelerating inflammasome activation in mice. Autophagy, 16, 1–13.
    1. Fazio, P., Fitzer-Attas, C.J., Mrzljak, L., Bronzova, J., Nag, S., Warner, J.H., Landwehrmeyer, B., Al-Tawil, N., Halldin, C., Forsberg, A. et al. (2020) PET molecular imaging of phosphodiesterase 10A: an early biomarker of Huntington's disease progression. Mov. Disord., 35, 606–615.
    1. Farmer, R., Burbano, S.D., Patel, N.S., Sarmiento, A., Smith, A.J. and Kelly, M.P. (2020) Phosphodiesterases PDE2A and PDE10A both change mRNA expression in the human brain with age, but only PDE2A changes in a region-specific manner with psychiatric disease. Cell. Signal., 70, 109592.
    1. Kelly, M.P. (2015) Does phosphodiesterase 11A (PDE11A) hold promise as a future therapeutic target? Curr. Pharm. Des., 21, 389–416.
    1. Kelly, M.P. (2017) A role for phosphodiesterase 11A (PDE11A) in the formation of social memories and the stabilization of mood. Adv. Neurobiol., 17, 201–230.
    1. Li, Z., Farias, F.H.G., Dube, U., Del-Aguila, J.L., Mihindukulasuriya, K.A., Fernandez, M.V., Ibanez, L., Budde, J.P., Wang, F., Lake, A.M. et al. (2020) The TMEM106B FTLD-protective variant, rs1990621, is also associated with increased neuronal proportion. Acta Neuropathol., 139, 45–61.
    1. Masutomi, H., Kawashima, S., Kondo, Y., Uchida, Y., Jang, B., Choi, E.K., Kim, Y.S., Shimokado, K. and Ishigami, A. (2017) Induction of peptidylarginine deiminase 2 and 3 by dibutyryl cAMP via cAMP-PKA signaling in human astrocytoma U-251MG cells. J. Neurosci. Res., 95, 1503–1512.
    1. Martinez, M., Fernandez, E., Frank, A., Guaza, C., de la Fuente, M. and Hernanz, A. (1999) Increased cerebrospinal fluid cAMP levels in Alzheimer’s disease. Brain Res., 846, 265–267.
    1. van der Harg, J.M., Eggels, L., Bangel, F.N., Ruigrok, S.R., Zwart, R., Hoozemans, J.J.M., la Fleur, S.E. and Scheper, W. (2017) Insulin deficiency results in reversible protein kinase A activation and tau phosphorylation. Neurobiol. Dis., 103, 163–173.
    1. Bonkale, W.L., Cowburn, R.F., Ohm, T.G., Bogdanovic, N. and Fastbom, J. (1999) A quantitative autoradiographic study of [3H]cAMP binding to cytosolic and particulate protein kinase A in post-mortem brain staged for Alzheimer’s disease neurofibrillary changes and amyloid deposits. Brain Res., 818, 383–396.
    1. Du, H., Guo, L., Wu, X., Sosunov, A.A., McKhann, G.M., Chen, J.X. and Yan, S.S. (2014) Cyclophilin D deficiency rescues Abeta-impaired PKA/CREB signaling and alleviates synaptic degeneration. Biochim. Biophys. Acta, 1842, 2517–2527.
    1. Kelly, M.P. (2018) Cyclic nucleotide signaling changes associated with normal aging and age-related diseases of the brain. Cell. Signal., 42, 281–291.
    1. Wang, H.H., Li, Y., Li, A., Yan, F., Li, Z.L., Liu, Z.Y., Zhang, L., Zhang, J., Dong, W.R. and Zhang, L. (2018) Forskolin induces hyperphosphorylation of tau accompanied by cell cycle reactivation in primary hippocampal neurons. Mol. Neurobiol., 55, 696–706.
    1. Carlyle, B.C., Nairn, A.C., Wang, M., Yang, Y., Jin, L.E., Simen, A.A., Ramos, B.P., Bordner, K.A., Craft, G.E., Davies, P. et al. (2014) cAMP-PKA phosphorylation of tau confers risk for degeneration in aging association cortex. Proc. Natl. Acad. Sci. U. S. A., 111, 5036–5041.
    1. Logue, M.W., Schu, M., Vardarajan, B.N., Farrell, J., Bennett, D.A., Buxbaum, J.D., Byrd, G.S., Ertekin-Taner, N., Evans, D., Foroud, T. et al. (2014) Two rare AKAP9 variants are associated with Alzheimer’s disease in African Americans. Alzheimers Dement., 10, 609–618.e611.
    1. Ikezu, T., Chen, C., DeLeo, A.M., Zeldich, E., Fallin, M.D., Kanaan, N.M., Lunetta, K.L., Abraham, C.R., Logue, M.W. and Farrer, L.A. (2018) Tau phosphorylation is impacted by rare AKAP9 mutations associated with Alzheimer disease in African Americans. J. Neuroimmune Pharmacol., 13, 254–264.
    1. Liu, F., Liang, Z., Shi, J., Yin, D., El-Akkad, E., Grundke-Iqbal, I., Iqbal, K. and Gong, C.X. (2006) PKA modulates GSK-3beta- and cdk5-catalyzed phosphorylation of tau in site- and kinase-specific manners. FEBS Lett., 580, 6269–6274.
    1. McKhann, G., Drachman, D., Folstein, M., Katzman, R., Price, D. and Stadlan, E.M. (1984) Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology, 34, 939–944.
    1. McKhann, G.M., Knopman, D.S., Chertkow, H., Hyman, B.T., Jack, C.R., Jr., Kawas, C.H., Klunk, W.E., Koroshetz, W.J., Manly, J.J., Mayeux, R. et al. (2011) The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement., 7, 263–269.

Source: PubMed

3
Abonneren